1
|
Salnikova DI, Shchekotikhin AE, Scherbakov AM. [A commentary: New carbonic anhydrase IX-targeted probes for imaging hypoxic tumors]. Bull Cancer 2025; 112:117-121. [PMID: 39690094 DOI: 10.1016/j.bulcan.2024.11.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 10/30/2024] [Accepted: 11/14/2024] [Indexed: 12/19/2024]
Abstract
In 2023, the journals "Bioconjugate Chemistry" and "Sensors and Actuators B: Chemical" published two papers describing new biosensors for imaging hypoxic regions in tumors. Cao et al. combined acetazolamide (AZA) to target carbonic anhydrase IX (CA IX) with two tyrosine-derived Mn(II)-ethylenediaminetetraacetic acid chelates (TyEDTA) on a rigid triazine (TA) scaffold. The aim of this synthesis was to create a Mn(II)-based magnetic resonance imaging probe named AZA-TA-Mn. In a murine hypoxia model of esophageal squamous cell carcinoma, AZA-TA-Mn, at a low dose of 0.05mmol/kg, produced selective contrast enhancement over non-specific Gd-diethylenetriaminepentaacetic acid (0.1mmol/kg). A competition study involving the co-injection of free AZA and a Mn(II)-based magnetic resonance imaging probe confirmed the in vivo tumor selectivity of AZA-TA-Mn. Immunofluorescence staining of tissue sections confirmed the positive correlation between tumor accumulation of AZA-TA-Mn and overexpression of AC IX. Using CA IX as a biomarker of hypoxia, Cao et al. have thus illustrated a practical strategy for the development of novel imaging probes for specific regions of hypoxia in a tumor of interest.
Collapse
Affiliation(s)
- Diana Igorevna Salnikova
- Département de biologie expérimentale des tumeurs, institut de carcinogenèse, centre national de recherche médicale en oncologie N.N. Blokhin, ministère de la Santé de Russie, 24 Kashirskoe shosse, Moscou 115522, Fédération de Russie.
| | - Andrey Egorovich Shchekotikhin
- Laboratoire de transformation chimique des antibiotiques, institut Gause des nouveaux antibiotiques, 11, rue Bol'chaya Pirogovskaya, Moscou 119021, Fédération de Russie
| | - Alexander Mikhailovich Scherbakov
- Département de biologie expérimentale des tumeurs, institut de carcinogenèse, centre national de recherche médicale en oncologie N.N. Blokhin, ministère de la Santé de Russie, 24 Kashirskoe shosse, Moscou 115522, Fédération de Russie.
| |
Collapse
|
2
|
Fujita N, Bondoc A, Simoes S, Ishida J, Taccone MS, Luck A, Srikanthan D, Siddaway R, Levine A, Sabha N, Krumholtz S, Kondo A, Arai H, Smith C, McDonald P, Hawkins C, Dedhar S, Rutka J. Combination treatment with histone deacetylase and carbonic anhydrase 9 inhibitors shows therapeutic potential in experimental diffuse intrinsic pontine glioma. Brain Tumor Pathol 2024; 41:117-131. [PMID: 39316272 DOI: 10.1007/s10014-024-00493-w] [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: 03/06/2024] [Accepted: 09/04/2024] [Indexed: 09/25/2024]
Abstract
Diffuse intrinsic pontine glioma (DIPG) remains a significant therapeutic challenge due to the lack of effective and safe treatment options. This study explores the potential of combining histone deacetylase (HDAC) and carbonic anhydrase 9 (CA9) inhibitors in treating DIPG. Analysis of RNA sequencing data and tumor tissue from patient samples for the expression of the carbonic anhydrase family and hypoxia signaling pathway activity revealed clinical relevance for targeting CA9 in DIPG. A synergy screen was conducted using CA9 inhibitor SLC-0111 and HDAC inhibitors panobinostat, vorinostat, entinostat, and pyroxamide. The combination of SLC-0111 and pyroxamide demonstrated the highest synergy and was selected for further analysis. Combining SLC-0111 and pyroxamide effectively inhibited DIPG cell proliferation, reduced cell migration and invasion potential, and enhanced histone acetylation, leading to decreased cell population in S Phase. Additionally, the combination therapy induced a greater reduction in intracellular pH than either agent alone. Data from this study suggest that the combination of SLC-0111 and pyroxamide holds promise for treating experimental DIPG, and further investigation of this combination therapy in preclinical models is warranted to evaluate its potential as a viable treatment for DIPG.
Collapse
Affiliation(s)
- Naohide Fujita
- Cell Biology Research Program, The Hospital for Sick Children, Toronto, ON, M5G 0A4, Canada
- Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, ON, M5G 0A4, Canada
- Department of Neurosurgery, Juntendo University School of Medicine, Bunkyo-Ku, Tokyo, 113-8421, Japan
| | - Andrew Bondoc
- Cell Biology Research Program, The Hospital for Sick Children, Toronto, ON, M5G 0A4, Canada
- Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, ON, M5G 0A4, Canada
| | - Sergio Simoes
- Cell Biology Research Program, The Hospital for Sick Children, Toronto, ON, M5G 0A4, Canada
- Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, ON, M5G 0A4, Canada
| | - Joji Ishida
- Cell Biology Research Program, The Hospital for Sick Children, Toronto, ON, M5G 0A4, Canada
- Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, ON, M5G 0A4, Canada
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, 700-8558, Japan
| | - Michael S Taccone
- Cell Biology Research Program, The Hospital for Sick Children, Toronto, ON, M5G 0A4, Canada
- Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, ON, M5G 0A4, Canada
- Department of Laboratory Medicine and Pathology, University of Toronto, Toronto, ON, M5S 1A8, Canada
- Division of Neurosurgery, Department of Surgery, The Ottawa Hospital, Ottawa, ON, K1Y4E9, Canada
| | - Amanda Luck
- Cell Biology Research Program, The Hospital for Sick Children, Toronto, ON, M5G 0A4, Canada
- Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, ON, M5G 0A4, Canada
| | - Dilakshan Srikanthan
- Cell Biology Research Program, The Hospital for Sick Children, Toronto, ON, M5G 0A4, Canada
- Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, ON, M5G 0A4, Canada
| | - Robert Siddaway
- Cell Biology Research Program, The Hospital for Sick Children, Toronto, ON, M5G 0A4, Canada
- Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, ON, M5G 0A4, Canada
- Division of Pathology, The Hospital for Sick Children, Toronto, ON, M5G 1X8, Canada
| | - Adrian Levine
- Cell Biology Research Program, The Hospital for Sick Children, Toronto, ON, M5G 0A4, Canada
- Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, ON, M5G 0A4, Canada
- Division of Pathology, The Hospital for Sick Children, Toronto, ON, M5G 1X8, Canada
| | - Nesrin Sabha
- Cell Biology Research Program, The Hospital for Sick Children, Toronto, ON, M5G 0A4, Canada
- Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, ON, M5G 0A4, Canada
| | - Stacey Krumholtz
- Cell Biology Research Program, The Hospital for Sick Children, Toronto, ON, M5G 0A4, Canada
- Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, ON, M5G 0A4, Canada
| | - Akihide Kondo
- Department of Neurosurgery, Juntendo University School of Medicine, Bunkyo-Ku, Tokyo, 113-8421, Japan
| | - Hajime Arai
- Department of Neurosurgery, Juntendo University School of Medicine, Bunkyo-Ku, Tokyo, 113-8421, Japan
| | - Christian Smith
- Cell Biology Research Program, The Hospital for Sick Children, Toronto, ON, M5G 0A4, Canada
- Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, ON, M5G 0A4, Canada
| | - Paul McDonald
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, BC, V5Z 1L3, Canada
| | - Cynthia Hawkins
- Cell Biology Research Program, The Hospital for Sick Children, Toronto, ON, M5G 0A4, Canada
- Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, ON, M5G 0A4, Canada
- Division of Pathology, The Hospital for Sick Children, Toronto, ON, M5G 1X8, Canada
- Department of Laboratory Medicine and Pathology, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Shoukat Dedhar
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, BC, V5Z 1L3, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada
| | - James Rutka
- Cell Biology Research Program, The Hospital for Sick Children, Toronto, ON, M5G 0A4, Canada.
- Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, ON, M5G 0A4, Canada.
- Division of Neurosurgery, The Hospital for Sick Children, 555 University Ave, Suite 1503, Toronto, ON, M5G 1X8, Canada.
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, ON, M5T 1P5, Canada.
| |
Collapse
|
3
|
Pravoverov K, Fatima I, Barman S, Jühling F, Primeaux M, Baumert TF, Singh AB, Dhawan P. IL-22 regulates MASTL expression in intestinal epithelial cells. Am J Physiol Gastrointest Liver Physiol 2024; 327:G123-G139. [PMID: 38771154 PMCID: PMC11687961 DOI: 10.1152/ajpgi.00260.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 05/09/2024] [Accepted: 05/09/2024] [Indexed: 05/22/2024]
Abstract
Microtubule-associated serine-threonine kinase-like (MASTL) has recently been identified as an oncogenic kinase given its overexpression in numerous cancers. Our group has shown that MASTL expression is upregulated in mouse models of sporadic colorectal cancer and colitis-associated cancer (CAC). CAC is one of the most severe complications of chronic inflammatory bowel disease (IBD), but a limited understanding of the mechanisms governing the switch from normal healing to neoplasia in IBD underscores the need for increased research in this area. However, MASTL levels in patients with IBD and its molecular regulation in IBD and CAC have not been studied. This study reveals that MASTL is upregulated by the cytokine interleukin (IL)-22, which promotes proliferation and has important functions in colitis recovery; however, IL-22 can also promote tumorigenesis when chronically elevated. Upon reviewing the publicly available data, we found significantly elevated MASTL and IL-22 levels in the biopsies from patients with late-stage ulcerative colitis compared with controls, and that MASTL upregulation was associated with high IL-22 expression. Our subsequent in vitro studies found that IL-22 increases MASTL expression in intestinal epithelial cell lines, which facilitates IL-22-mediated cell proliferation and downstream survival signaling. Inhibition of AKT activation abrogated IL-22-induced MASTL upregulation. We further found an increased association of carbonic anhydrase IX (CAIX) with MASTL in IL-22-treated cells, which stabilized MASTL expression. Inhibition of CAIX prevented IL-22-induced MASTL expression and cell survival. Overall, we show that IL-22/AKT signaling increases MASTL expression to promote cell survival and proliferation. Furthermore, CAIX associates with and stabilizes MASTL in response to IL-22 stimulation.NEW & NOTEWORTHY MASTL is upregulated in colorectal cancer; however, its role in colitis and colitis-associated cancer is poorly understood. This study is the first to draw a link between MASTL and IL-22, a proinflammatory/intestinal epithelial recovery-promoting cytokine that is also implicated in colon tumorigenesis. We propose that IL-22 increases MASTL protein stability by promoting its association with CAIX potentially via AKT signaling to promote cell survival and proliferation.
Collapse
Affiliation(s)
- Kristina Pravoverov
- Eppley Institute, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska, United States
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, United States
| | - Iram Fatima
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, United States
| | - Susmita Barman
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, United States
| | - Frank Jühling
- Inserm U1110, Université de Strasbourg, Institute for Translational Medicine and Liver Disease (ITM), Strasbourg, France
- Inserm U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, Université de Strasbourg, Strasbourg, France
| | - Mark Primeaux
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, United States
| | - Thomas F Baumert
- Inserm U1110, Université de Strasbourg, Institute for Translational Medicine and Liver Disease (ITM), Strasbourg, France
- Inserm U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, Université de Strasbourg, Strasbourg, France
- IHU Strasbourg and Gastroenterology-Hepatology Service, Strasbourg University Hospitals, Strasbourg, France
- Institut Universitaire de France (IUF), Paris, France
| | - Amar B Singh
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, United States
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska, United States
- Veteran Affairs Nebraska-Western Iowa Health Care System, Omaha, Nebraska, United States
| | - Punita Dhawan
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, United States
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska, United States
- Veteran Affairs Nebraska-Western Iowa Health Care System, Omaha, Nebraska, United States
| |
Collapse
|
4
|
Kalniņa Z, Liekniņa I, Skeltona V, Akopjana I, Kazāks A, Tārs K. Preclinical Evaluation of virus-like particle Vaccine Against Carbonic Anhydrase IX Efficacy in a Mouse Breast Cancer Model System. Mol Biotechnol 2024; 66:1206-1219. [PMID: 38217826 DOI: 10.1007/s12033-023-01021-5] [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: 07/12/2023] [Accepted: 12/05/2023] [Indexed: 01/15/2024]
Abstract
Carbonic anhydrase IX (CAIX) is a cancer-associated membrane protein frequently overexpressed in hypoxic solid tumours leading to enhanced tumour cell survival and invasion, and it has been proposed to be an attractive tumour-specific molecule for antibody-mediated targeting. This study aimed to generate a virus-like particle (VLP)-based CAIX vaccine candidate and evaluate its efficacy in a mouse model of breast cancer. The prototype murine vaccine was developed based on the ssRNA bacteriophage Qbeta VLPs with chemically coupled murine CAIX protein catalytic domains on their surfaces. The vaccine was shown to efficiently break the natural B cell tolerance against autologous murine CAIX and to induce high-titre Th1-oriented IgG responses in the BALB/c mice. This vaccine was tested in a therapeutic setting by using a triple-negative breast cancer mouse model system comprising 4T1, 4T1-Car9KI and 4T1-Car9KO cells, the latter representing positive and negative controls for murine CAIX production, respectively. The humoural immune responses induced in tumour-bearing animals were predominantly of Th1-type and higher anti-mCAIXc titres correlated with slower growth and lung metastasis development of 4T1 tumours constitutively expressing mCAIX in vivo in the syngeneic host.
Collapse
Affiliation(s)
- Zane Kalniņa
- Latvian Biomedical Research and Study Centre, Ratsupites 1, k-1, Riga, LV-1067, Latvia.
- Faculty of Biology, University of Latvia, Jelgavas 1, Riga, LV-1004, Latvia.
| | - Ilva Liekniņa
- Latvian Biomedical Research and Study Centre, Ratsupites 1, k-1, Riga, LV-1067, Latvia
| | - Vendija Skeltona
- Faculty of Biology, University of Latvia, Jelgavas 1, Riga, LV-1004, Latvia
| | - Ināra Akopjana
- Latvian Biomedical Research and Study Centre, Ratsupites 1, k-1, Riga, LV-1067, Latvia
| | - Andris Kazāks
- Latvian Biomedical Research and Study Centre, Ratsupites 1, k-1, Riga, LV-1067, Latvia
| | - Kaspars Tārs
- Latvian Biomedical Research and Study Centre, Ratsupites 1, k-1, Riga, LV-1067, Latvia
- Faculty of Biology, University of Latvia, Jelgavas 1, Riga, LV-1004, Latvia
| |
Collapse
|
5
|
Łukasiewicz-Śmietańska D, Godlewski D, Nowakowska E, Szpak A, Chabros E, Juszczyk G, Charzewska J, Rybaczyk-Pathak D. Association of the bitter taste genes TAS2R38 and CA6 and breast cancer risk; a case-control study of Polish women in Poland and Polish immigrants in USA. PLoS One 2024; 19:e0300061. [PMID: 38687739 PMCID: PMC11060581 DOI: 10.1371/journal.pone.0300061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 02/21/2024] [Indexed: 05/02/2024] Open
Abstract
It is known that the perception of bitterness is mediated by type 2 bitter taste receptors (TAS2Rs). However, recent reports have suggested that the carbonic anhydrase 6 (CA6) gene may also influence bitterness sensing. Genetic variants in these genes could influence dietary intake of brassica vegetables, whose increased consumption has been observed in the literature, though inconsistently, to decrease breast cancer (BC) risk. We hypothesized that the estimated odds ratios (ORs) for the association between BC and taster diplotype (PAV/PAV) and/or genotype A/A, will be in the direction of increased BC risk, potentially due to reduced consumption of brassica vegetables. Using a case-control study of BC in Polish women in Poland (210 cases and 262 controls) and Polish immigrant women to USA (78 cases and 170 controls) we evaluated the association of the taster diplotypes in TAS2R38 gene and genotypes in the CA6 gene and BC risk in these two populations individually and jointly. No significant increase in risk was observed for the TAS2R38 PAV/PAV diplotype (tasters) in each population individually or in the joint population. For the CA6 gene, in the joint population, we observed an increased BC risk for the combined G/A and G/G genotypes (non-tasters) vs A/A (tasters), OR = 1.41 (95% CI 1.04-1.90, p = 0.026) which after adjustment for False Discovery Rate (FDR), was not significant at p≤0.05 level. However, for the joint population and for the combined genotype of the two genes AVI/AVI+G* (non-tasters) vs. PAV/*+A/A (tasters), we observed a significant increase in BC risk, OR = 1.77 (95%CI 1.47-2.74, p = 0.01), for the non-tasters, which remained significant after FDR adjustment. In conclusion for the joint population and the joint effect for the two bitter sensing genes, we observed an increase in BC risk for the bitterness non-tasters, association which is in the opposite direction to our original hypothesis.
Collapse
Affiliation(s)
- Dorota Łukasiewicz-Śmietańska
- Department of Nutrition and Nutritional Value of Food, National Institute of Public Health NIH- National Research Institute, Warsaw, Poland
| | | | | | | | | | - Grzegorz Juszczyk
- National Institute of Public Health NIH- National Research Institute, Warsaw, Poland
| | - Jadwiga Charzewska
- Department of Nutrition and Nutritional Value of Food, National Institute of Public Health NIH- National Research Institute, Warsaw, Poland
| | - Dorothy Rybaczyk-Pathak
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI, United States of America
| |
Collapse
|
6
|
Buckenmeyer MJ, Brooks EA, Taylor MS, Yang L, Holewinski RJ, Meyer TJ, Galloux M, Garmendia-Cedillos M, Pohida TJ, Andresson T, Croix B, Wolf MT. Engineering Tumor Stroma Morphogenesis Using Dynamic Cell-Matrix Spheroid Assembly. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.19.585805. [PMID: 38903106 PMCID: PMC11188064 DOI: 10.1101/2024.03.19.585805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/22/2024]
Abstract
The tumor microenvironment consists of resident tumor cells organized within a compositionally diverse, three-dimensional (3D) extracellular matrix (ECM) network that cannot be replicated in vitro using bottom-up synthesis. We report a new self-assembly system to engineer ECM-rich 3D MatriSpheres wherein tumor cells actively organize and concentrate microgram quantities of decellularized ECM dispersions which modulate cell phenotype. 3D colorectal cancer (CRC) MatriSpheres were created using decellularized small intestine submucosa (SIS) as an orthotopic ECM source that had greater proteomic homology to CRC tumor ECM than traditional ECM formulations such as Matrigel. SIS ECM was rapidly concentrated from its environment and assembled into ECM-rich 3D stroma-like regions by mouse and human CRC cell lines within 4-5 days via a mechanism that was rheologically distinct from bulk hydrogel formation. Both ECM organization and transcriptional regulation by 3D ECM cues affected programs of malignancy, lipid metabolism, and immunoregulation that corresponded with an in vivo MC38 tumor cell subpopulation identified via single cell RNA sequencing. This 3D modeling approach stimulates tumor specific tissue morphogenesis that incorporates the complexities of both cancer cell and ECM compartments in a scalable, spontaneous assembly process that may further facilitate precision medicine.
Collapse
Affiliation(s)
- Michael J. Buckenmeyer
- Cancer Biomaterials Engineering Laboratory, Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, 21702, USA
| | - Elizabeth A. Brooks
- Cancer Biomaterials Engineering Laboratory, Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, 21702, USA
| | - Madison S. Taylor
- Cancer Biomaterials Engineering Laboratory, Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, 21702, USA
| | - Liping Yang
- Tumor Angiogenesis Unit, Mouse Cancer Genetics Program, National Cancer Institute, National Institutes of Health, Frederick, MD, 21702, USA
| | - Ronald J. Holewinski
- Protein Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD, 21701, USA
| | - Thomas J. Meyer
- CCR Collaborative Bioinformatics Resource, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Mélissa Galloux
- Independent Bioinformatician, Marseille, Provence-Alpes-Côte d’Azur, France
| | - Marcial Garmendia-Cedillos
- Instrumentation Development and Engineering Application Solutions, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Thomas J. Pohida
- Instrumentation Development and Engineering Application Solutions, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Thorkell Andresson
- Protein Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD, 21701, USA
| | - Brad Croix
- Tumor Angiogenesis Unit, Mouse Cancer Genetics Program, National Cancer Institute, National Institutes of Health, Frederick, MD, 21702, USA
| | - Matthew T. Wolf
- Cancer Biomaterials Engineering Laboratory, Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, 21702, USA
| |
Collapse
|
7
|
Wang Y, Buck A, Piel B, Zerefa L, Murugan N, Coherd CD, Miklosi AG, Johal H, Bastos RN, Huang K, Ficial M, Laimon YN, Signoretti S, Zhong Z, Hoang SM, Kastrunes GM, Grimaud M, Fayed A, Yuan HC, Nguyen QD, Thai T, Ivanova EV, Paweletz CP, Wu MR, Choueiri TK, Wee JO, Freeman GJ, Barbie DA, Marasco WA. Affinity fine-tuning anti-CAIX CAR-T cells mitigate on-target off-tumor side effects. Mol Cancer 2024; 23:56. [PMID: 38491381 PMCID: PMC10943873 DOI: 10.1186/s12943-024-01952-w] [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: 10/23/2023] [Accepted: 01/31/2024] [Indexed: 03/18/2024] Open
Abstract
One of the major hurdles that has hindered the success of chimeric antigen receptor (CAR) T cell therapies against solid tumors is on-target off-tumor (OTOT) toxicity due to sharing of the same epitopes on normal tissues. To elevate the safety profile of CAR-T cells, an affinity/avidity fine-tuned CAR was designed enabling CAR-T cell activation only in the presence of a highly expressed tumor associated antigen (TAA) but not when recognizing the same antigen at a physiological level on healthy cells. Using direct stochastic optical reconstruction microscopy (dSTORM) which provides single-molecule resolution, and flow cytometry, we identified high carbonic anhydrase IX (CAIX) density on clear cell renal cell carcinoma (ccRCC) patient samples and low-density expression on healthy bile duct tissues. A Tet-On doxycycline-inducible CAIX expressing cell line was established to mimic various CAIX densities, providing coverage from CAIX-high skrc-59 tumor cells to CAIX-low MMNK-1 cholangiocytes. Assessing the killing of CAR-T cells, we demonstrated that low-affinity/high-avidity fine-tuned G9 CAR-T has a wider therapeutic window compared to high-affinity/high-avidity G250 that was used in the first anti-CAIX CAR-T clinical trial but displayed serious OTOT effects. To assess the therapeutic effect of G9 on patient samples, we generated ccRCC patient derived organotypic tumor spheroid (PDOTS) ex vivo cultures and demonstrated that G9 CAR-T cells exhibited superior efficacy, migration and cytokine release in these miniature tumors. Moreover, in an RCC orthotopic mouse model, G9 CAR-T cells showed enhanced tumor control compared to G250. In summary, G9 has successfully mitigated OTOT side effects and in doing so has made CAIX a druggable immunotherapeutic target.
Collapse
Affiliation(s)
- Yufei Wang
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
- Harvard Medical School, Boston, MA, 02115, USA
| | - Alicia Buck
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | - Brandon Piel
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | - Luann Zerefa
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | - Nithyassree Murugan
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | - Christian D Coherd
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | | | | | | | - Kun Huang
- Molecular Imaging Core, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | - Miriam Ficial
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - Yasmin Nabil Laimon
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - Sabina Signoretti
- Harvard Medical School, Boston, MA, 02115, USA
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, 02115, USA
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | | | | | - Gabriella M Kastrunes
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | - Marion Grimaud
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | - Atef Fayed
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | - Hsien-Chi Yuan
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | - Quang-De Nguyen
- Lurie Family Imaging Center, Center for Biomedical Imaging in Oncology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | - Tran Thai
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | - Elena V Ivanova
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
- Belfer Center of Applied Cancer Science, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | - Cloud P Paweletz
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
- Belfer Center of Applied Cancer Science, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | - Ming-Ru Wu
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
- Harvard Medical School, Boston, MA, 02115, USA
| | - Toni K Choueiri
- Harvard Medical School, Boston, MA, 02115, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | - Jon O Wee
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | - Gordon J Freeman
- Harvard Medical School, Boston, MA, 02115, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | - David A Barbie
- Harvard Medical School, Boston, MA, 02115, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
- Belfer Center of Applied Cancer Science, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | - Wayne A Marasco
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA.
- Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
8
|
Wang SSY. Advancing biomarker development for diagnostics and therapeutics using solid tumour cancer stem cell models. TUMORI JOURNAL 2024; 110:10-24. [PMID: 36964664 DOI: 10.1177/03008916231158411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/26/2023]
Abstract
The cancer stem cell model hopes to explain solid tumour carcinogenesis, tumour progression and treatment failure in cancers. However, the cancer stem cell model has led to minimal clinical translation to cancer stem cell biomarkers and targeted therapies in solid tumours. Many reasons underlie the challenges, one being the imperfect understanding of the cancer stem cell model. This review hopes to spur further research into clinically translatable cancer stem cell biomarkers through first defining cancer stem cells and their associated models. With a better understanding of these models there would be a development of more accurate biomarkers. Making the clinical translation of biomarkers into diagnostic tools and therapeutic agents more feasible.
Collapse
|
9
|
Jiang N, Zheng Y, Ding J, Wang J, Zhu F, Wang M, Sobhani N, Neeli P, Wang G, Li H, Zheng J, Chai D. The co-delivery of adenovirus-based immune checkpoint vaccine elicits a potent anti-tumor effect in renal carcinoma. NPJ Vaccines 2023; 8:109. [PMID: 37542081 PMCID: PMC10403580 DOI: 10.1038/s41541-023-00706-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 07/11/2023] [Indexed: 08/06/2023] Open
Abstract
Immune-based checkpoint therapy has made significant progress in cancer treatment, but its therapeutic effect is limited. A replication-defective adenovirus (Ad) vaccine encoding tumor antigen carbonic anhydrase IX (CAIX) combined with Ad-encoding immune checkpoint PD-L1 was developed to treat renal carcinoma. Three tumor models, subcutaneous, lung metastasis and orthotopic tumor were established, and Ad vaccines were used to immunize them and evaluate the vaccine's therapeutic effect. Compared to the single Ad vaccine group, the subcutaneous tumor growth was significantly reduced in Ad-CAIX/Ad-PD-L1 combination group. Co-immunization of Ad-CAIX/Ad-PD-L1 enhanced the induction and maturation of CD11c+ or CD8+CD11c+ DCs in the spleen and tumor and promoted the strong tumor-specific CD8+ T cell immune responses. In vivo CD8 T cell deletion assay showed that the anti-tumor effect of the Ad-CAIX/Ad-PD-L1 vaccine was mainly dependent on functional CD8+ T cell immune responses. Furthermore, the Ad-CAIX/Ad-PD-L1 vaccine effectively inhibited tumor growth and lung metastasis in metastatic or orthotopic models. These results indicate that the combination strategy of the immune checkpoint vaccine shows promising potential as an approach for malignant tumor therapy.
Collapse
Affiliation(s)
- Nan Jiang
- Department of Urology, Suqian Hospital of Chinese Medicine Department of Pharmacy, Suqian, Jiangsu, China
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yanyan Zheng
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Jiage Ding
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Jiawei Wang
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Fei Zhu
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Meng Wang
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Navid Sobhani
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Praveen Neeli
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Gang Wang
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Hailong Li
- Department of Urology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.
| | - Junnian Zheng
- Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China.
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China.
| | - Dafei Chai
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China.
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China.
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
10
|
Minakata N, Sakashita S, Wakabayashi M, Nakamura Y, Sunakawa H, Yoda Y, Ishii G, Yano T. Immunohistochemistry and oxygen saturation endoscopic imaging reveal hypoxia in submucosal invasive esophageal squamous cell carcinoma. Cancer Med 2023; 12:15809-15819. [PMID: 37329213 PMCID: PMC10469640 DOI: 10.1002/cam4.6217] [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: 04/04/2023] [Revised: 05/24/2023] [Accepted: 05/29/2023] [Indexed: 06/18/2023] Open
Abstract
BACKGROUND Hypoxic microenvironment is prominent in advanced esophageal squamous cell carcinoma (ESCC). However, it is unclear whether ESCC becomes hypoxic when it remains in the mucosal layer or as it invades the submucosal layer. We aimed to investigate whether intramucosal (Tis-T1a) or submucosal invasive (T1b) ESCC becomes hypoxic using endoscopic submucosal dissection samples. METHODS We evaluated the expression of hypoxia markers including hypoxia inducible factor 1α (HIF-1α), carbonic anhydrase IX (CAIX), and glucose transporter 1 (GLUT1) by H-score and vessel density by microvessel count (MVC) and microvessel density (MVD) for CD31 and α-smooth muscle actin (α-SMA) with immunohistochemical staining (n = 109). Further, we quantified oxygen saturation (StO2 ) with oxygen saturation endoscopic imaging (OXEI) (n = 16) and compared them to non-neoplasia controls, Tis-T1a, and T1b. RESULTS In Tis-T1a, cccIX (13.0 vs. 0.290, p < 0.001) and GLUT1 (199 vs. 37.6, p < 0.001) were significantly increased. Similarly, median MVC (22.7/mm2 vs. 14.2/mm2 , p < 0.001) and MVD (0.991% vs. 0.478%, p < 0.001) were markedly augmented. Additionally, in T1b, the mean expression of HIF-1α (16.0 vs. 4.95, p < 0.001), CAIX (15.7 vs. 0.290, p < 0.001), and GLUT1 (177 vs. 37.6, p < 0.001) were significantly heightened, and median MVC (24.8/mm2 vs. 14.2/mm2 , p < 0.001) and MVD (1.51% vs. 0.478%, p < 0.001) were markedly higher. Furthermore, OXEI revealed that median StO2 was significantly lower in T1b than in non-neoplasia (54% vs. 61.5%, p = 0.00131) and tended to be lower in T1b than in Tis-T1a (54% vs. 62%, p = 0.0606). CONCLUSION These results suggest that ESCC becomes hypoxic even at an early stage, and is especially prominent in T1b.
Collapse
Affiliation(s)
- Nobuhisa Minakata
- Department of Gastroenterology and EndoscopyNational Cancer Center Hospital EastKashiwaJapan
- Course of Advanced Clinical Research of CancerJuntendo University Graduate School of MedicineBunkyo‐kuJapan
| | - Shingo Sakashita
- Division of PathologyExploratory Oncology Research and Clinical Trial Center, National Cancer CenterKashiwaJapan
| | - Masashi Wakabayashi
- Biostatistics Division, Center for Research Administration and SupportNational Cancer CenterKashiwaJapan
| | - Yuka Nakamura
- Department of Strategic ProgramsExploratory Oncology Research and Clinical Trial Center, National Cancer CenterKashiwaJapan
| | - Hironori Sunakawa
- Department of Gastroenterology and EndoscopyNational Cancer Center Hospital EastKashiwaJapan
- NEXT Medical Device Innovation CenterNational Cancer Center Hospital EastKashiwaJapan
| | - Yusuke Yoda
- Department of Gastroenterology and EndoscopyNational Cancer Center Hospital EastKashiwaJapan
- Department of Gastroenterology and EndoscopySaitama Cancer CenterSaitamaJapan
| | - Genichiro Ishii
- Course of Advanced Clinical Research of CancerJuntendo University Graduate School of MedicineBunkyo‐kuJapan
- Department of Pathology and Clinical LaboratoriesNational Cancer Center Hospital EastKashiwaJapan
| | - Tomonori Yano
- Department of Gastroenterology and EndoscopyNational Cancer Center Hospital EastKashiwaJapan
- Course of Advanced Clinical Research of CancerJuntendo University Graduate School of MedicineBunkyo‐kuJapan
- NEXT Medical Device Innovation CenterNational Cancer Center Hospital EastKashiwaJapan
| |
Collapse
|
11
|
Cui J, Xu H, Shi J, Fang K, Liu J, Liu F, Chen Y, Liang H, Zhang Y, Piao H. Carbonic anhydrase IX inhibitor S4 triggers release of DAMPs related to immunogenic cell death in glioma cells via endoplasmic reticulum stress pathway. Cell Commun Signal 2023; 21:167. [PMID: 37386564 PMCID: PMC10311836 DOI: 10.1186/s12964-023-01180-7] [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: 12/26/2022] [Accepted: 06/02/2023] [Indexed: 07/01/2023] Open
Abstract
BACKGROUND Immunogenic cell death (ICD), which releases danger-associated molecular patterns (DAMP) that induce potent anticancer immune response, has emerged as a key component of therapy-induced anti-tumor immunity. The aim of this work was to analyze whether the carbonic anhydrase IX inhibitor S4 can elicit ICD in glioma cells. METHODS The effects of S4 on glioma cell growth were evaluated using the CCK-8, clonogenic and sphere assays. Glioma cell apoptosis was determined by flow cytometry. Surface-exposed calreticulin (CRT) was inspected by confocal imaging. The supernatants of S4-treated cells were concentrated for the determination of HMGB1and HSP70/90 expression by immunoblotting. RNA-seq was performed to compare gene expression profiles between S4-treated and control cells. Pharmacological inhibition of apoptosis, autophagy, necroptosis and endoplasmic reticulum (ER) stress was achieved by inhibitors. In vivo effects of S4 were evaluated in glioma xenografts. Immunohistochemistry (IHC) was performed to stain Ki67 and CRT. RESULTS S4 significantly decreased the viability of glioma cells and induced apoptosis and autophagy. Moreover, S4 triggered CRT exposure and the release of HMGB1 and HSP70/90. Inhibition of either apoptosis or autophagy significantly reversed S4-induced release of DAMP molecules. RNA-seq analysis indicated that the ER stress pathway was deregulated upon exposure to S4. Both PERK-eIF2α and IRE1α- XBP1 axes were activated in S4-treated cells. Furthermore, pharmacological inhibition of PERK significantly suppressed S4-triggered ICD markers and autophagy. In glioma xenografts, S4 significantly reduced tumor growth. CONCLUSIONS Altogether, these findings suggest S4 as a novel ICD inducer in glioma and might have implications for S4-based immunotherapy. Video Abstract.
Collapse
Affiliation(s)
- Jing Cui
- Department of Neurosurgery, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, No.44 Xiaoheyan Road, Dadong District, Shenyang, 110042, China
| | - Huizhe Xu
- Central Laboratory, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, No.44 Xiaoheyan Road, Dadong District, Shenyang, 110042, China
| | - Ji Shi
- Department of Neurosurgery, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, No.44 Xiaoheyan Road, Dadong District, Shenyang, 110042, China
| | - Kun Fang
- Central Laboratory, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, No.44 Xiaoheyan Road, Dadong District, Shenyang, 110042, China
| | - Jia Liu
- Department of Neurosurgery, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, No.44 Xiaoheyan Road, Dadong District, Shenyang, 110042, China
| | - Feng Liu
- Department of Neurosurgery, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, No.44 Xiaoheyan Road, Dadong District, Shenyang, 110042, China
- Institute of Cancer Stem Cell, Dalian Medical University, No.9 Lvshun South Road, Lvshunkou District, Dalian, 116044, China
| | - Yi Chen
- Department of Neurosurgery, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, No.44 Xiaoheyan Road, Dadong District, Shenyang, 110042, China
| | - Haiyang Liang
- Department of Neurosurgery, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, No.44 Xiaoheyan Road, Dadong District, Shenyang, 110042, China
| | - Ye Zhang
- Department of Neurosurgery, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, No.44 Xiaoheyan Road, Dadong District, Shenyang, 110042, China.
| | - Haozhe Piao
- Department of Neurosurgery, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, No.44 Xiaoheyan Road, Dadong District, Shenyang, 110042, China.
- Central Laboratory, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, No.44 Xiaoheyan Road, Dadong District, Shenyang, 110042, China.
| |
Collapse
|
12
|
Lee S, Toft NJ, Axelsen TV, Espejo MS, Pedersen TM, Mele M, Pedersen HL, Balling E, Johansen T, Burton M, Thomassen M, Vahl P, Christiansen P, Boedtkjer E. Carbonic anhydrases reduce the acidity of the tumor microenvironment, promote immune infiltration, decelerate tumor growth, and improve survival in ErbB2/HER2-enriched breast cancer. Breast Cancer Res 2023; 25:46. [PMID: 37098526 PMCID: PMC10127511 DOI: 10.1186/s13058-023-01644-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 03/30/2023] [Indexed: 04/27/2023] Open
Abstract
BACKGROUND Carbonic anhydrases catalyze CO2/HCO3- buffer reactions with implications for effective H+ mobility, pH dynamics, and cellular acid-base sensing. Yet, the integrated consequences of carbonic anhydrases for cancer and stromal cell functions, their interactions, and patient prognosis are not yet clear. METHODS We combine (a) bioinformatic analyses of human proteomic data and bulk and single-cell transcriptomic data coupled to clinicopathologic and prognostic information; (b) ex vivo experimental studies of gene expression in breast tissue based on quantitative reverse transcription and polymerase chain reactions, intracellular and extracellular pH recordings based on fluorescence confocal microscopy, and immunohistochemical protein identification in human and murine breast cancer biopsies; and (c) in vivo tumor size measurements, pH-sensitive microelectrode recordings, and microdialysis-based metabolite analyses in mice with experimentally induced breast carcinomas. RESULTS Carbonic anhydrases-particularly the extracellular isoforms CA4, CA6, CA9, CA12, and CA14-undergo potent expression changes during human and murine breast carcinogenesis. In patients with basal-like/triple-negative breast cancer, elevated expression of the extracellular carbonic anhydrases negatively predicts survival, whereas, surprisingly, the extracellular carbonic anhydrases positively predict patient survival in HER2/ErbB2-enriched breast cancer. Carbonic anhydrase inhibition attenuates cellular net acid extrusion and extracellular H+ elimination from diffusion-restricted to peripheral and well-perfused regions of human and murine breast cancer tissue. Supplied in vivo, the carbonic anhydrase inhibitor acetazolamide acidifies the microenvironment of ErbB2-induced murine breast carcinomas, limits tumor immune infiltration (CD3+ T cells, CD19+ B cells, F4/80+ macrophages), lowers inflammatory cytokine (Il1a, Il1b, Il6) and transcription factor (Nfkb1) expression, and accelerates tumor growth. Supporting the immunomodulatory influences of carbonic anhydrases, patient survival benefits associated with high extracellular carbonic anhydrase expression in HER2-enriched breast carcinomas depend on the tumor inflammatory profile. Acetazolamide lowers lactate levels in breast tissue and blood without influencing breast tumor perfusion, suggesting that carbonic anhydrase inhibition lowers fermentative glycolysis. CONCLUSIONS We conclude that carbonic anhydrases (a) elevate pH in breast carcinomas by accelerating net H+ elimination from cancer cells and across the interstitial space and (b) raise immune infiltration and inflammation in ErbB2/HER2-driven breast carcinomas, restricting tumor growth and improving patient survival.
Collapse
Affiliation(s)
- Soojung Lee
- Department of Biomedicine, Aarhus University, Hoegh-Guldbergs Gade 10, Building 1115, DK-8000, Aarhus C, Denmark
| | - Nicolai J Toft
- Department of Biomedicine, Aarhus University, Hoegh-Guldbergs Gade 10, Building 1115, DK-8000, Aarhus C, Denmark
| | - Trine V Axelsen
- Department of Biomedicine, Aarhus University, Hoegh-Guldbergs Gade 10, Building 1115, DK-8000, Aarhus C, Denmark
| | - Maria Sofia Espejo
- Department of Biomedicine, Aarhus University, Hoegh-Guldbergs Gade 10, Building 1115, DK-8000, Aarhus C, Denmark
| | - Tina M Pedersen
- Department of Biomedicine, Aarhus University, Hoegh-Guldbergs Gade 10, Building 1115, DK-8000, Aarhus C, Denmark
| | - Marco Mele
- Department of Surgery, Randers Regional Hospital, Randers, Denmark
| | - Helene L Pedersen
- Department of Pathology, Randers Regional Hospital, Randers, Denmark
| | - Eva Balling
- Department of Surgery, Randers Regional Hospital, Randers, Denmark
| | - Tonje Johansen
- Department of Pathology, Randers Regional Hospital, Randers, Denmark
| | - Mark Burton
- Department of Clinical Genetics, University of Southern Denmark, Odense, Denmark
- Clinical Genome Center, University and Region of Southern Denmark, Odense, Denmark
- Department of Clinical Medicine, University of Southern Denmark, Odense, Denmark
| | - Mads Thomassen
- Department of Clinical Genetics, University of Southern Denmark, Odense, Denmark
- Clinical Genome Center, University and Region of Southern Denmark, Odense, Denmark
| | - Pernille Vahl
- Department of Pathology, Aarhus University Hospital, Aarhus, Denmark
| | - Peer Christiansen
- Department of Surgery, Randers Regional Hospital, Randers, Denmark
- Department of Plastic and Breast Surgery, Aarhus University Hospital, Aarhus, Denmark
| | - Ebbe Boedtkjer
- Department of Biomedicine, Aarhus University, Hoegh-Guldbergs Gade 10, Building 1115, DK-8000, Aarhus C, Denmark.
| |
Collapse
|
13
|
Numprasit W, Yangngam S, Prasopsiri J, Quinn JA, Edwards J, Thuwajit C. Carbonic anhydrase IX-related tumoral hypoxia predicts worse prognosis in breast cancer: A systematic review and meta-analysis. Front Med (Lausanne) 2023; 10:1087270. [PMID: 37007798 PMCID: PMC10063856 DOI: 10.3389/fmed.2023.1087270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 02/17/2023] [Indexed: 03/19/2023] Open
Abstract
BackgroundTumoral hypoxia is associated with aggressiveness in many cancers including breast cancer. However, measuring hypoxia is complicated. Carbonic anhydrase IX (CAIX) is a reliable endogenous marker of hypoxia under the control of the master regulator hypoxia-inducible factor-1α (HIF-1α). The expression of CAIX is associated with poor prognosis in many solid malignancies; however, its role in breast cancer remains controversial.MethodsThe present study performed a meta-analysis to evaluate the correlation between CAIX expression and disease-free survival (DFS) and overall survival (OS) in breast cancer.ResultsA total of 2,120 publications from EMBASE, PubMed, Cochrane, and Scopus were screened. Of these 2,120 publications, 272 full texts were reviewed, and 27 articles were included in the meta-analysis. High CAIX was significantly associated with poor DFS (HR = 1.70, 95% CI = 1.39–2.07, p < 0.00001) and OS (HR = 2.02, 95% CI 1.40–2.91, p = 0.0002) in patients with breast cancer. When stratified by subtype, the high CAIX group was clearly associated with shorter DFS (HR = 2.09, 95% CI =1.11–3.92, p = 0.02) and OS (HR = 2.50, 95% CI =1.53–4.07, p = 0.0002) in TNBC and shorter DFS in ER+ breast cancer (HR = 1.81 95% CI =1.38–2.36, p < 0.0001).ConclusionHigh CAIX expression is a negative prognostic marker of breast cancer regardless of the subtypes.
Collapse
Affiliation(s)
- Warapan Numprasit
- Division of Head Neck and Breast Surgery, Department of Surgery, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- School of Cancer Sciences, Wolfson Wohl Cancer Research Centre, University of Glasgow, Glasgow, United Kingdom
| | - Supaporn Yangngam
- Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Jaturawitt Prasopsiri
- Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Jean A. Quinn
- School of Cancer Sciences, Wolfson Wohl Cancer Research Centre, University of Glasgow, Glasgow, United Kingdom
| | - Joanne Edwards
- School of Cancer Sciences, Wolfson Wohl Cancer Research Centre, University of Glasgow, Glasgow, United Kingdom
| | - Chanitra Thuwajit
- Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- *Correspondence: Chanitra Thuwajit,
| |
Collapse
|
14
|
Metwally NH, El-Desoky EA. Novel Thiopyrano[2,3- d]thiazole-pyrazole Hybrids as Potential Nonsulfonamide Human Carbonic Anhydrase IX and XII Inhibitors: Design, Synthesis, and Biochemical Studies. ACS OMEGA 2023; 8:5571-5592. [PMID: 36816682 PMCID: PMC9933482 DOI: 10.1021/acsomega.2c06954] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 01/10/2023] [Indexed: 06/18/2023]
Abstract
In recent years, molecular hybridization strategies have developed into a potent strategy for drug discovery. A series of novel thiopyrano[2,3-d]thiazoles linked to the pyrazole moiety was designed and developed as anticancer agents by a molecular hybridization. Target compounds were synthesized and characterized by spectroscopic tools as well as X-ray crystallography analysis as in the case of thiopyrano[2,3-d]thiazole derivative 5a. The MTT assay was used to demonstrate the in vitro efficacy of compounds 5a-g and 7a-j on MCF-7 and HePG-2. The results showed that some cycloadducts such as bromophenyl-4-thioxo-2-thiazolidinone 3e, 4-methylphenyl derivative of thiopyrano[2,3-d]thiazole 5d, and 6-substituted-thiopyrano[2,3-d]thiazoles 7e-j displayed good to excellent IC50 in the range of 10.08 ± 1.5 to 25.95 ± 2.8 μg/mL against the MCF-7 cell line and from 7.83 ±2.1 to 13.37 ± 1.2 μg/mL against the HePG-2 cell line. To explore the enzymatic tests for isozymes hCAIX and hCAXII, the most promising eight compounds 3e, 5d, and 7e-j with IC50 ranging from 7.83 ± 2.1 to 25.95 ± 2.8 μM were chosen. Compound 7e exhibited an IC50 (0.067 ± 0.003 μM) similar to that of the standard drug AZA against CAIX (0.059 ± 0.003 μM)). For CAXII, the compound 7i had an IC50 equal to 0.123 ± 0.007 μM compared to that of AZA (0.083 ± 0.005 μM). In addition, using flow cytometry, cell cycle analysis and apoptosis studies in HePG-2 were performed for the two potent anticancer and selective carbonic anhydrase agents (7e and 7i). An enzymatic assay of these two compounds against caspase-9 was also examined. Interestingly, the molecular docking studies revealed that compounds 7e and 7i successfully embedded themselves in the active pockets of the CAIX and CAXII enzymes through different interactions. Overall, the novel thiopyrano[2,3-d]thiazole-pyrazole hybrids (7e and 7i) were suggested to be potent and selective inhibitors of CAIX and CAXII.
Collapse
|
15
|
Verhoeff SR, Oosting SF, Elias SG, van Es SC, Gerritse SL, Angus L, Heskamp S, Desar IM, Menke-van der Houven van Oordt CW, van der Veldt AA, Arens AI, Brouwers AH, Eisses B, Mulders PF, Hoekstra OS, Zwezerijnen GJ, van der Graaf WT, Aarntzen EH, Oyen WJ, van Herpen CM. [89Zr]Zr-DFO-girentuximab and [18F]FDG PET/CT to Predict Watchful Waiting Duration in Patients with Metastatic Clear-cell Renal Cell Carcinoma. Clin Cancer Res 2023; 29:592-601. [PMID: 36394882 PMCID: PMC9890134 DOI: 10.1158/1078-0432.ccr-22-0921] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Revised: 05/11/2022] [Accepted: 11/15/2022] [Indexed: 11/18/2022]
Abstract
PURPOSE Watchful waiting (WW) can be considered for patients with metastatic clear-cell renal cell carcinoma (mccRCC) with good or intermediate prognosis, especially those with <2 International Metastatic RCC Database Consortium criteria and ≤2 metastatic sites [referred to as watch and wait ("W&W") criteria]. The IMaging PAtients for Cancer drug SelecTion-Renal Cell Carcinoma study objective was to assess the predictive value of [18F]FDG PET/CT and [89Zr]Zr-DFO-girentuximab PET/CT for WW duration in patients with mccRCC. EXPERIMENTAL DESIGN Between February 2015 and March 2018, 48 patients were enrolled, including 40 evaluable patients with good (n = 14) and intermediate (n = 26) prognosis. Baseline contrast-enhanced CT, [18F]FDG and [89Zr]Zr-DFO-girentuximab PET/CT were performed. Primary endpoint was the time to disease progression warranting systemic treatment. Maximum standardized uptake values (SUVmax) were measured using lesions on CT images coregistered to PET/CT. High and low uptake groups were defined on the basis of median geometric mean SUVmax of RECIST-measurable lesions across patients. RESULTS The median WW time was 16.1 months [95% confidence interval (CI): 9.0-31.7]. The median WW period was shorter in patients with high [18F]FDG tumor uptake than those with low uptake (9.0 vs. 36.2 months; HR, 5.6; 95% CI: 2.4-14.7; P < 0.001). Patients with high [89Zr]Zr-DFO-girentuximab tumor uptake had a median WW period of 9.3 versus 21.3 months with low uptake (HR, 1.7; 95% CI: 0.9-3.3; P = 0.13). Patients with "W&W criteria" had a longer median WW period of 21.3 compared with patients without: 9.3 months (HR, 1.9; 95% CI: 0.9-3.9; Pone-sided = 0.034). Adding [18F]FDG uptake to the "W&W criteria" improved the prediction of WW duration (P < 0.001); whereas [89Zr]Zr-DFO-girentuximab did not (P = 0.53). CONCLUSIONS In patients with good- or intermediate-risk mccRCC, low [18F]FDG uptake is associated with prolonged WW. This study shows the predictive value of the "W&W criteria" for WW duration and shows the potential of [18F]FDG-PET/CT to further improve this.
Collapse
Affiliation(s)
- Sarah R. Verhoeff
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Sjoukje F. Oosting
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Sjoerd G. Elias
- Department of Epidemiology, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Suzanne C. van Es
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Sophie L. Gerritse
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, the Netherlands.,Department of Medical Oncology, Amsterdam UMC location VUMC, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Lindsay Angus
- Departments of Medical Oncology and Radiology & Nuclear Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Sandra Heskamp
- Department of Radiology, Nuclear Medicine and Anatomy, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Ingrid M.E. Desar
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, the Netherlands
| | | | - Astrid A.M. van der Veldt
- Departments of Medical Oncology and Radiology & Nuclear Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Anne I.J. Arens
- Department of Radiology, Nuclear Medicine and Anatomy, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Adrienne H. Brouwers
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Bertha Eisses
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Peter F.A. Mulders
- Department of Urology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Otto S. Hoekstra
- Department of Radiology and Nuclear Medicine, Amsterdam UMC location VUMC, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Gerben J.C. Zwezerijnen
- Department of Radiology and Nuclear Medicine, Amsterdam UMC location VUMC, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Winette T.A. van der Graaf
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, the Netherlands.,Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Erik H.J.G. Aarntzen
- Department of Radiology, Nuclear Medicine and Anatomy, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Wim J.G. Oyen
- Department of Radiology, Nuclear Medicine and Anatomy, Radboud University Medical Center, Nijmegen, the Netherlands.,Department of Radiology and Nuclear Medicine, Rijnstate, Arnhem, the Netherlands.,Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Carla M.L. van Herpen
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, the Netherlands.,Corresponding Author: Carla M.L. van Herpen, Radboud University Nijmegen Medical Center, Nijmegen 6500 HB, the Netherlands. Phone: 312-4361-4038; Fax: 312-4361-5025; E-mail:
| |
Collapse
|
16
|
Liao C, Liu X, Zhang C, Zhang Q. Tumor hypoxia: From basic knowledge to therapeutic implications. Semin Cancer Biol 2023; 88:172-186. [PMID: 36603793 PMCID: PMC9929926 DOI: 10.1016/j.semcancer.2022.12.011] [Citation(s) in RCA: 51] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 12/07/2022] [Accepted: 12/31/2022] [Indexed: 01/04/2023]
Abstract
Diminished oxygen availability, termed hypoxia, within solid tumors is one of the most common characteristics of cancer. Hypoxia shapes the landscape of the tumor microenvironment (TME) into a pro-tumorigenic and pro-metastatic niche through arrays of pathological alterations such as abnormal vasculature, altered metabolism, immune-suppressive phenotype, etc. In addition, emerging evidence suggests that limited efficacy or the development of resistance towards antitumor therapy may be largely due to the hypoxic TME. This review will focus on summarizing the knowledge about the molecular machinery that mediates the hypoxic cellular responses and adaptations, as well as highlighting the effects and consequences of hypoxia, especially for angiogenesis regulation, cellular metabolism alteration, and immunosuppressive response within the TME. We also outline the current advances in novel therapeutic implications through targeting hypoxia in TME. A deep understanding of the basics and the role of hypoxia in the tumor will help develop better therapeutic avenues in cancer treatment.
Collapse
Affiliation(s)
- Chengheng Liao
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Xijuan Liu
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, School of Medicine, Chapel Hill, NC 27599, USA
| | - Cheng Zhang
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Qing Zhang
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
17
|
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
|
18
|
Zhang Z, Wu B, Shao Y, Chen Y, Wang D. A systematic review verified by bioinformatic analysis based on TCGA reveals week prognosis power of CAIX in renal cancer. PLoS One 2022; 17:e0278556. [PMID: 36542612 PMCID: PMC9770376 DOI: 10.1371/journal.pone.0278556] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 11/21/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Carbonic anhydrase IX (CAIX) protein has been correlated with progression and survival in patients with some tumors such as head and neck carcinoma. But renal cell carcinoma is an exception. The prognostic value of CAIX in RCC used to be associated with patients' survival according to published works. This study aimed to rectify the former conclusion. METHODS This study was registered in PROSPERO (CRD42020160181). A literature search of the PubMed, Embase, Cochrane library and Web of Science databases was performed to retrieve original studies until April of 2022. Twenty-seven studies, including a total of 5462 patients with renal cell carcinoma, were reviewed. Standard meta-analysis methods were used to evaluate the prognostic impact of CAIX expression on patient prognosis. The hazard ratio and its 95% confidence interval were recorded for the relationship between CAIX expression and survival, and the data were analyzed using Stata 11.0. Then we verify the meta-analysis resort to bioinformatics (TCGA). RESULTS Our initial search resulted in 908 articles in total. From PubMed, Embase, Web of Science electronic and Cochrane library databases, 493, 318 and 97 potentially relevant articles were discovered, respectively. We took the analysis between CA9 and disease-specific survival (HR = 1.18, 95% CI: 0.82-1.70, I2 = 79.3%, P<0.05), a subgroup then was performed to enhance the result (HR = 1.63, 95%CI: 1.30-2.03, I2 = 26.3%, P = 0.228); overall survival was also parallel with the former (HR = 1.13, 95%CI: 0.82-1.56, I2 = 79.8%, P<0.05), then a subgroup also be performed (HR = 0.90, 95%CI:0.75-1.07, I2 = 23.1%, P = 0.246) to verify the result; the analysis between CAIX and progression-free survival got the similar result (HR = 1.73, 95%CI:0.97-3.09, I2 = 82.4%, P<0.05), we also verify the result by subgroup analysis (HR = 1.04, 95%CI:0.79-1.36, I2 = 0.0%, P = 0.465); at last the relationship between CAIX and recurrence-free survival got the same result, too (HR = 0.99, 95%CI: 0.95-1.02, I2 = 57.8%, P = 0.050), the subgroup's result was also parallel with the former (HR = 1.01, 95%CI: 0.91-1.03, I2 = 0.00%, P = 0.704). To validate our meta-analysis, we took a bioinformatic analysis based on TCGA database, survival curve between low and high CAIX expression in four endpoints (DSS, OS, PFI, DFI) have corresponding P value (DSS:P = 0.23, OS:P = 0.77, PFI:P = 0.25, DFI:P = 0.78). CONCLUSIONS CAIX expression in patients with RCC is an exception to predict tumor survival. Both low CAIX expression and high expression are not associated with survivals in RCC patients.
Collapse
Affiliation(s)
- Zikuan Zhang
- Basic Medicine of Shanxi Medical University, Taiyuan, China
| | - Bo Wu
- Basic Medicine of Shanxi Medical University, Taiyuan, China
| | - Yuan Shao
- Basic Medicine of Shanxi Medical University, Taiyuan, China
| | - Yongquan Chen
- Basic Medicine of Shanxi Medical University, Taiyuan, China
| | - Dongwen Wang
- Basic Medicine of Shanxi Medical University, Taiyuan, China
| |
Collapse
|
19
|
Amin MU, Ali S, Ali MY, Fuhrmann DC, Tariq I, Seitz BS, Preis E, Brüßler J, Brüne B, Bakowsky U. Co-delivery of carbonic anhydrase IX inhibitor and doxorubicin as a promising approach to address hypoxia-induced chemoresistance. Drug Deliv 2022; 29:2072-2085. [PMID: 35848469 PMCID: PMC9297722 DOI: 10.1080/10717544.2022.2092234] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Hypoxia, an oxygen-deprived condition of the tumor, is one of the major reasons for resistance to chemotherapy. Carbonic anhydrases are generally involved in pH homeostasis in normal conditions, but in solid tumors having a strong relation with hypoxia, the carbonic anhydrase IX (CA-IX) enzyme is overexpressed and results in an extracellular acidic environment. For most weakly basic anticancer drugs, including doxorubicin (Dox), the ionization in an acidic environment limits their cellular uptake, and consequently, the tumor exposure to the drug at sub-therapeutic concentration comes out as chemoresistance. Herein, a combined drug delivery system of liposomes and mesoporous silica nanoparticles (MSNPs) was developed for the co-delivery of the CA-IX enzyme inhibitor and Dox in hypoxic condition. The unique structure of MSNPs with higher surface area was utilized for higher drug loading and sustained release of Dox. Additionally, the biocompatible nature of liposomal coating as a second loading site for the CA-IX enzyme inhibitor has provided gatekeeping effects at pore opening to avoid premature drug release. Lipid coated MSNPs as a co-delivery system for Dox and the CA-IX inhibitor have synergistic cytotoxic effects against MDA-MB 231 breast cancer cells in hypoxic conditions. These findings assure the potential of this drug delivery system to overcome hypoxia-related chemoresistance.
Collapse
Affiliation(s)
- Muhammad Umair Amin
- Department of Pharmaceutics and Biopharmaceutics, University of Marburg, Marburg, Germany
| | - Sajid Ali
- Department of Pharmaceutics and Biopharmaceutics, University of Marburg, Marburg, Germany.,Department of Chemistry, Angström Laboratory, Uppsala University, Uppsala, Sweden
| | - Muhammad Yasir Ali
- Department of Pharmaceutics and Biopharmaceutics, University of Marburg, Marburg, Germany.,Faculty of Pharmaceutical Sciences, GC University Faisalabad, Faisalabad, Pakistan
| | - Dominik C Fuhrmann
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, Frankfurt, Germany
| | - Imran Tariq
- Department of Pharmaceutics and Biopharmaceutics, University of Marburg, Marburg, Germany.,Punjab University College of Pharmacy, University of Punjab, Lahore, Pakistan
| | - Benjamin S Seitz
- Department of Pharmaceutics and Biopharmaceutics, University of Marburg, Marburg, Germany
| | - Eduard Preis
- Department of Pharmaceutics and Biopharmaceutics, University of Marburg, Marburg, Germany
| | - Jana Brüßler
- Department of Pharmaceutics and Biopharmaceutics, University of Marburg, Marburg, Germany
| | - Bernhard Brüne
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, Frankfurt, Germany
| | - Udo Bakowsky
- Department of Pharmaceutics and Biopharmaceutics, University of Marburg, Marburg, Germany
| |
Collapse
|
20
|
Yakushov S, Menyailo M, Denisov E, Karlina I, Zainullina V, Kirgizov K, Romantsova O, Timashev P, Ulasov I. Identification of Factors Driving Doxorubicin-Resistant Ewing Tumor Cells to Survival. Cancers (Basel) 2022; 14:cancers14225498. [PMID: 36428591 PMCID: PMC9688843 DOI: 10.3390/cancers14225498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 10/26/2022] [Accepted: 11/03/2022] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Ewing sarcoma (ES) cells exhibit extreme plasticity that contributes to the cell's survival and recurrence. Although multiple studies reveal various signaling pathways mediated by the EWSR1/FLI1 fusion, the specific transcriptional control of tumor cell resistance to doxorubicin is unknown. Understanding the molecular hubs that contribute to this behavior provides a new perspective on valuable therapeutic options against tumor cells. METHODS Single-cell RNA sequencing and LC-MS/MS-based quantitative proteomics were used. RESULTS A goal of this study was to identify protein hubs that would help elucidate tumor resistance which prompted ES to relapse or metastasize. Several differentially expressed genes and proteins, including adhesion, cytoskeletal, and signaling molecules, were observed between embryonic fibroblasts and control and doxorubicin-treated tumor cell lines. While several cancer-associated genes/proteins exhibited similar expression across fibroblasts and non-treated cells, upregulation of some proteins belonging to metabolic, stress response, and growth pathway activation was uniquely observed in doxorubicin-treated sarcoma cells, respectively. The novel information on differentially expressed genes/proteins provides insights into the biology of ES cells, which could help elucidate mechanisms of their recurrence. CONCLUSIONS Collectively, our results identify a novel role of cellular proteins in contributing to tumor cell resistance and escape from doxorubicin therapy and contributing to ES progression.
Collapse
Affiliation(s)
- Semyon Yakushov
- Group of Experimental Biotherapy and Diagnostics, Institute for Regenerative Medicine, World-Class Research Centre “Digital Biodesign and Personalized Healthcare”, I.M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia
| | - Maxim Menyailo
- Laboratory of Cancer Progression Biology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 634009 Tomsk, Russia
| | - Evgeny Denisov
- Laboratory of Cancer Progression Biology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 634009 Tomsk, Russia
| | - Irina Karlina
- Group of Experimental Biotherapy and Diagnostics, Institute for Regenerative Medicine, World-Class Research Centre “Digital Biodesign and Personalized Healthcare”, I.M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia
| | - Viktoria Zainullina
- Laboratory of Cancer Progression Biology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 634009 Tomsk, Russia
| | - Kirill Kirgizov
- Research Institute of Pediatric Oncology and Hematology at N.N. Blokhin National Medical Research Center of Oncology, Ministry of Health of Russia, 115478 Moscow, Russia
| | - Olga Romantsova
- Research Institute of Pediatric Oncology and Hematology at N.N. Blokhin National Medical Research Center of Oncology, Ministry of Health of Russia, 115478 Moscow, Russia
| | - Peter Timashev
- World-Class Research Centre “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University, 119991 Moscow, Russia
| | - Ilya Ulasov
- Group of Experimental Biotherapy and Diagnostics, Institute for Regenerative Medicine, World-Class Research Centre “Digital Biodesign and Personalized Healthcare”, I.M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia
- Correspondence: ; Tel.: +7-901-797-5406
| |
Collapse
|
21
|
Matulienė J, Žvinys G, Petrauskas V, Kvietkauskaitė A, Zakšauskas A, Shubin K, Zubrienė A, Baranauskienė L, Kačenauskaitė L, Kopanchuk S, Veiksina S, Paketurytė-Latvė V, Smirnovienė J, Juozapaitienė V, Mickevičiūtė A, Michailovienė V, Jachno J, Stravinskienė D, Sližienė A, Petrošiūtė A, Becker HM, Kazokaitė-Adomaitienė J, Yaromina A, Čapkauskaitė E, Rinken A, Dudutienė V, Dubois LJ, Matulis D. Picomolar fluorescent probes for compound affinity determination to carbonic anhydrase IX expressed in live cancer cells. Sci Rep 2022; 12:17644. [PMID: 36271018 PMCID: PMC9586938 DOI: 10.1038/s41598-022-22436-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 10/14/2022] [Indexed: 01/18/2023] Open
Abstract
Numerous human cancers, especially hypoxic solid tumors, express carbonic anhydrase IX (CAIX), a transmembrane protein with its catalytic domain located in the extracellular space. CAIX acidifies the tumor microenvironment, promotes metastases and invasiveness, and is therefore considered a promising anticancer target. We have designed a series of high affinity and high selectivity fluorescein-labeled compounds targeting CAIX to visualize and quantify CAIX expression in cancer cells. The competitive binding model enabled the determination of common CA inhibitors' dissociation constants for CAIX expressed in exponentially growing cancer cells. All tested sulfonamide compounds bound the proliferating cells with similar affinity as to recombinantly purified CAIX. The probes are applicable for the design of selective drug-like compounds for CAIX and the competition strategy could be applied to other drug targets.
Collapse
Affiliation(s)
- Jurgita Matulienė
- grid.6441.70000 0001 2243 2806Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences Center, Vilnius University, Saulėtekio 7, LT-10257 Vilnius, Lithuania
| | - Gediminas Žvinys
- grid.6441.70000 0001 2243 2806Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences Center, Vilnius University, Saulėtekio 7, LT-10257 Vilnius, Lithuania
| | - Vytautas Petrauskas
- grid.6441.70000 0001 2243 2806Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences Center, Vilnius University, Saulėtekio 7, LT-10257 Vilnius, Lithuania
| | - Agnė Kvietkauskaitė
- grid.6441.70000 0001 2243 2806Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences Center, Vilnius University, Saulėtekio 7, LT-10257 Vilnius, Lithuania
| | - Audrius Zakšauskas
- grid.6441.70000 0001 2243 2806Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences Center, Vilnius University, Saulėtekio 7, LT-10257 Vilnius, Lithuania
| | - Kirill Shubin
- grid.419212.d0000 0004 0395 6526Latvian Institute of Organic Synthesis, Aizkraukles 21, Riga, LV-1006 Latvia
| | - Asta Zubrienė
- grid.6441.70000 0001 2243 2806Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences Center, Vilnius University, Saulėtekio 7, LT-10257 Vilnius, Lithuania
| | - Lina Baranauskienė
- grid.6441.70000 0001 2243 2806Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences Center, Vilnius University, Saulėtekio 7, LT-10257 Vilnius, Lithuania
| | - Lina Kačenauskaitė
- grid.6441.70000 0001 2243 2806Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences Center, Vilnius University, Saulėtekio 7, LT-10257 Vilnius, Lithuania
| | - Sergei Kopanchuk
- grid.10939.320000 0001 0943 7661Institute of Chemistry, University of Tartu, Ravila 14a, 50411 Tartu, Estonia
| | - Santa Veiksina
- grid.10939.320000 0001 0943 7661Institute of Chemistry, University of Tartu, Ravila 14a, 50411 Tartu, Estonia
| | - Vaida Paketurytė-Latvė
- grid.6441.70000 0001 2243 2806Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences Center, Vilnius University, Saulėtekio 7, LT-10257 Vilnius, Lithuania
| | - Joana Smirnovienė
- grid.6441.70000 0001 2243 2806Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences Center, Vilnius University, Saulėtekio 7, LT-10257 Vilnius, Lithuania
| | - Vaida Juozapaitienė
- grid.6441.70000 0001 2243 2806Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences Center, Vilnius University, Saulėtekio 7, LT-10257 Vilnius, Lithuania
| | - Aurelija Mickevičiūtė
- grid.6441.70000 0001 2243 2806Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences Center, Vilnius University, Saulėtekio 7, LT-10257 Vilnius, Lithuania
| | - Vilma Michailovienė
- grid.6441.70000 0001 2243 2806Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences Center, Vilnius University, Saulėtekio 7, LT-10257 Vilnius, Lithuania
| | - Jelena Jachno
- grid.6441.70000 0001 2243 2806Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences Center, Vilnius University, Saulėtekio 7, LT-10257 Vilnius, Lithuania
| | - Dovilė Stravinskienė
- grid.6441.70000 0001 2243 2806Department of Immunology and Cell Biology, Institute of Biotechnology, Life Sciences Center, Vilnius University, Saulėtekio 7, LT-10257 Vilnius, Lithuania
| | - Aistė Sližienė
- grid.6441.70000 0001 2243 2806Department of Immunology and Cell Biology, Institute of Biotechnology, Life Sciences Center, Vilnius University, Saulėtekio 7, LT-10257 Vilnius, Lithuania
| | - Agnė Petrošiūtė
- grid.6441.70000 0001 2243 2806Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences Center, Vilnius University, Saulėtekio 7, LT-10257 Vilnius, Lithuania
| | - Holger M. Becker
- Zoology and Animal Physiology, Institute of Zoology, TU Dresden, 01217 Dresden, Germany
| | - Justina Kazokaitė-Adomaitienė
- grid.6441.70000 0001 2243 2806Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences Center, Vilnius University, Saulėtekio 7, LT-10257 Vilnius, Lithuania ,grid.430814.a0000 0001 0674 1393Present Address: Division of Biochemistry, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Ala Yaromina
- grid.5012.60000 0001 0481 6099The M-Lab, Department of Precision Medicine, GROW – School for Oncology and Reproduction, Maastricht University, Universiteitssingel 50/23, 6200 MD Maastricht, The Netherlands
| | - Edita Čapkauskaitė
- grid.6441.70000 0001 2243 2806Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences Center, Vilnius University, Saulėtekio 7, LT-10257 Vilnius, Lithuania
| | - Ago Rinken
- grid.10939.320000 0001 0943 7661Institute of Chemistry, University of Tartu, Ravila 14a, 50411 Tartu, Estonia
| | - Virginija Dudutienė
- grid.6441.70000 0001 2243 2806Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences Center, Vilnius University, Saulėtekio 7, LT-10257 Vilnius, Lithuania
| | - Ludwig J Dubois
- grid.5012.60000 0001 0481 6099The M-Lab, Department of Precision Medicine, GROW – School for Oncology and Reproduction, Maastricht University, Universiteitssingel 50/23, 6200 MD Maastricht, The Netherlands
| | - Daumantas Matulis
- grid.6441.70000 0001 2243 2806Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences Center, Vilnius University, Saulėtekio 7, LT-10257 Vilnius, Lithuania
| |
Collapse
|
22
|
Nguyen PL, Elkamhawy A, Choi YH, Lee CH, Lee K, Cho J. Suppression of Tumor Growth and Cell Migration by Indole-Based Benzenesulfonamides and Their Synergistic Effects in Combination with Doxorubicin. Int J Mol Sci 2022; 23:ijms23179903. [PMID: 36077298 PMCID: PMC9456432 DOI: 10.3390/ijms23179903] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 08/26/2022] [Accepted: 08/29/2022] [Indexed: 11/25/2022] Open
Abstract
Pharmacological inhibition of the enzyme activity targeting carbonic anhydrases (CAs) demonstrated antiglaucoma and anticancer effects through pH control. Recently, we reported a series of indole-based benzenesulfonamides as potent CA inhibitors. The present study aimed to evaluate the antitumor effects of these compounds against various cancer cell lines, including breast cancer (MDA-MB-231, MCF-7, and SK-BR-3), lung cancer (A549), and pancreatic cancer (Panc1) cells. Overall, more potent cytotoxicity was observed on MCF-7 and SK-BR-3 cells than on lung or pancreatic cancer cells. Among the 15 compounds tested, A6 and A15 exhibited potent cytotoxic and antimigratory activities against MCF-7 and SK-BR-3 cells in the CoCl2-induced hypoxic condition. While A6 and A15 markedly reduced the viability of control siRNA-treated cells, these compounds could not significantly reduce the viability of CA IX-knockdown cells, suggesting the role of CA IX in their anticancer activities. To assess whether these compounds exerted synergism with a conventional anticancer drug doxorubicin (DOX), the cytotoxic effects of A6 or A15 combined with DOX were analyzed using Chou−Talalay and Bliss independence methods. Our data revealed that both A6 and A15 significantly enhanced the anticancer activity of DOX. Among the tested pairs, the combination of DOX with A15 showed the strongest synergism on SK-BR-3 cells. Moreover, this combination further attenuated cell migration compared to the respective drug. Collectively, our results demonstrated that A6 and A15 suppressed tumor growth and cell migration of MCF-7 and SK-BR-3 cells through inhibition of CA IX, and the combination of these compounds with DOX exhibited synergistic cytotoxic effects on these breast cancer cells. Therefore, A6 and A15 may serve as potential anticancer agents alone or in combination with DOX against breast cancer.
Collapse
Affiliation(s)
| | - Ahmed Elkamhawy
- College of Pharmacy, Dongguk University-Seoul, Goyang 10326, Korea
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| | - Young Hee Choi
- College of Pharmacy, Dongguk University-Seoul, Goyang 10326, Korea
| | - Chang Hoon Lee
- College of Pharmacy, Dongguk University-Seoul, Goyang 10326, Korea
| | - Kyeong Lee
- College of Pharmacy, Dongguk University-Seoul, Goyang 10326, Korea
- Correspondence: (K.L.); (J.C.)
| | - Jungsook Cho
- College of Pharmacy, Dongguk University-Seoul, Goyang 10326, Korea
- Correspondence: (K.L.); (J.C.)
| |
Collapse
|
23
|
Russell S, Xu L, Kam Y, Abrahams D, Ordway B, Lopez AS, Bui MM, Johnson J, Epstein T, Ruiz E, Lloyd MC, Swietach P, Verduzco D, Wojtkowiak J, Gillies RJ. Proton export upregulates aerobic glycolysis. BMC Biol 2022; 20:163. [PMID: 35840963 PMCID: PMC9287933 DOI: 10.1186/s12915-022-01340-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Accepted: 05/30/2022] [Indexed: 01/06/2023] Open
Abstract
INTRODUCTION Aggressive cancers commonly ferment glucose to lactic acid at high rates, even in the presence of oxygen. This is known as aerobic glycolysis, or the "Warburg Effect." It is widely assumed that this is a consequence of the upregulation of glycolytic enzymes. Oncogenic drivers can increase the expression of most proteins in the glycolytic pathway, including the terminal step of exporting H+ equivalents from the cytoplasm. Proton exporters maintain an alkaline cytoplasmic pH, which can enhance all glycolytic enzyme activities, even in the absence of oncogene-related expression changes. Based on this observation, we hypothesized that increased uptake and fermentative metabolism of glucose could be driven by the expulsion of H+ equivalents from the cell. RESULTS To test this hypothesis, we stably transfected lowly glycolytic MCF-7, U2-OS, and glycolytic HEK293 cells to express proton-exporting systems: either PMA1 (plasma membrane ATPase 1, a yeast H+-ATPase) or CA-IX (carbonic anhydrase 9). The expression of either exporter in vitro enhanced aerobic glycolysis as measured by glucose consumption, lactate production, and extracellular acidification rate. This resulted in an increased intracellular pH, and metabolomic analyses indicated that this was associated with an increased flux of all glycolytic enzymes upstream of pyruvate kinase. These cells also demonstrated increased migratory and invasive phenotypes in vitro, and these were recapitulated in vivo by more aggressive behavior, whereby the acid-producing cells formed higher-grade tumors with higher rates of metastases. Neutralizing tumor acidity with oral buffers reduced the metastatic burden. CONCLUSIONS Therefore, cancer cells which increase export of H+ equivalents subsequently increase intracellular alkalization, even without oncogenic driver mutations, and this is sufficient to alter cancer metabolism towards an upregulation of aerobic glycolysis, a Warburg phenotype. Overall, we have shown that the traditional understanding of cancer cells favoring glycolysis and the subsequent extracellular acidification is not always linear. Cells which can, independent of metabolism, acidify through proton exporter activity can sufficiently drive their metabolism towards glycolysis providing an important fitness advantage for survival.
Collapse
Affiliation(s)
- Shonagh Russell
- Cancer Physiology, Moffitt Cancer Center, 12902 USF Magnolia Dr, Tampa, FL 33612 USA
- Graduate School, University of South Florida, 4202 E Fowler Ave, Tampa, FL 33620 USA
| | - Liping Xu
- Cancer Physiology, Moffitt Cancer Center, 12902 USF Magnolia Dr, Tampa, FL 33612 USA
| | - Yoonseok Kam
- Agilent Technologies, 5301 Stevens Creek Blvd, Santa Clara, CA 9505 USA
| | - Dominique Abrahams
- Cancer Physiology, Moffitt Cancer Center, 12902 USF Magnolia Dr, Tampa, FL 33612 USA
| | - Bryce Ordway
- Cancer Physiology, Moffitt Cancer Center, 12902 USF Magnolia Dr, Tampa, FL 33612 USA
- Graduate School, University of South Florida, 4202 E Fowler Ave, Tampa, FL 33620 USA
| | - Alex S. Lopez
- Anatomic Pathology, Moffitt Cancer Center, 12902 USF Magnolia Dr, Tampa, FL 33612 USA
| | - Marilyn M. Bui
- Anatomic Pathology, Moffitt Cancer Center, 12902 USF Magnolia Dr, Tampa, FL 33612 USA
- Analytic Microscopy Core, Moffitt Cancer Center, 12902 USF Magnolia Dr, Tampa, FL 33612 USA
| | - Joseph Johnson
- Analytic Microscopy Core, Moffitt Cancer Center, 12902 USF Magnolia Dr, Tampa, FL 33612 USA
| | | | - Epifanio Ruiz
- Small Animal Imaging Department, Moffitt Cancer Center, 12902 USF Magnolia Dr, Tampa, FL 33612 USA
| | - Mark C. Lloyd
- Inspirata, Inc., One North Dale Mabry Hwy. Suite 600, Tampa, FL 33609 USA
| | - Pawel Swietach
- Department of Physiology, Anatomy and Genetics, University of Oxford, Parks Road, Oxford, OX1 3PT UK
| | - Daniel Verduzco
- Cancer Physiology, Moffitt Cancer Center, 12902 USF Magnolia Dr, Tampa, FL 33612 USA
| | - Jonathan Wojtkowiak
- Cancer Physiology, Moffitt Cancer Center, 12902 USF Magnolia Dr, Tampa, FL 33612 USA
| | - Robert J. Gillies
- Cancer Physiology, Moffitt Cancer Center, 12902 USF Magnolia Dr, Tampa, FL 33612 USA
| |
Collapse
|
24
|
Hypoxia-driven metabolic heterogeneity and immune evasive behaviour of gastrointestinal cancers: Elements of a recipe for disaster. Cytokine 2022; 156:155917. [PMID: 35660715 DOI: 10.1016/j.cyto.2022.155917] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 04/28/2022] [Accepted: 05/16/2022] [Indexed: 11/24/2022]
Abstract
Gastrointestinal (GI) cancers refer to a group of malignancies associated with the GI tract (GIT). Like other solid tumors, hypoxic regions consistently feature inside the GI tumor microenvironment (TME) and contribute towards metabolic reprogramming of tumor-resident cells by modulating hypoxia-induced factors. We highlight here how the metabolic crosstalk between cancer cells and immune cells generate immunosuppressive environment inside hypoxic tumors. Given the fluctuating nature of tumor hypoxia, the metabolic fluxes between immune cells and cancer cells change dynamically. These changes alter cellular phenotypes and functions, resulting in the acceleration of cancer progression. These evolved properties of hypoxic tumors make metabolism-targeting monotherapy approaches or immunotherapy-measures unsuccessful. The current review highlights the advantages of combined immunometabolic treatment strategies to target hypoxic GI cancers and also identifies research areas to develop better combinational therapeutics for future.
Collapse
|
25
|
Sarogni P, Mapanao AK, Gonnelli A, Ermini ML, Marchetti S, Kusmic C, Paiar F, Voliani V. Chorioallantoic membrane tumor models highlight the effects of cisplatin compounds in oral carcinoma treatment. iScience 2022; 25:103980. [PMID: 35310338 PMCID: PMC8924639 DOI: 10.1016/j.isci.2022.103980] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/31/2022] [Accepted: 02/19/2022] [Indexed: 12/12/2022] Open
Abstract
The European Society for Medical Oncology (ESMO) suggests the use of chemotherapy as neoadjuvant, adjuvant, and concomitant to surgery and radiotherapy for the treatment of oral carcinoma by depending on the cancer stage. The usual drug of choice belongs to the platinum compounds. In this context, the evaluation of the cancer behavior associated with the administration of standard or emerging cisplatin compounds supports the establishment of optimal cancer management. Here, we have assessed and compared the performance of cisplatin alone and contained in biodegradable nanocapsules on standardized chorioallantoic membrane (CAM) tumor models. The vascularized environment and optimized grafting procedure allowed the establishment of solid tumors. The treatments showed antitumor and anti-angiogenic activities together with deregulation of pivotal genes responsible of treatment resistance and tumor aggressiveness. This study further supports the significance of CAM tumor models in oncological research for the comprehension of the molecular mechanisms involved in tumor treatment response.
Collapse
Affiliation(s)
- Patrizia Sarogni
- Center for Nanotechnology Innovation@NEST, Istituto Italiano di Tecnologia, Piazza San Silvestro 12, Pisa, Italy
| | - Ana Katrina Mapanao
- Center for Nanotechnology Innovation@NEST, Istituto Italiano di Tecnologia, Piazza San Silvestro 12, Pisa, Italy
- NEST-Scuola Normale Superiore, Piazza San Silvestro 12, Pisa, Italy
| | - Alessandra Gonnelli
- Center for Nanotechnology Innovation@NEST, Istituto Italiano di Tecnologia, Piazza San Silvestro 12, Pisa, Italy
- Radiation Oncology Unit, Pisa University Hospital, Via Roma 67, Pisa, Italy
| | - Maria Laura Ermini
- Center for Nanotechnology Innovation@NEST, Istituto Italiano di Tecnologia, Piazza San Silvestro 12, Pisa, Italy
| | - Sabrina Marchetti
- Institute of Clinical Physiology, CNR, Via G. Moruzzi 1, Pisa, Italy
| | - Claudia Kusmic
- Institute of Clinical Physiology, CNR, Via G. Moruzzi 1, Pisa, Italy
| | - Fabiola Paiar
- Radiation Oncology Unit, Pisa University Hospital, Via Roma 67, Pisa, Italy
| | - Valerio Voliani
- Center for Nanotechnology Innovation@NEST, Istituto Italiano di Tecnologia, Piazza San Silvestro 12, Pisa, Italy
| |
Collapse
|
26
|
Koyuncu I, Temiz E, Durgun M, Kocyigit A, Yuksekdag O, Supuran CT. Intracellular pH-mediated induction of apoptosis in HeLa cells by a sulfonamide carbonic anhydrase inhibitor. Int J Biol Macromol 2022; 201:37-46. [PMID: 34999037 DOI: 10.1016/j.ijbiomac.2021.12.190] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 12/16/2021] [Accepted: 12/30/2021] [Indexed: 01/26/2023]
Abstract
Carbonic anhydrase IX (CAIX) is a hypoxia-associated transmembrane protein that is critical in the survival of cells. Because CAIX has a key role in pH regulation, its therapeutic effects have been heavily studied by different research laboratories. This study aims to investigate how a synthetic CAIX inhibitor triggers apoptosis in a cancer cell line, HeLa. In this regard, we investigated the effects of the compound I, synthesized as a CAIX inhibitor, on the survival of cancer cells. The compound I inhibited the proliferation of the CAIX+ HeLa cells, kept the cells in G0/G1 phase (74.7%) and altered the cells morphologies (AO/EtBr staining) and the nuclear structure (γ-H2AX staining). CAIX inhibition triggered apoptosis in HeLa cells with a rate of 47.4%. According to the expression of mediator genes (CASP-3, -8, -9, BAX, BCL-2, BECLIN, LC3), the both death pathways were activated in HeLa cells with the inhibition of CAIX with the compound I. The compound I was also determined to affect the genes and proteins that have a critical role in the regulation of apoptotic pathways (pro casp-3, cleaved casp-3, -8, -9, cleaved PARP and CAIX). Furthermore, CAIX inhibition caused changes in pH balance, disruption in organelle integrity of mitochondria, and increase intracellular reactive oxygen level of HeLa cells. Taken together, our findings suggest that CAIX inhibition has a potential in cancer treatment, and the compound I, a CAIX inhibitor, could be a promising therapeutic strategy in the treatment of aggressive tumours.
Collapse
Affiliation(s)
- Ismail Koyuncu
- Department of Medical Biochemistry, Faculty of Medicine, Harran University, Sanliurfa 63290, Turkey.
| | - Ebru Temiz
- Program of Medical Promo and Marketing, Health Services Vocational School, Harran University, Sanliurfa 63300, Turkey
| | - Mustafa Durgun
- Department of Chemistry, Faculty of Arts and Sciences, Harran University, Sanliurfa 63290, Turkey.
| | - Abdurrahim Kocyigit
- Department of Medical Biochemistry, Faculty of Medicine, Bezmialem Vakif University, Istanbul 34093, Turkey.
| | - Ozgur Yuksekdag
- Department of Medical Biochemistry, Faculty of Medicine, Harran University, Sanliurfa 63290, Turkey
| | - Claudiu T Supuran
- NEUROFARBA Department, Section of Pharmaceutical and Nutraceutical Sciences, Università degli Studi di Firenze, Sesto Fiorentino, Florence 50019, Italy.
| |
Collapse
|
27
|
Huo Z, Bilang R, Supuran CT, von der Weid N, Bruder E, Holland-Cunz S, Martin I, Muraro MG, Gros SJ. Perfusion-Based Bioreactor Culture and Isothermal Microcalorimetry for Preclinical Drug Testing with the Carbonic Anhydrase Inhibitor SLC-0111 in Patient-Derived Neuroblastoma. Int J Mol Sci 2022; 23:ijms23063128. [PMID: 35328549 PMCID: PMC8955558 DOI: 10.3390/ijms23063128] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 03/04/2022] [Accepted: 03/05/2022] [Indexed: 12/23/2022] Open
Abstract
Neuroblastoma is a rare disease. Rare are also the possibilities to test new therapeutic options for neuroblastoma in clinical trials. Despite the constant need to improve therapy and outcomes for patients with advanced neuroblastoma, clinical trials currently only allow for testing few substances in even fewer patients. This increases the need to improve and advance preclinical models for neuroblastoma to preselect favorable candidates for novel therapeutics. Here we propose the use of a new patient-derived 3D slice-culture perfusion-based 3D model in combination with rapid treatment evaluation using isothermal microcalorimetry exemplified with treatment with the novel carbonic anhydrase IX and XII (CAIX/CAXII) inhibitor SLC-0111. Patient samples showed a CAIX expression of 18% and a CAXII expression of 30%. Corresponding with their respective CAIX expression patterns, the viability of SH-EP cells was significantly reduced upon treatment with SLC-0111, while LAN1 cells were not affected. The inhibitory effect on SH-SY5Y cells was dependent on the induction of CAIX expression under hypoxia. These findings corresponded to thermogenesis of the cells. Patient-derived organotypic slice cultures were treated with SLC-0111, which was highly effective despite heterogeneity of CAIX/CAXII expression. Thermogenesis, in congruence with the findings of the histological observations, was significantly reduced in SLC-0111-treated samples. In order to extend the evaluation time, we established a perfusion-based approach for neuroblastoma tissue in a 3D perfusion-based bioreactor system. Using this system, excellent tissue quality with intact tumor cells and stromal structure in neuroblastoma tumors can be maintained for 7 days. The system was successfully used for consecutive drug response monitoring with isothermal microcalorimetry. The described approach for drug testing, relying on an advanced 3D culture system combined with a rapid and highly sensitive metabolic assessment, can facilitate development of personalized treatment strategies for neuroblastoma.
Collapse
Affiliation(s)
- Zihe Huo
- Department of Pediatric Surgery, University Children’s Hospital Basel, 4031 Basel, Switzerland; (Z.H.); (R.B.); (S.H.-C.)
- Department of Clinical Research, University of Basel, 4031 Basel, Switzerland;
| | - Remo Bilang
- Department of Pediatric Surgery, University Children’s Hospital Basel, 4031 Basel, Switzerland; (Z.H.); (R.B.); (S.H.-C.)
- Department of Clinical Research, University of Basel, 4031 Basel, Switzerland;
| | - Claudiu T. Supuran
- Department Neurofarba, Sezione di Scienze Farmaceutiche, University of Florence, 50121 Florence, Italy;
| | - Nicolas von der Weid
- Department of Clinical Research, University of Basel, 4031 Basel, Switzerland;
- Department of Hematology and Oncology, University Children’s Hospital Basel, 4031 Basel, Switzerland
| | - Elisabeth Bruder
- Institute of Pathology, University Hospital Basel, 4031 Basel, Switzerland;
| | - Stefan Holland-Cunz
- Department of Pediatric Surgery, University Children’s Hospital Basel, 4031 Basel, Switzerland; (Z.H.); (R.B.); (S.H.-C.)
- Department of Clinical Research, University of Basel, 4031 Basel, Switzerland;
| | - Ivan Martin
- Tissue Engineering, Department of Biomedicine, University of Basel and University Hospital Basel, 4031 Basel, Switzerland; (I.M.); (M.G.M.)
| | - Manuele G. Muraro
- Tissue Engineering, Department of Biomedicine, University of Basel and University Hospital Basel, 4031 Basel, Switzerland; (I.M.); (M.G.M.)
| | - Stephanie J. Gros
- Department of Pediatric Surgery, University Children’s Hospital Basel, 4031 Basel, Switzerland; (Z.H.); (R.B.); (S.H.-C.)
- Department of Clinical Research, University of Basel, 4031 Basel, Switzerland;
- Correspondence:
| |
Collapse
|
28
|
Harguindey S, Alfarouk K, Polo Orozco J, Reshkin SJ, Devesa J. Hydrogen Ion Dynamics as the Fundamental Link between Neurodegenerative Diseases and Cancer: Its Application to the Therapeutics of Neurodegenerative Diseases with Special Emphasis on Multiple Sclerosis. Int J Mol Sci 2022; 23:ijms23052454. [PMID: 35269597 PMCID: PMC8910484 DOI: 10.3390/ijms23052454] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 02/17/2022] [Accepted: 02/21/2022] [Indexed: 02/01/2023] Open
Abstract
The pH-related metabolic paradigm has rapidly grown in cancer research and treatment. In this contribution, this recent oncological perspective has been laterally assessed for the first time in order to integrate neurodegeneration within the energetics of the cancer acid-base conceptual frame. At all levels of study (molecular, biochemical, metabolic, and clinical), the intimate nature of both processes appears to consist of opposite mechanisms occurring at the far ends of a physiopathological intracellular pH/extracellular pH (pHi/pHe) spectrum. This wide-ranging original approach now permits an increase in our understanding of these opposite processes, cancer and neurodegeneration, and, as a consequence, allows us to propose new avenues of treatment based upon the intracellular and microenvironmental hydrogen ion dynamics regulating and deregulating the biochemistry and metabolism of both cancer and neural cells. Under the same perspective, the etiopathogenesis and special characteristics of multiple sclerosis (MS) is an excellent model for the study of neurodegenerative diseases and, utilizing this pioneering approach, we find that MS appears to be a metabolic disease even before an autoimmune one. Furthermore, within this paradigm, several important aspects of MS, from mitochondrial failure to microbiota functional abnormalities, are analyzed in depth. Finally, and for the first time, a new and integrated model of treatment for MS can now be advanced.
Collapse
Affiliation(s)
- Salvador Harguindey
- Division of Oncology, Institute of Clinical Biology and Metabolism, 01004 Vitoria, Spain;
- Correspondence: ; Tel.: +34-629-047-141
| | - Khalid Alfarouk
- Institute of Endemic Diseases, University of Khartoum, Khartoum 11111, Sudan;
| | - Julián Polo Orozco
- Division of Oncology, Institute of Clinical Biology and Metabolism, 01004 Vitoria, Spain;
| | - Stephan J Reshkin
- Department of Biosciences, Biotechnology and Biopharmaceutics, University of Bari, 70125 Bari, Italy;
| | - Jesús Devesa
- Scientific Direction, Foltra Medical Centre, 15886 Teo, Spain;
| |
Collapse
|
29
|
Zatovicova M, Kajanova I, Barathova M, Takacova M, Labudova M, Csaderova L, Jelenska L, Svastova E, Pastorekova S, Harris AL, Pastorek J. Novel humanized monoclonal antibodies for targeting hypoxic human tumors via two distinct extracellular domains of carbonic anhydrase IX. Cancer Metab 2022; 10:3. [PMID: 35109923 PMCID: PMC8811981 DOI: 10.1186/s40170-022-00279-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 01/11/2022] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Hypoxia in the tumor microenvironment (TME) is often the main factor in the cancer progression. Moreover, low levels of oxygen in tumor tissue may signal that the first- or second-line therapy will not be successful. This knowledge triggers the inevitable search for different kinds of treatment that will successfully cure aggressive tumors. Due to its exclusive expression on cancer cells, carbonic anhydrase IX belongs to the group of the most precise targets in hypoxic tumors. CA IX possesses several exceptional qualities that predetermine its crucial role in targeted therapy. Its expression on the cell membrane makes it an easily accessible target, while its absence in healthy corresponding tissues makes the treatment practically harmless. The presence of CA IX in solid tumors causes an acidic environment that may lead to the failure of standard therapy. METHODS Parental mouse hybridomas (IV/18 and VII/20) were humanized to antibodies which were subsequently named CA9hu-1 and CA9hu-2. From each hybridoma, we obtained 25 clones. Each clone was tested for antibody-dependent cellular cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC) activity, affinity, extracellular pH measurement, multicellular aggregation analysis, and real-time monitoring of invasion with the xCELLigence system. RESULTS Based on the results from in vivo experiments, we have selected mouse monoclonal antibodies VII/20 and IV/18. The first one is directed at the conformational epitope of the catalytic domain, internalizes after binding to the antigen, and halts tumor growth while blocking extracellular acidification. The second targets the sequential epitope of the proteo-glycan domain, does not internalize, and is able to block the attachment of cancer cells to the matrix preventing metastasis formation. In vitro experiments prove that humanized versions of the parental murine antibodies, CA9hu-1 and CA9hu-2, have preserved these characteristics. They can reverse the failure of standard therapy as a result of an acidic environment by modulating the TME, and both are able to induce an immune response and have high affinity, as well as ADCC and CDC activity. CONCLUSION CA9hu-1 and CA9hu-2 are the very first humanized antibodies against CA IX that are likely to become suitable therapies for hypoxic tumors. These antibodies can be applied in the treatment therapy of primary tumors and suppression of metastases formation.
Collapse
Affiliation(s)
- Miriam Zatovicova
- MABPRO, a.s., Dubravska cesta 2, 841 04, Bratislava, Slovakia
- Department of Tumor Biology, Institute of Virology, Biomedical Research Center, Slovak Academy of Sciences, Dubravska cesta 9, 84505, Bratislava, Slovakia
| | - Ivana Kajanova
- Department of Tumor Biology, Institute of Virology, Biomedical Research Center, Slovak Academy of Sciences, Dubravska cesta 9, 84505, Bratislava, Slovakia
| | - Monika Barathova
- MABPRO, a.s., Dubravska cesta 2, 841 04, Bratislava, Slovakia
- Department of Tumor Biology, Institute of Virology, Biomedical Research Center, Slovak Academy of Sciences, Dubravska cesta 9, 84505, Bratislava, Slovakia
| | - Martina Takacova
- Department of Tumor Biology, Institute of Virology, Biomedical Research Center, Slovak Academy of Sciences, Dubravska cesta 9, 84505, Bratislava, Slovakia
| | - Martina Labudova
- MABPRO, a.s., Dubravska cesta 2, 841 04, Bratislava, Slovakia
- Department of Tumor Biology, Institute of Virology, Biomedical Research Center, Slovak Academy of Sciences, Dubravska cesta 9, 84505, Bratislava, Slovakia
| | - Lucia Csaderova
- Department of Tumor Biology, Institute of Virology, Biomedical Research Center, Slovak Academy of Sciences, Dubravska cesta 9, 84505, Bratislava, Slovakia
| | - Lenka Jelenska
- Department of Tumor Biology, Institute of Virology, Biomedical Research Center, Slovak Academy of Sciences, Dubravska cesta 9, 84505, Bratislava, Slovakia
| | - Eliska Svastova
- Department of Tumor Biology, Institute of Virology, Biomedical Research Center, Slovak Academy of Sciences, Dubravska cesta 9, 84505, Bratislava, Slovakia
| | - Silvia Pastorekova
- Department of Tumor Biology, Institute of Virology, Biomedical Research Center, Slovak Academy of Sciences, Dubravska cesta 9, 84505, Bratislava, Slovakia
| | - Adrian L Harris
- Department of Oncology, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford, OX3 7DQ, UK
| | | |
Collapse
|
30
|
Understanding metabolic alterations and heterogeneity in cancer progression through validated immunodetection of key molecular components: a case of carbonic anhydrase IX. Cancer Metastasis Rev 2022; 40:1035-1053. [PMID: 35080763 PMCID: PMC8825433 DOI: 10.1007/s10555-021-10011-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 12/08/2021] [Indexed: 12/22/2022]
Abstract
Cancer metabolic heterogeneity develops in response to both intrinsic factors (mutations leading to activation of oncogenic pathways) and extrinsic factors (physiological and molecular signals from the extracellular milieu). Here we review causes and consequences of metabolic alterations in cancer cells with focus on hypoxia and acidosis, and with particular attention to carbonic anhydrase IX (CA IX). CA IX is a cancer-associated enzyme induced and activated by hypoxia in a broad range of tumor types, where it participates in pH regulation as well as in molecular mechanisms supporting cancer cells’ invasion and metastasis. CA IX catalyzes reversible conversion of carbon dioxide to bicarbonate ion plus proton and cooperates with a spectrum of molecules transporting ions or metabolites across the plasma membrane. Thereby CA IX contributes to extracellular acidosis as well as to buffering intracellular pH, which is essential for cell survival, metabolic performance, and proliferation of cancer cells. Since CA IX expression pattern reflects gradients of oxygen, pH, and other intratumoral factors, we use it as a paradigm to discuss an impact of antibody quality and research material on investigating metabolic reprogramming of tumor tissue. Based on the validation, we propose the most reliable CA IX-specific antibodies and suggest conditions for faithful immunohistochemical analysis of molecules contributing to heterogeneity in cancer progression.
Collapse
|
31
|
van Genugten EAJ, Weijers JAM, Heskamp S, Kneilling M, van den Heuvel MM, Piet B, Bussink J, Hendriks LEL, Aarntzen EHJG. Imaging the Rewired Metabolism in Lung Cancer in Relation to Immune Therapy. Front Oncol 2022; 11:786089. [PMID: 35070990 PMCID: PMC8779734 DOI: 10.3389/fonc.2021.786089] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 12/10/2021] [Indexed: 12/14/2022] Open
Abstract
Metabolic reprogramming is recognized as one of the hallmarks of cancer. Alterations in the micro-environmental metabolic characteristics are recognized as important tools for cancer cells to interact with the resident and infiltrating T-cells within this tumor microenvironment. Cancer-induced metabolic changes in the micro-environment also affect treatment outcomes. In particular, immune therapy efficacy might be blunted because of somatic mutation-driven metabolic determinants of lung cancer such as acidity and oxygenation status. Based on these observations, new onco-immunological treatment strategies increasingly include drugs that interfere with metabolic pathways that consequently affect the composition of the lung cancer tumor microenvironment (TME). Positron emission tomography (PET) imaging has developed a wide array of tracers targeting metabolic pathways, originally intended to improve cancer detection and staging. Paralleling the developments in understanding metabolic reprogramming in cancer cells, as well as its effects on stromal, immune, and endothelial cells, a wave of studies with additional imaging tracers has been published. These tracers are yet underexploited in the perspective of immune therapy. In this review, we provide an overview of currently available PET tracers for clinical studies and discuss their potential roles in the development of effective immune therapeutic strategies, with a focus on lung cancer. We report on ongoing efforts that include PET/CT to understand the outcomes of interactions between cancer cells and T-cells in the lung cancer microenvironment, and we identify areas of research which are yet unchartered. Thereby, we aim to provide a starting point for molecular imaging driven studies to understand and exploit metabolic features of lung cancer to optimize immune therapy.
Collapse
Affiliation(s)
- Evelien A J van Genugten
- Department of Medical Imaging, Radboud University Medical Centre (Radboudumc), Nijmegen, Netherlands
| | - Jetty A M Weijers
- Department of Medical Imaging, Radboud University Medical Centre (Radboudumc), Nijmegen, Netherlands
| | - Sandra Heskamp
- Department of Medical Imaging, Radboud University Medical Centre (Radboudumc), Nijmegen, Netherlands
| | - Manfred Kneilling
- Department of Preclinical Imaging and Radiopharmacy, Werner Siemens Imaging Center, Eberhard Karls University, Tuebingen, Germany.,Department of Dermatology, Eberhard Karls University, Tuebingen, Germany
| | | | - Berber Piet
- Department of Respiratory Diseases, Radboudumc, Nijmegen, Netherlands
| | - Johan Bussink
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboudumc, Netherlands
| | - Lizza E L Hendriks
- Department of Pulmonary Diseases, GROW - School for Oncology and Developmental Biology, Maastricht University Medical Centre (UMC), Maastricht, Netherlands
| | - Erik H J G Aarntzen
- Department of Medical Imaging, Radboud University Medical Centre (Radboudumc), Nijmegen, Netherlands
| |
Collapse
|
32
|
Therapeutic targeting of the hypoxic tumour microenvironment. Nat Rev Clin Oncol 2021; 18:751-772. [PMID: 34326502 DOI: 10.1038/s41571-021-00539-4] [Citation(s) in RCA: 242] [Impact Index Per Article: 60.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/18/2021] [Indexed: 02/07/2023]
Abstract
Hypoxia is prevalent in human tumours and contributes to microenvironments that shape cancer evolution and adversely affect therapeutic outcomes. Historically, two different tumour microenvironment (TME) research communities have been discernible. One has focused on physicochemical gradients of oxygen, pH and nutrients in the tumour interstitium, motivated in part by the barrier that hypoxia poses to effective radiotherapy. The other has focused on cellular interactions involving tumour and non-tumour cells within the TME. Over the past decade, strong links have been established between these two themes, providing new insights into fundamental aspects of tumour biology and presenting new strategies for addressing the effects of hypoxia and other microenvironmental features that arise from the inefficient microvascular system in solid tumours. This Review provides a perspective on advances at the interface between these two aspects of the TME, with a focus on translational therapeutic opportunities relating to the elimination and/or exploitation of tumour hypoxia.
Collapse
|
33
|
Datta A, West C, O'Connor JPB, Choudhury A, Hoskin P. Impact of hypoxia on cervical cancer outcomes. Int J Gynecol Cancer 2021; 31:1459-1470. [PMID: 34593564 DOI: 10.1136/ijgc-2021-002806] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 09/14/2021] [Indexed: 01/22/2023] Open
Abstract
The annual global incidence of cervical cancer is approximately 604 000 cases/342 000 deaths, making it the fourth most common cancer in women. Cervical cancer is a major healthcare problem in low and middle income countries where 85% of new cases and deaths occur. Secondary prevention measures have reduced incidence and mortality in developed countries over the past 30 years, but cervical cancer remains a major cause of cancer deaths in women. For women who present with Fédération Internationale de Gynécologie et d'Obstétrique (FIGO 2018) stages IB3 or upwards, chemoradiation is the established treatment. Despite high rates of local control, overall survival is less than 50%, largely due to distant relapse. Reducing the health burden of cervical cancer requires greater individualization of treatment, identifying those at risk of relapse and progression for modified or intensified treatment. Hypoxia is a well known feature of solid tumors and an established therapeutic target. Low tumorous oxygenation increases the risk of local invasion, metastasis and treatment failure. While meta-analyses show benefit, many individual trials targeting hypoxia failed in part due to not selecting patients most likely to benefit. This review summarizes the available hypoxia-targeted strategies and identifies further research and new treatment paradigms needed to improve patient outcomes. The applications and limitations of hypoxia biomarkers for treatment selection and response monitoring are discussed. Finally, areas of greatest unmet clinical need are identified to measure and target hypoxia and therefore improve cervical cancer outcomes.
Collapse
Affiliation(s)
- Anubhav Datta
- Division of Cancer Sciences, The University of Manchester Faculty of Biology Medicine and Health, Manchester, UK
- Clinical Radiology, The Christie NHS Foundation Trust, Manchester, UK
| | - Catharine West
- Division of Cancer Sciences, The University of Manchester Faculty of Biology Medicine and Health, Manchester, UK
| | - James P B O'Connor
- Division of Cancer Sciences, The University of Manchester Faculty of Biology Medicine and Health, Manchester, UK
- Division of Radiotherapy and Imaging, Institute of Cancer Research, London, UK
| | - Ananya Choudhury
- Division of Cancer Sciences, The University of Manchester Faculty of Biology Medicine and Health, Manchester, UK
- Clinical Oncology, The Christie Hospital NHS Trust, Manchester, UK
| | - Peter Hoskin
- Division of Cancer Sciences, The University of Manchester Faculty of Biology Medicine and Health, Manchester, UK
- Clinical Oncology, Mount Vernon Cancer Centre, Northwood, Middlesex, UK
| |
Collapse
|
34
|
Keerthiga R, Pei DS, Fu A. Mitochondrial dysfunction, UPR mt signaling, and targeted therapy in metastasis tumor. Cell Biosci 2021; 11:186. [PMID: 34717757 PMCID: PMC8556915 DOI: 10.1186/s13578-021-00696-0] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 10/02/2021] [Indexed: 12/13/2022] Open
Abstract
In modern research, mitochondria are considered a more crucial energy plant in cells. Mitochondrial dysfunction, including mitochondrial DNA (mtDNA) mutation and denatured protein accumulation, is a common feature of tumors. The dysfunctional mitochondria reprogram molecular metabolism and allow tumor cells to proliferate in the hostile microenvironment. One of the crucial signaling pathways of the mitochondrial dysfunction activation in the tumor cells is the retrograde signaling of mitochondria-nucleus interaction, mitochondrial unfolded protein response (UPRmt), which is initiated by accumulation of denatured protein and excess ROS production. In the process of UPRmt, various components are activitated to enhance the mitochondria-nucleus retrograde signaling to promote carcinoma progression, including hypoxia-inducible factor (HIF), activating transcription factor ATF-4, ATF-5, CHOP, AKT, AMPK. The retrograde signaling molecules of overexpression ATF-5, SIRT3, CREB, SOD1, SOD2, early growth response protein 1 (EGR1), ATF2, CCAAT/enhancer-binding protein-d, and CHOP also involved in the process. Targeted blockage of the UPRmt pathway could obviously inhibit tumor proliferation and metastasis. This review indicates the UPRmt pathways and its crucial role in targeted therapy of metastasis tumors.
Collapse
Affiliation(s)
| | - De-Sheng Pei
- School of Public Health and Management, Chongqing Medical University, Chongqing, 400016, China.
| | - Ailing Fu
- College of Pharmaceutical Sciences, Southwest University, Chongqing, China.
| |
Collapse
|
35
|
Nortunen M, Parkkila S, Saarnio J, Huhta H, Karttunen TJ. Carbonic Anhydrases II and IX in Non-ampullary Duodenal Adenomas and Adenocarcinoma. J Histochem Cytochem 2021; 69:677-690. [PMID: 34636283 DOI: 10.1369/00221554211050133] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Non-ampullary duodenal adenocarcinoma (DAC) is a rare malignancy. Little information is available concerning the histopathological prognostic factors associated with DAC. Carbonic anhydrases (CAs) are metalloenzymes catalyzing the universal reaction of CO2 hydration. Isozymes CAII, CAIX, and CAXII are associated with prognosis in various cancers. Our aim was to analyze the immunohistochemical expressions of CAII, CAIX, and CAXII in normal duodenal epithelium, duodenal adenomas, and adenocarcinoma and their associations with clinicopathological variables and survival. Our retrospective study included all 27 DACs treated in Oulu University Hospital during years 2000-2020. For comparison, samples of 42 non-ampullary adenomas were collected. CAII expression was low in duodenal adenomas and adenocarcinoma. CAIX expression in adenomas and adenocarcinoma was comparable with the high expression of normal duodenal crypts. Expression patterns in carcinomas were largely not related to clinicopathological features. However, low expression of CAII associated with poorer differentiation of the tumor (p=0.049) and low expression of CAIX showed a trend for association with nodal spread, although statistical significance was not reached (p=0.091). CAII and CAIX lost their epithelial polarization and staining intensity in adenomas. CAXII expression was not detected in the studied samples. CAs were not associated with survival. The prognostic value of CAII and CAIX downregulation should be further investigated. Both isozymes may serve as biomarkers of epithelial dysplasia in the duodenum.
Collapse
Affiliation(s)
- Minna Nortunen
- Research Unit of Surgery, Anesthesia and Intensive Care, University of Oulu, Oulu, Finland.,Department of Surgery, Oulu University Hospital and Medical Research Center Oulu, Oulu, Finland.,Cancer and Translational Medicine Research Unit, Medical Research Center Oulu, University of Oulu and Oulu University Hospital, Oulu, Finland
| | - Seppo Parkkila
- Faculty of Medicine and Health Technology, Tampere University and Fimlab Ltd, Tampere University Hospital, Tampere, Finland (SP)
| | - Juha Saarnio
- Research Unit of Surgery, Anesthesia and Intensive Care, University of Oulu, Oulu, Finland.,Department of Surgery, Oulu University Hospital and Medical Research Center Oulu, Oulu, Finland.,Cancer and Translational Medicine Research Unit, Medical Research Center Oulu, University of Oulu and Oulu University Hospital, Oulu, Finland
| | - Heikki Huhta
- Research Unit of Surgery, Anesthesia and Intensive Care, University of Oulu, Oulu, Finland.,Department of Surgery, Oulu University Hospital and Medical Research Center Oulu, Oulu, Finland.,Cancer and Translational Medicine Research Unit, Medical Research Center Oulu, University of Oulu and Oulu University Hospital, Oulu, Finland
| | - Tuomo J Karttunen
- Cancer and Translational Medicine Research Unit, Medical Research Center Oulu, University of Oulu and Oulu University Hospital, Oulu, Finland
| |
Collapse
|
36
|
Kim EK, Kim HM, Koo JS. Expression of Glucose Metabolism-Related Proteins in Adrenal Neoplasms. Pathobiology 2021; 88:424-433. [PMID: 34518477 DOI: 10.1159/000518208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 07/01/2021] [Indexed: 11/19/2022] Open
Abstract
PURPOSE The aim of this study was to investigate the expression patterns of glucose metabolism-related proteins and their clinicopathologic implications in adrenal cortical neoplasms (ACN) and pheochromocytoma (PCC). METHODS Immunohistochemical staining was performed to evaluate glucose metabolism-related proteins (GLUT1, CAIX, hexokinase II, G6PDH, PHGDH, and SHMT1) in 132 ACN cases (115 adrenal cortical adenoma [ACA] and 17 adrenal cortical carcinoma [ACC]) and 189 PCC cases. RESULTS Expression levels of GLUT1 in tumor cells ([T]; p < 0.001), GLUT1 in stromal cells ([S]; p < 0.001), G6PDH (p < 0.001), and SHMT1 (p = 0.002) were higher in ACN than in PCC. GLUT1 (T; p = 0.045) and PHGDH (p = 0.043) levels were higher in ACC than in ACA. In a univariate analysis of ACN, GLUT1 (T; p = 0.017), CAIX (S; p = 0.003), and PHGDH (p = 0.009) levels were correlated with a shorter overall survival (OS). GLUT1 (T; p = 0.001) and PHGDH (p < 0.001) were related to a shorter OS in PCC. GLUT1 (T) positivity (p = 0.043) in ACN predicted a poor OS in a multivariate Cox analysis. In PCC, high GAPP score (p = 0.026), GLUT1 (T; p = 0.002), and PHGDH (p < 0.001) were independent prognostic factors for poor OS. CONCLUSIONS The adrenal gland tumors ACN and PCC had different expression patterns of glucose metabolism-related proteins (GLUT1, G6PDH, and SHMT1), with higher expression levels in ACN than in PCC. GLUT1 and PHGDH were significant prognostic factors in these adrenal neoplasms.
Collapse
Affiliation(s)
- Eun Kyung Kim
- Department of Pathology, National Health Insurance Service Ilsan Hospital, Goyang, Republic of Korea
| | - Hye Min Kim
- Department of Pathology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Ja Seung Koo
- Department of Pathology, Yonsei University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
37
|
Huizing FJ, Hoeben BAW, Lok J, Boerman OC, Heskamp S, Bussink J. Imaging carbonic anhydrase IX as a method for monitoring hypoxia-related radioresistance in preclinical head and neck cancer models. PHYSICS & IMAGING IN RADIATION ONCOLOGY 2021; 19:145-150. [PMID: 34485720 PMCID: PMC8397885 DOI: 10.1016/j.phro.2021.08.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 07/21/2021] [Accepted: 08/11/2021] [Indexed: 12/03/2022]
Abstract
Background and purpose Tumor hypoxia is an important cause of radioresistance and is associated with poor outcome. SPECT (Single-photon emission computed tomography) imaging enables visualizing tumor characteristics. We investigated the SPECT-radiotracer [111In]-girentuximab-F(ab’)2 to image Carbonic Anhydrase IX (CAIX), an enzyme upregulated under hypoxic conditions. Materials and methods Athymic mice with subcutaneous FaDu or SCCNij202 head and neck squamous cell carcinoma (HNSCC) xenografts were treated with atovaquone or were housed in a hypoxic chamber (8% O2). Next, [111In]-girentuximab-F(ab’)2 was injected and 24 h later mice were euthanized for ex vivo biodistribution, autoradiography of the tumor, and immunohistochemical staining of the tumor. Tumor sections were analyzed for hypoxia, CAIX expression, vessels, and perfusion. Also, the effect of atovaquone on microSPECT scans was determined in the FaDu model. Results Atovaquone decreased CAIX expression by 69% (p = 0.017) compared with control tumors in FaDu, while in the SCCNij202 tumors no difference was observed. Hypoxic breathing did not increase CAIX expression or hypoxia staining in either tumor model, but did affect the necrotic tumor fraction. Ex vivo tracer uptake in the atovaquone treated group did not differ significantly from the control group, despite the difference in CAIX expression. Furthermore, SPECT imaging with [111In]-girentuximab-F(ab’)2 did not discriminate atovaquone-treated versus control tumors. Conclusion Atovaquone decreased CAIX expression only in the FaDu tumor model. [111In]-girentuximab-F(ab’)2 specifically targets CAIX-expressing areas in HNSCC xenografts, but differences in vessel density and necrosis most likely affected tracer uptake in the tumors and therefore complicated quantification of changes in CAIX expression.
Collapse
Affiliation(s)
- Fokko J Huizing
- Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Bianca A W Hoeben
- Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Jasper Lok
- Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Otto C Boerman
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Sandra Heskamp
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Johan Bussink
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
38
|
Carbonic Anhydrase IX and Survivin in Colorectal Adenocarcinoma Cells: Slovakian Population Study. BIOLOGY 2021; 10:biology10090872. [PMID: 34571749 PMCID: PMC8466885 DOI: 10.3390/biology10090872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 08/20/2021] [Accepted: 08/28/2021] [Indexed: 11/17/2022]
Abstract
Simple Summary This retrospective study (Slovakian population study) brings information about immunohistochemical detection of CAIX and survivin in 74 samples of human colorectal adenocarcinoma and comparison their expression with expression in healthy colon tissue. Our results show that all of samples with healthy colon tissue were CAIX and survivin-negative and there is no statistically significant dependence of these proteins and the chosen clinicopathological parameters. These findings demonstrate that detection of these proteins could be useful for tumor diagnostic and prognostic and CAIX and survivin could represent independent negative prognostic markers of colorectal cancer. Abstract The aim of this study was to detect carbonic anhydrase IX (CAIX) and survivin in the colorectal adenocarcinoma cells of the Slovakian population. We used an indirect three-step immunohistochemical method with DAB staining for the localization of the proteins and investigation their expression. We compared their expression with expression in healthy colorectal tissue. In 74 tissues of colorectal adenocarcinomas, 42% showed CAIX positivity and 20% showed survivin positivity. Brown membrane immunostaining was visible in CAIX-positive tumors. Survivin-positive tumors had strong brown cytoplasmic immunostaining. Co-expression of both proteins was present in five specimens (7%). The samples of normal colorectal tissue (without carcinoma) were CAIX-negative and survivin-negative. We also applied the Chi-squared test for evaluation statistically significant association between the expression of proteins and selected clinical and histopathological parameters. We did not find any statistically significant correlations between CAIX or survivin expression and sex of patients, the grade of the tumor, nodal status and presence of metastasis (p > 0.05). The fact that all samples of normal colorectal tissue were CAIX- and survivin-negative could lead to the possibility of using these two proteins as potential tumor diagnostic markers. On the basic of the available publications and data, we suggest that CAIX and survivin could be negative independent prognostic markers of colorectal cancer, which could affect response to therapy.
Collapse
|
39
|
Hypoxic Jumbo Spheroids On-A-Chip (HOnAChip): Insights into Treatment Efficacy. Cancers (Basel) 2021; 13:cancers13164046. [PMID: 34439199 PMCID: PMC8394550 DOI: 10.3390/cancers13164046] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 08/01/2021] [Accepted: 08/07/2021] [Indexed: 12/25/2022] Open
Abstract
Hypoxia is a key characteristic of the tumor microenvironment, too rarely considered during drug development due to the lack of a user-friendly method to culture naturally hypoxic 3D tumor models. In this study, we used soft lithography to engineer a microfluidic platform allowing the culture of up to 240 naturally hypoxic tumor spheroids within an 80 mm by 82.5 mm chip. These jumbo spheroids on a chip are the largest to date (>750 µm), and express gold-standard hypoxic protein CAIX at their core only, a feature absent from smaller spheroids of the same cell lines. Using histopathology, we investigated response to combined radiotherapy (RT) and hypoxic prodrug Tirapazamine (TPZ) on our jumbo spheroids produced using two sarcoma cell lines (STS117 and SK-LMS-1). Our results demonstrate that TPZ preferentially targets the hypoxic core (STS117: p = 0.0009; SK-LMS-1: p = 0.0038), but the spheroids' hypoxic core harbored as much DNA damage 24 h after irradiation as normoxic spheroid cells. These results validate our microfluidic device and jumbo spheroids as potent fundamental and pre-clinical tools for the study of hypoxia and its effects on treatment response.
Collapse
|
40
|
Esteves M, Monteiro MP, Duarte JA. The Effects of Physical Exercise on Tumor Vasculature: Systematic Review and Meta-analysis. Int J Sports Med 2021; 42:1237-1249. [PMID: 34341974 DOI: 10.1055/a-1533-1876] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
A wealth of evidence supports an association between physical exercise, decreased tumor growth rate, and reduced risk of cancer mortality. In this context, the tumor vascular microenvironment may play a key role in modulating tumor biologic behavior. The present systematic review and meta-analysis aimed to summarize the evidence regarding the effects of physical exercise on tumor vasculature in pre-clinical studies. We performed a computerized research on the PubMed, Scopus, and EBSCO databases to identify pre-clinical studies that evaluated the effect of physical exercise on tumor vascular outcomes. Mean differences were calculated through a random effects model. The present systematic review included 13 studies involving 373 animals. From these, 11 studies evaluated chronic intratumoral vascular adaptations and 2 studies assessed the acute intratumoral vascular adaptations to physical exercise. The chronic intratumoral vascular adaptations resulted in higher tumor microvessel density in 4 studies, increased tumor perfusion in 2 studies, and reduced intratumoral hypoxia in 3 studies. Quantitatively, regular physical exercise induced an increased tumor vascularization of 2.13 [1.07, 3.20] (p<0.0001). The acute intratumoral vascular adaptations included increased vascular conductance and reduced vascular resistance, which improved tumor perfusion and attenuated intratumoral hypoxia. In pre-clinical studies, physical exercise seems to improve tumor vascularization.
Collapse
Affiliation(s)
- Mário Esteves
- Laboratory of Biochemistry and Experimental Morphology, CIAFEL, Porto, Portugal.,Department of Physical Medicine and Rehabilitation, Teaching Hospital of the Fernando Pessoa University, Gondomar, Portugal
| | - Mariana P Monteiro
- Department of Anatomy, Universidade do Porto Instituto de Ciências Biomédicas Abel Salazar, Porto, Portugal
| | - Jose Alberto Duarte
- Laboratory of Biochemistry and Experimental Morphology, CIAFEL, Porto, Portugal.,TOXRUN - Toxicology Research Unit, University Institute of Health Sciences, CESPU, CRL, Gandra, Portugal
| |
Collapse
|
41
|
Xu P, Zhang Y, Ge F, Zhang F, He X, Gao X. Modulation of Tumor Microenvironment to Enhance Radiotherapy Efficacy in Esophageal Squamous Cell Carcinoma by Inhibiting Carbonic Anhydrase IX. Front Oncol 2021; 11:637252. [PMID: 34249682 PMCID: PMC8267588 DOI: 10.3389/fonc.2021.637252] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 06/09/2021] [Indexed: 02/01/2023] Open
Abstract
The radiotherapy outcomes of patients with advanced esophageal squamous cell carcinoma (ESCC) remain poor due to hypoxia. Carbonic anhydrase IX (CAIX) is a membrane-associated enzyme that induces hypoxia, extracellular acidity, and upregulation of hypoxia-related factors in tumor microenvironment, thereby promoting tumor metastasis. CAIX is upregulated in ESCC tissues compared to normal surrounding tissues. In the current study, we aimed to investigate the effect of CAIX inhibition on the modulation of tumor microenvironment and radiotherapy efficacy in ESCC. Higher CAIX expression was correlated with poorer progression-free survival in ESCC patients. Then, the ethyl N-(4-methylphenyl) sulfonylcarbamate (S4) was used to inhibit CAIX expression in ESCC cells and mice xenografts. The pretreatment of ESCC cells with S4 significantly downregulated CAIX expression, decreased intracellular pH, reduced cell viability, resulting in decreased oxygen consumption and more sensitive response to X-ray irradiation. In mice inoculated with ESCC cells, the combination of X-ray irradiation with S4 further improved survival, delayed tumor growth, decreased hypoxia level, exaggerated DNA damage, and increased apoptosis compared with the groups treated solely with S4 or radiotherapy. In conclusion, our study showed that the inhibition of CAIX by S4 treatment altered hypoxic tumor micro-environment, exaggerated DNA damage, increased apoptosis, and thus enhanced radiotherapy efficacy in ESCC. These findings provided a potential therapeutic strategy for patients with resistant ESCC.
Collapse
Affiliation(s)
- Pengqin Xu
- The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, China.,The Affiliated Tumor Hospital of Nantong University, Nantong Tumor Hospital, Nantong, China
| | - Yu Zhang
- The Affiliated Tumor Hospital of Nantong University, Nantong Tumor Hospital, Nantong, China
| | - Fanghong Ge
- The Affiliated Tumor Hospital of Nantong University, Nantong Tumor Hospital, Nantong, China
| | - Fuming Zhang
- The Affiliated Tumor Hospital of Nantong University, Nantong Tumor Hospital, Nantong, China
| | - Xia He
- The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, China
| | - Xingya Gao
- Department of Physiology, Nanjing Medical University, Nanjing, China
| |
Collapse
|
42
|
Bonatelli M, Fornari IF, Bernécule PN, Pinheiro LE, Costa RFA, Longatto-Filho A, Junior JNA, Silva ECA, Cárcano FM, Pinheiro C. Expression of Glycolysis-Related Proteins in Cancer of Unknown Primary Origin. Front Oncol 2021; 11:682665. [PMID: 34249728 PMCID: PMC8264765 DOI: 10.3389/fonc.2021.682665] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 06/07/2021] [Indexed: 12/27/2022] Open
Abstract
Introduction Cancer of unknown primary origin (CUP) is defined as metastatic cancer without identification of the primary site. Considering that only 15–20% of patients with CUP show a favorable outcome, identifying biomarkers may help improve the clinical management of patients who do not respond well to conventional therapies. In this context, the study of the metabolic profile of CUP may pave the way to establish new biomarkers and/or therapeutic targets; therefore, this study aimed to characterize the expression of metabolism-related proteins in CUP. Materials and Methods The expression of monocarboxylate transporters MCT1, MCT2 and MCT4, their chaperone CD147, the glucose transporter GLUT1 and the pH regulator CAIX was evaluated by immunohistochemistry in a series of 118 CUP patients, and the results were associated with the available clinicopathological information. Results The metabolism-related proteins MCT1, MCT4, CD147, GLUT1 and CAIX were expressed in a critical portion of the CUP (approximately 20 to 70%). MCT1 and CD147 were both more frequently expressed in cases with lymph nodes as metastasis dominant sites (p = 0.001) as well as in samples from lymph nodes (p <0.001 and p = 0.002, respectively), while MCT1 expression was more frequently expressed in squamous cell carcinomas (p = 0.045). A higher overall survival was observed in patients with tumors positive for GLUT1 and CAIX expression (p = 0.011 and p = 0.041, respectively), but none of the proteins was an independent prognostic factor for overall survival in multivariable analysis. Conclusion The results suggest that a portion of CUPs present a hyperglycolytic phenotype, which is associated with higher overall survival.
Collapse
Affiliation(s)
- Murilo Bonatelli
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, Brazil
| | - Isabella Fernandes Fornari
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, Brazil.,Barretos School of Health Sciences Dr. Paulo Prata-FACISB, Barretos, Brazil
| | - Priscila Neves Bernécule
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, Brazil.,Barretos School of Health Sciences Dr. Paulo Prata-FACISB, Barretos, Brazil
| | - Lara Esquiapatti Pinheiro
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, Brazil.,Barretos School of Health Sciences Dr. Paulo Prata-FACISB, Barretos, Brazil
| | - Ricardo Filipe Alves Costa
- Barretos School of Health Sciences Dr. Paulo Prata-FACISB, Barretos, Brazil.,Research and Teaching Institute, Barretos Cancer Hospital, Barretos, Brazil
| | - Adhemar Longatto-Filho
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, Brazil.,Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga, Portugal.,Laboratory of Medical Investigation (LIM-14), School of Medicine, University of São Paulo, São Paulo, Brazil
| | | | | | - Flávio Mavignier Cárcano
- Barretos School of Health Sciences Dr. Paulo Prata-FACISB, Barretos, Brazil.,Medical Oncology Department, Barretos Cancer Hospital, Barretos, Brazil
| | - Céline Pinheiro
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, Brazil.,Barretos School of Health Sciences Dr. Paulo Prata-FACISB, Barretos, Brazil
| |
Collapse
|
43
|
Würnschimmel C, Wenzel M, Maurer T, Valdés Olmos RA, Vidal-Sicart S. Contemporary update of SPECT tracers and novelties in radioguided surgery: a perspective based on urology. THE QUARTERLY JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING : OFFICIAL PUBLICATION OF THE ITALIAN ASSOCIATION OF NUCLEAR MEDICINE (AIMN) [AND] THE INTERNATIONAL ASSOCIATION OF RADIOPHARMACOLOGY (IAR), [AND] SECTION OF THE SOCIETY OF RADIOPHARMACEUTICAL CHEMISTRY AND BIOLOGY 2021; 65:215-228. [PMID: 33829716 DOI: 10.23736/s1824-4785.21.03345-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Recent technical advances and implementation of novel radiotracers have further increased the potential of radioguided surgery for a broad variety of malignancies. Indeed, the possibilities for future applications of novel radiotracers in diverse oncological strategies has become more promising than ever. This literature review aims to provide a contemporary update on a selected group of radiotracers and evaluates the usability of radioguided surgery and sentinel node procedures, focusing on most promising advances. For example, the impact of targeted radiotracers on prostate specific membrane antigen (PSMA), CD206 receptor-targeted agents (99mTc-tilmanocept), and hybrid tracers adding fluorescence to radioguidance (ICG-99mTc-nanocolloid) as well as targeting hypoxia-induced carbonic anhydrase IX (CAIX) will be covered. Furthermore, future outlooks on the implementation of gold nanoparticles (AuNP's), but also technical advances in improved radiotracer detection by hybrid gamma devices will be discussed.
Collapse
Affiliation(s)
- Christoph Würnschimmel
- Martini-Klinik Prostate Cancer Center, University Hospital of Hamburg-Eppendorf, Hamburg, Germany -
| | - Mike Wenzel
- Department of Urology, University Hospital of Frankfurt, Frankfurt, Germany
| | - Tobias Maurer
- Martini-Klinik Prostate Cancer Center, University Hospital of Hamburg-Eppendorf, Hamburg, Germany.,Department of Urology, University Hospital of Hamburg-Eppendorf, Hamburg, Germany
| | - Renato A Valdés Olmos
- Section of Nuclear Medicine, Department of Radiology, Interventional Molecular Imaging Laboratory, Leiden University Medical Center, Leiden, the Netherlands
| | - Sergi Vidal-Sicart
- Department of Nuclear Medicine, Clinic of Barcelona, Barcelona, Spain.,Institut d'Investigació Biomèdica August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| |
Collapse
|
44
|
Thiruthaneeswaran N, Bibby BAS, Yang L, Hoskin PJ, Bristow RG, Choudhury A, West C. Lost in application: Measuring hypoxia for radiotherapy optimisation. Eur J Cancer 2021; 148:260-276. [PMID: 33756422 DOI: 10.1016/j.ejca.2021.01.039] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 01/21/2021] [Accepted: 01/28/2021] [Indexed: 12/15/2022]
Abstract
The history of radiotherapy is intertwined with research on hypoxia. There is level 1a evidence that giving hypoxia-targeting treatments with radiotherapy improves locoregional control and survival without compromising late side-effects. Despite coming in and out of vogue over decades, there is now an established role for hypoxia in driving molecular alterations promoting tumour progression and metastases. While tumour genomic complexity and immune profiling offer promise, there is a stronger evidence base for personalising radiotherapy based on hypoxia status. Despite this, there is only one phase III trial targeting hypoxia modification with full transcriptomic data available. There are no biomarkers in routine use for patients undergoing radiotherapy to aid management decisions, and a roadmap is needed to ensure consistency and provide a benchmark for progression to application. Gene expression signatures address past limitations of hypoxia biomarkers and could progress biologically optimised radiotherapy. Here, we review recent developments in generating hypoxia gene expression signatures and highlight progress addressing the challenges that must be overcome to pave the way for their clinical application.
Collapse
Affiliation(s)
- Niluja Thiruthaneeswaran
- Division of Cancer Sciences, The University of Manchester, Manchester, UK; Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia.
| | - Becky A S Bibby
- Division of Cancer Sciences, The University of Manchester, Manchester, UK
| | - Lingjang Yang
- Division of Cancer Sciences, The University of Manchester, Manchester, UK
| | - Peter J Hoskin
- Division of Cancer Sciences, The University of Manchester, Manchester, UK; Mount Vernon Cancer Centre, Northwood, UK
| | - Robert G Bristow
- Division of Cancer Sciences, The University of Manchester, Manchester, UK; CRUK Manchester Institute and Manchester Cancer Research Centre, Manchester, UK
| | - Ananya Choudhury
- Division of Cancer Sciences, The University of Manchester, Christie Hospital NHS Foundation Trust, Manchester, UK
| | - Catharine West
- Division of Cancer Sciences, The University of Manchester, Christie Hospital NHS Foundation Trust, Manchester, UK
| |
Collapse
|
45
|
Kast RE. Adding high-dose celecoxib to increase effectiveness of standard glioblastoma chemoirradiation. ANNALES PHARMACEUTIQUES FRANÇAISES 2021; 79:481-488. [PMID: 33689795 DOI: 10.1016/j.pharma.2021.03.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 02/25/2021] [Accepted: 03/01/2021] [Indexed: 11/28/2022]
Abstract
Over one hundred clinical trials since 2005 have failed to significantly improve the prognosis of glioblastoma. Since 2005, the standard of care has been maximal resection followed by 60Gy irradiation over six weeks with daily temozolomide. With this, a median survival of 2 years can be expected. This short paper reviewed how the pharmacodynamic attributes of an EMA/FDA approved, cheap, generic drug to treat pain, celecoxib, intersect with pathophysiological elements driving glioblastoma growth, such that growth drive inhibition can be expected from celecoxib. The two main attributes of celecoxib are carbonic anhydrase inhibition and cyclooxygenase-2 inhibition. Both attributes individually have been in active study as adjuncts during current cancer treatment, including that of glioblastoma. That research is briefly reviewed here. This paper concludes from the collected data, that starting celecoxib, 600 to 800mg twice daily before surgery and continuing it through the chemoirradiation phase of treatment would be a low-risk intervention with sound rationale.
Collapse
Affiliation(s)
- R E Kast
- IIAIGC study centre, 05401 Burlington, VT, USA.
| |
Collapse
|
46
|
Nakashima K, Iikuni S, Okada Y, Watanabe H, Shimizu Y, Nakamoto Y, Ono M. Synthesis and evaluation of 68Ga-labeled imidazothiadiazole sulfonamide derivatives for PET imaging of carbonic anhydrase-IX. Nucl Med Biol 2021; 93:46-53. [PMID: 33316738 DOI: 10.1016/j.nucmedbio.2020.11.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 10/18/2020] [Accepted: 11/22/2020] [Indexed: 01/26/2023]
Abstract
INTRODUCTION Carbonic anhydrase-IX (CA-IX) is markedly overexpressed in many types of solid tumors promoting tumorigenicity and tumor growth. We synthesized novel 68Ga-labeled imidazothiadiazole sulfonamide (IS) derivatives ([68Ga]Ga-DO3A-IS1 and [68Ga]Ga-DO2A-IS2), and evaluated their utility as positron emission tomography (PET) probes targeting CA-IX. METHODS [67/68Ga]Ga-DO3A-IS1 and [67/68Ga]Ga-DO2A-IS2 were synthesized from corresponding precursors by ligand substitution reaction in acetate buffer. Cell binding assays were performed using HT-29 cells, which highly express CA-IX, and MDA-MB-231 cells, which show lower-level expression of CA-IX, and a biodistribution assay with model mice bearing the HT-29 or MDA-MB-231 tumor was performed. [68Ga]Ga-DO3A-IS1 was further evaluated by PET/CT. RESULTS To evaluate their fundamental properties, [67Ga]Ga-DO3A-IS1 and [67Ga]Ga-DO2A-IS2 were synthesized by conjugation with 67Ga, which has a much longer decay half-life and can be utilized more easily than 68Ga. [67/68Ga]Ga-DO3A-IS1 and [67/68Ga]Ga-DO2A-IS2 were prepared from corresponding precursors with preferable yield and purity. [67Ga]Ga-DO3A-IS1 and [67Ga]Ga-DO2A-IS2 showed significantly greater binding to HT-29 cells than MDA-MB-231 cells in vitro and the binding of [67Ga]Ga-DO2A-IS2 to HT-29 cells was much greater than that of [67Ga]Ga-DO3A-IS1, suggesting multivalent interactions. [67Ga]Ga-DO3A-IS1 and [67Ga]Ga-DO2A-IS2 showed significant selectivity for the HT-29 tumor in vivo, while tumor uptake of [67Ga]Ga-DO3A-IS1 was greater than that of [67Ga]Ga-DO2A-IS2. PET/CT of [68Ga]Ga-DO3A-IS1 showed selectivity for the HT-29 tumor, although [68Ga]Ga-DO3A-IS1 could not be used to visualize the HT-29 tumor clearly because of its strong background signals. CONCLUSION These results indicate that 68Ga-labeled IS derivatives may be useful 68Ga-PET probes targeting CA-IX with further structural modifications.
Collapse
Affiliation(s)
- Kazuma Nakashima
- Department of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Shimpei Iikuni
- Department of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Yuya Okada
- Department of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Hiroyuki Watanabe
- Department of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Yoichi Shimizu
- Department of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan; Department of Diagnostic Imaging and Nuclear Medicine, Graduate School of Medicine, Kyoto University, 54 Shogoinkawara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Yuji Nakamoto
- Department of Diagnostic Imaging and Nuclear Medicine, Graduate School of Medicine, Kyoto University, 54 Shogoinkawara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Masahiro Ono
- Department of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan.
| |
Collapse
|
47
|
Ruzzolini J, Laurenzana A, Andreucci E, Peppicelli S, Bianchini F, Carta F, Supuran CT, Romanelli MN, Nediani C, Calorini L. A potentiated cooperation of carbonic anhydrase IX and histone deacetylase inhibitors against cancer. J Enzyme Inhib Med Chem 2020; 35:391-397. [PMID: 31865754 PMCID: PMC6968260 DOI: 10.1080/14756366.2019.1706090] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 12/10/2019] [Accepted: 12/12/2019] [Indexed: 12/19/2022] Open
Abstract
The emergence of tumour recurrence and resistance limits the survival rate for most tumour-bearing patients. Only, combination therapies targeting pathways involved in the induction and in the maintenance of cancer growth and progression might potentially result in an enhanced therapeutic efficacy. Herein, we provided a prospective combination treatment that includes suberoylanilide hydroxamic acid (SAHA), a well-known inhibitor of histone deacetylases (HDACs), and SLC-0111, a novel inhibitor of carbonic anhydrase (CA) IX. We proved that HDAC inhibition with SAHA in combination with SLC-0111 affects cell viability and colony forming capability to greater extent than either treatment alone of breast, colorectal and melanoma cancer cells. At the molecular level, this therapeutic regimen resulted in a synergistically increase of histone H4 and p53 acetylation in all tested cell lines. Overall, our findings showed that SAHA and SLC-0111 can be regarded as very attractive combination providing a potential therapeutic strategy against different cancer models.
Collapse
Affiliation(s)
- Jessica Ruzzolini
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, Florence, Italy
| | - Anna Laurenzana
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, Florence, Italy
| | - Elena Andreucci
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, Florence, Italy
| | - Silvia Peppicelli
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, Florence, Italy
| | - Francesca Bianchini
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, Florence, Italy
| | - Fabrizio Carta
- Department of NEUROFARBA, University of Florence, Florence, Italy
| | | | | | - Chiara Nediani
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, Florence, Italy
| | - Lido Calorini
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, Florence, Italy
- Center of Excellence for Research, Transfer and High Education, DenoTHE University of Florence, Florence, Italy
| |
Collapse
|
48
|
Influence of paclitaxel therapy on expression of ßIII-Tubulin and Carbonic anhydrase IX proteins in chemically-induced rat mammary tumors. Biologia (Bratisl) 2020. [DOI: 10.2478/s11756-020-00496-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
49
|
Abstract
Imaging has played a critical role in the management of patients with cancer. Novel therapies are emerging rapidly; however, they are effective only in some patients. With the advent of new targeted therapeutics and immunotherapy, the limitations of conventional imaging methods are becoming more evident. FDG-PET imaging is restricted to the optimal assessment of immune therapies. There is a critical unmet need for pharmacodynamic and prognostic imaging biomarkers. Radiolabeled antibodies or small molecules can allow for specific assessment of targets in expression and concentration. Several such imaging agents have been under preclinical development. Early human studies with radiolabeled monoclonal antibodies or small molecules targeted to the epidermal growth factor receptor pathway have shown potential; targeted imaging of CA19.9 and CA-IX and are being further explored. Immune-directed imaging agents are highly desirable as biomarkers and preliminary studies with radiolabeled antibodies targeting immune mechanisms appear promising. While novel agents are being developed, larger well-designed studies are needed to validate the role of these agents as biomarkers in the clinical management of patients.
Collapse
Affiliation(s)
- Neeta Pandit-Taskar
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY; Weill Cornell Medical College, New York, NY.
| | - Michael A Postow
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| |
Collapse
|
50
|
Sapio L, Salzillo A, Ragone A, Illiano M, Spina A, Naviglio S. Targeting CREB in Cancer Therapy: A Key Candidate or One of Many? An Update. Cancers (Basel) 2020; 12:3166. [PMID: 33126560 PMCID: PMC7693618 DOI: 10.3390/cancers12113166] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 10/23/2020] [Accepted: 10/27/2020] [Indexed: 12/11/2022] Open
Abstract
Intratumor heterogeneity (ITH) is considered the major disorienting factor in cancer treatment. As a result of stochastic genetic and epigenetic alterations, the appearance of a branched evolutionary shape confers tumor plasticity, causing relapse and unfavorable clinical prognosis. The growing evidence in cancer discovery presents to us "the great paradox" consisting of countless potential targets constantly discovered and a small number of candidates being effective in human patients. Among these, cyclic-AMP response element-binding protein (CREB) has been proposed as proto-oncogene supporting tumor initiation, progression and metastasis. Overexpression and hyperactivation of CREB are frequently observed in cancer, whereas genetic and pharmacological CREB downregulation affects proliferation and apoptosis. Notably, the present review is designed to investigate the feasibility of targeting CREB in cancer therapy. In particular, starting with the latest CREB evidence in cancer pathophysiology, we evaluate the advancement state of CREB inhibitor design, including the histone lysine demethylases JMJD3/UTX inhibitor GSKJ4 that we newly identified as a promising CREB modulator in leukemia cells. Moreover, an accurate analysis of strengths and weaknesses is also conducted to figure out whether CREB can actually represent a therapeutic candidate or just one of the innumerable preclinical cancer targets.
Collapse
Affiliation(s)
| | | | | | | | | | - Silvio Naviglio
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, Via L. De Crecchio 7, 80138 Naples, Italy; (L.S.); (A.S.); (A.R.); (M.I.); (A.S.)
| |
Collapse
|