1
|
Napolitano R, Adamo A, Biondi L, Cordaro A, Iaboni M, Stummo F, La Cava F, Arena F, Faletto D, Chianale F, Reitano E, Poggi L, Blasi F. Novel Near Infrared Dyes Targeting Carbonic Anhydrase IX for Fluorescence Imaging Applications. Invest Radiol 2025:00004424-990000000-00335. [PMID: 40334098 DOI: 10.1097/rli.0000000000001204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/09/2025]
Abstract
OBJECTIVES Fluorescence tumor-targeted imaging is a sensitive technique that may assist surgeons to remove residual cancer tissue during resection. Carbonic anhydrase IX (CAIX) is a tumor-associated cell-surface glycoprotein, upregulated in hypoxic environments, and a suitable biomarker to develop targeted dyes for fluorescence-guided surgery. This study describes design, synthesis, in vitro and in vivo assessment of novel CAIX-targeting fluorescent probes based on the well-known drug acetazolamide, addressing the contribution of both targeting moiety and fluorophore structure on imaging efficacy. MATERIALS AND METHODS All the CAIX-targeting heptamethine cyanines synthesized and described in the present work were characterized in terms of their optical properties in different media. The affinity to human serum albumin was evaluated by UV-VIS spectrophotometry. The affinity to the CA catalytic site was determined on a recombinant bovine CAII enzyme (bCAII), with a fluorescent-based assay. Human colon adenocarcinoma HT-29 cells, highly expressing CAIX, were used for the in vitro characterization, including cell binding, uptake and competition assays, by flow cytometry. Finally, the in vivo tumor targeting efficacy of a selected group of probes was assessed by Optical Imaging in a mouse subcutaneous tumor from HT-29 cells, characterized by both expression of CAIX and a hypoxic tumor microenvironment. RESULTS First, a family of CAIX-targeting probes was prepared by functionalizing a novel glucamine-bearing heptamethine cyanine (Dye1) with a modified acetazolamide moiety, whose acetyl group was replaced with i) aminooctanoic acid C8, ii) phenylalanine, iii) amino-PEG2-acid and iv) the longer linker 4a, or 2 commercially available benzenesulfonamides. From the in vitro screening of this first group of compounds, the C8-AZA targeting moiety was selected due to its highest affinity. Indeed, Dye1-C8-AZA exhibited the lowest KD values for both bCAII (6.1 ± 1.6 nM) and CAIX-expressing HT-29 cells (58 ± 9 nM), even lower than HypoxyFluor-1 (HF-1), a CAIX-targeted dye already reported in the literature. Then, other heptamethine cyanines (Dye2-Dye5, linear or cyclic, with different substituents on the indolenines and different conjugation position of the targeting vector) were functionalized with C8-AZA and fully characterized both in vitro and in vivo, to evaluate the combinatory effect of vector and fluorophore on the performance of the resulting probes. The different chemical features of the cyanines influenced the optical properties, solubility, binding with albumin, biodistribution, and imaging efficacy of the probes, while leaving unaffected the high affinity to the target. When tested in vivo for the visualization of CAIX-expressing HT-29 tumors, all C8-AZA probes showed high and specific tumor accumulation, often superior to HF-1. CONCLUSIONS Several CAIX-targeting probes were synthesized to test the combinatory effect of different molecular vectors and dyes on the biological properties. All probes containing the C8-AZA targeting moiety displayed higher affinity and specificity to the target, while imaging efficacy in vivo was strongly influenced also by the structure of the labelling dye. All probes, and among them especially Dye1-C8-AZA, displayed efficient in vivo tumor accumulation. These results support further studies toward clinical testing of CAIX as suitable target for tumor fluorescence imaging and pave the way for future clinical applications.
Collapse
|
2
|
Annušová A, Labudová M, Truchan D, Hegedűšová V, Švajdlenková H, Mičušík M, Kotlár M, Pribusová Slušná L, Hulman M, Salehtash F, Kálosi A, Csáderová L, Švastová E, Šiffalovič P, Jergel M, Pastoreková S, Majková E. Selective Tumor Hypoxia Targeting Using M75 Antibody Conjugated Photothermally Active MoO x Nanoparticles. ACS OMEGA 2023; 8:44497-44513. [PMID: 38046334 PMCID: PMC10688043 DOI: 10.1021/acsomega.3c01934] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 07/24/2023] [Accepted: 11/02/2023] [Indexed: 12/05/2023]
Abstract
Photothermal therapy (PTT) mediated at the nanoscale has a unique advantage over currently used cancer treatments, by being spatially highly specific and minimally invasive. Although PTT combats traditional tumor treatment approaches, its clinical implementation has not yet been successful. The reasons for its disadvantage include an insufficient treatment efficiency or low tumor accumulation. Here, we present a promising new PTT platform combining a recently emerged two-dimensional (2D) inorganic nanomaterial, MoOx, and a tumor hypoxia targeting element, the monoclonal antibody M75. M75 specifically binds to carbonic anhydrase IX (CAIX), a hypoxia marker associated with many solid tumors with a poor prognosis. The as-prepared nanoconjugates showed highly specific binding to cancer cells expressing CAIX while being able to produce significant photothermal yield after irradiation with near-IR wavelengths. Small aminophosphonic acid linkers were recognized to be more effective over the combination of poly(ethylene glycol) chain and biotin-avidin-biotin bridge in constructing a PTT platform with high tumor-binding efficacy. The in vitro cellular uptake of nanoconjugates was visualized by high-resolution fluorescence microscopy and label-free live cell confocal Raman microscopy. The key to effective cancer treatment may be the synergistic employment of active targeting and noninvasive, tumor-selective therapeutic approaches, such as nanoscale-mediated PTT. The use of active targeting can streamline nanoparticle delivery increasing photothermal yield and therapeutic success.
Collapse
Affiliation(s)
- Adriana Annušová
- Institute
of Physics, Slovak Academy of Sciences, Dúbravská cesta 9, 845 11 Bratislava, Slovakia
- Centre
for Advanced Materials Application, Slovak
Academy of Sciences, Dúbravská cesta 9, 845
11 Bratislava, Slovakia
| | - Martina Labudová
- Centre
for Advanced Materials Application, Slovak
Academy of Sciences, Dúbravská cesta 9, 845
11 Bratislava, Slovakia
- Institute
of Virology, Biomedical Research Center,
Slovak Academy of Sciences, Dúbravská cesta 9, 845 05 Bratislava, Slovakia
- Faculty
of Natural Sciences, Comenius University
in Bratislava, Ilkovičova
6, 842 15 Bratislava, Slovakia
| | - Daniel Truchan
- Institute
of Physics, Slovak Academy of Sciences, Dúbravská cesta 9, 845 11 Bratislava, Slovakia
- Université
Sorbonne Paris Nord, Université Paris
Cité, Laboratory for Vascular Translational Science, LVTS,
INSERM, UMR 1148, Bobigny F-93017, France
| | - Veronika Hegedűšová
- Institute
of Physics, Slovak Academy of Sciences, Dúbravská cesta 9, 845 11 Bratislava, Slovakia
- Faculty
of Natural Sciences, Comenius University
in Bratislava, Ilkovičova
6, 842 15 Bratislava, Slovakia
| | - Helena Švajdlenková
- Faculty
of Natural Sciences, Comenius University
in Bratislava, Ilkovičova
6, 842 15 Bratislava, Slovakia
- Polymer
Institute, Slovak Academy of Sciences, Dúbravská cesta 9, 845 41 Bratislava, Slovakia
| | - Matej Mičušík
- Polymer
Institute, Slovak Academy of Sciences, Dúbravská cesta 9, 845 41 Bratislava, Slovakia
| | - Mário Kotlár
- Centre
for Nanodiagnostics of Materials, Slovak
University of Technology in Bratislava, Vazovova 5, 812 43 Bratislava, Slovakia
| | - Lenka Pribusová Slušná
- Centre
for Advanced Materials Application, Slovak
Academy of Sciences, Dúbravská cesta 9, 845
11 Bratislava, Slovakia
- Institute
of Electrical Engineering, Slovak Academy of Sciences, Dúbravská cesta 9, 841 04 Bratislava, Slovakia
| | - Martin Hulman
- Institute
of Electrical Engineering, Slovak Academy of Sciences, Dúbravská cesta 9, 841 04 Bratislava, Slovakia
| | - Farnoush Salehtash
- Institute
of Physics, Slovak Academy of Sciences, Dúbravská cesta 9, 845 11 Bratislava, Slovakia
| | - Anna Kálosi
- Institute
of Physics, Slovak Academy of Sciences, Dúbravská cesta 9, 845 11 Bratislava, Slovakia
- Centre
for Advanced Materials Application, Slovak
Academy of Sciences, Dúbravská cesta 9, 845
11 Bratislava, Slovakia
| | - Lucia Csáderová
- Centre
for Advanced Materials Application, Slovak
Academy of Sciences, Dúbravská cesta 9, 845
11 Bratislava, Slovakia
- Institute
of Virology, Biomedical Research Center,
Slovak Academy of Sciences, Dúbravská cesta 9, 845 05 Bratislava, Slovakia
| | - Eliška Švastová
- Centre
for Advanced Materials Application, Slovak
Academy of Sciences, Dúbravská cesta 9, 845
11 Bratislava, Slovakia
- Institute
of Virology, Biomedical Research Center,
Slovak Academy of Sciences, Dúbravská cesta 9, 845 05 Bratislava, Slovakia
| | - Peter Šiffalovič
- Institute
of Physics, Slovak Academy of Sciences, Dúbravská cesta 9, 845 11 Bratislava, Slovakia
- Centre
for Advanced Materials Application, Slovak
Academy of Sciences, Dúbravská cesta 9, 845
11 Bratislava, Slovakia
| | - Matej Jergel
- Institute
of Physics, Slovak Academy of Sciences, Dúbravská cesta 9, 845 11 Bratislava, Slovakia
- Centre
for Advanced Materials Application, Slovak
Academy of Sciences, Dúbravská cesta 9, 845
11 Bratislava, Slovakia
| | - Silvia Pastoreková
- Institute
of Virology, Biomedical Research Center,
Slovak Academy of Sciences, Dúbravská cesta 9, 845 05 Bratislava, Slovakia
| | - Eva Majková
- Institute
of Physics, Slovak Academy of Sciences, Dúbravská cesta 9, 845 11 Bratislava, Slovakia
- Centre
for Advanced Materials Application, Slovak
Academy of Sciences, Dúbravská cesta 9, 845
11 Bratislava, Slovakia
| |
Collapse
|
3
|
Cancer Therapeutic Targeting of Hypoxia Induced Carbonic Anhydrase IX: From Bench to Bedside. Cancers (Basel) 2022; 14:cancers14143297. [PMID: 35884358 PMCID: PMC9322110 DOI: 10.3390/cancers14143297] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 07/04/2022] [Indexed: 02/01/2023] Open
Abstract
Simple Summary Tumor hypoxia remains a significant problem in the effective treatment of most cancers. Tumor cells within hypoxic niches tend to be largely resistant to most therapeutic modalities, and adaptation of the cells within the hypoxic microenvironment imparts the cells with aggressive, invasive behavior. Thus, a major goal of successful cancer therapy should be the eradication of hypoxic tumor cells. Carbonic Anhydrase IX (CAIX) is an exquisitely hypoxia induced protein, selectively expressed on hypoxic tumor cells, and thus has garnered significant attention as a therapeutic target. In this Commentary, we discuss the current status of targeting CAIX, and future strategies for effective, durable cancer treatment. Abstract Carbonic Anhydrase IX (CAIX) is a major metabolic effector of tumor hypoxia and regulates intra- and extracellular pH and acidosis. Significant advances have been made recently in the development of therapeutic targeting of CAIX. These approaches include antibody-based immunotherapy, as well as use of antibodies to deliver toxic and radioactive payloads. In addition, a large number of small molecule inhibitors which inhibit the enzymatic activity of CAIX have been described. In this commentary, we highlight the current status of strategies targeting CAIX in both the pre-clinical and clinical space, and discuss future perspectives that leverage inhibition of CAIX in combination with additional targeted therapies to enable effective, durable approaches for cancer therapy.
Collapse
|
4
|
Pérez de la Lastra JM, Baca-González V, González-Acosta S, Asensio-Calavia P, Otazo-Pérez A, Morales-delaNuez A. Antibodies targeting enzyme inhibition as potential tools for research and drug development. Biomol Concepts 2021; 12:215-232. [PMID: 35104929 DOI: 10.1515/bmc-2021-0021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 12/31/2021] [Indexed: 12/29/2022] Open
Abstract
Antibodies have transformed biomedical research and are now being used for different experimental applications. Generally, the interaction of enzymes with their specific antibodies can lead to a reduction in their enzymatic activity. The effect of the antibody is dependent on its narrow i.e. the regions of the enzyme to which it is directed. The mechanism of this inhibition is rarely a direct combination of the antibodies with the catalytic site, but is rather due to steric hindrance, barring the substrate access to the active site. In several systems, however, the interaction with the antibody induces conformational changes on the enzyme that can either inhibit or enhance its catalytic activity. The extent of enzyme inhibition or enhancement is, therefore, a reflection of the nature and distribution of the various antigenic determinants on the enzyme molecule. Currently, the mode of action of many enzymes has been elucidated at the molecular level. We here review the molecular mechanisms and recent trends by which antibodies inhibit the catalytic activity of enzymes and provide examples of how specific antibodies can be useful for the neutralization of biologically active molecules.
Collapse
Affiliation(s)
- José Manuel Pérez de la Lastra
- Biotechnology of macromolecules. Instituto de Productos Naturales y Agrobiología (IPNA-CSIC), San Cristóbal de la Laguna, Tenerife, Spain
| | - Victoria Baca-González
- Biotechnology of macromolecules. Instituto de Productos Naturales y Agrobiología (IPNA-CSIC), San Cristóbal de la Laguna, Tenerife, Spain.,Escuela Doctorado y Estudios de Posgrado. Universidad de La Laguna (ULL). C/ Pedro Zerolo, s/n. 38200. San Cristóbal de La Laguna. S/C de Tenerife, Spain
| | - Sergio González-Acosta
- Biotechnology of macromolecules. Instituto de Productos Naturales y Agrobiología (IPNA-CSIC), San Cristóbal de la Laguna, Tenerife, Spain
| | - Patricia Asensio-Calavia
- Biotechnology of macromolecules. Instituto de Productos Naturales y Agrobiología (IPNA-CSIC), San Cristóbal de la Laguna, Tenerife, Spain.,Escuela Doctorado y Estudios de Posgrado. Universidad de La Laguna (ULL). C/ Pedro Zerolo, s/n. 38200. San Cristóbal de La Laguna. S/C de Tenerife, Spain
| | - Andrea Otazo-Pérez
- Biotechnology of macromolecules. Instituto de Productos Naturales y Agrobiología (IPNA-CSIC), San Cristóbal de la Laguna, Tenerife, Spain.,Escuela Doctorado y Estudios de Posgrado. Universidad de La Laguna (ULL). C/ Pedro Zerolo, s/n. 38200. San Cristóbal de La Laguna. S/C de Tenerife, Spain
| | - Antonio Morales-delaNuez
- Biotechnology of macromolecules. Instituto de Productos Naturales y Agrobiología (IPNA-CSIC), San Cristóbal de la Laguna, Tenerife, Spain
| |
Collapse
|
5
|
Sheff JG, Kelly JF, Robotham A, Sulea T, Malenfant F, L'Abbé D, Duchesne M, Pelletier A, Lefebvre J, Acel A, Parat M, Gosselin M, Wu C, Fortin Y, Baardsnes J, Van Faassen H, Awrey S, Chafe SC, McDonald PC, Dedhar S, Lenferink AEG. Hydrogen-deuterium exchange mass spectrometry reveals three unique binding responses of mAbs directed to the catalytic domain of hCAIX. MAbs 2021; 13:1997072. [PMID: 34812124 PMCID: PMC8632303 DOI: 10.1080/19420862.2021.1997072] [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] [Indexed: 12/21/2022] Open
Abstract
Human carbonic anhydrase (hCAIX), an extracellular enzyme that catalyzes the reversible hydration of CO2, is often overexpressed in solid tumors. This enzyme is instrumental in maintaining the survival of cancer cells in a hypoxic and acidic tumor microenvironment. Absent in most normal tissues, hCAIX is a promising therapeutic target for detection and treatment of solid tumors. Screening of a library of anti-hCAIX monoclonal antibodies (mAbs) previously identified three therapeutic candidates (mAb c2C7, m4A2 and m9B6) with distinct biophysical and functional characteristics. Selective binding to the catalytic domain was confirmed by yeast surface display and isothermal calorimetry, and deeper insight into the dynamic binding profiles of these mAbs upon binding were highlighted by bottom-up hydrogen-deuterium exchange mass spectrometry (HDX-MS). Here, a conformational and allosterically silent epitope was identified for the antibody-drug conjugate candidate c2C7. Unique binding profiles are described for both inhibitory antibodies, m4A2 and m9B6. M4A2 reduces the ability of the enzyme to hydrate CO2 by steric gating at the entrance of the catalytic cavity. Conversely, m9B6 disrupts the secondary structure that is necessary for substrate binding and hydration. The synergy of these two inhibitory mechanisms is demonstrated in in vitro activity assays and HDX-MS. Finally, the ability of m4A2 to modulate extracellular pH and intracellular metabolism is reported. By highlighting three unique modes by which hCAIX can be targeted, this study demonstrates both the utility of HDX-MS as an important tool in the characterization of anti-cancer biotherapeutics, and the underlying value of CAIX as a therapeutic target.
Collapse
Affiliation(s)
- Joey G Sheff
- Human Health Therapeutics Research Centre, National Research Council Canada, Ottawa, Ontario, Canada
| | - John F Kelly
- Human Health Therapeutics Research Centre, National Research Council Canada, Ottawa, Ontario, Canada
| | - Anna Robotham
- Human Health Therapeutics Research Centre, National Research Council Canada, Ottawa, Ontario, Canada
| | - Traian Sulea
- Human Health Therapeutics Research Centre, National Research Council Canada, Montreal, Quebec, Canada
| | - Félix Malenfant
- Human Health Therapeutics Research Centre, National Research Council Canada, Montreal, Quebec, Canada
| | - Denis L'Abbé
- Human Health Therapeutics Research Centre, National Research Council Canada, Montreal, Quebec, Canada
| | - Mélanie Duchesne
- Human Health Therapeutics Research Centre, National Research Council Canada, Montreal, Quebec, Canada
| | - Alex Pelletier
- Human Health Therapeutics Research Centre, National Research Council Canada, Montreal, Quebec, Canada
| | - Jean Lefebvre
- Human Health Therapeutics Research Centre, National Research Council Canada, Montreal, Quebec, Canada
| | - Andrea Acel
- Human Health Therapeutics Research Centre, National Research Council Canada, Montreal, Quebec, Canada
| | - Marie Parat
- Human Health Therapeutics Research Centre, National Research Council Canada, Montreal, Quebec, Canada
| | - Mylene Gosselin
- Human Health Therapeutics Research Centre, National Research Council Canada, Montreal, Quebec, Canada
| | - Cunle Wu
- Human Health Therapeutics Research Centre, National Research Council Canada, Montreal, Quebec, Canada
| | - Yves Fortin
- Human Health Therapeutics Research Centre, National Research Council Canada, Montreal, Quebec, Canada
| | - Jason Baardsnes
- Human Health Therapeutics Research Centre, National Research Council Canada, Montreal, Quebec, Canada
| | - Henk Van Faassen
- Human Health Therapeutics Research Centre, National Research Council Canada, Ottawa, Ontario, Canada
| | - Shannon Awrey
- Department of Integrative Oncology, Bc Cancer Research Institute, Vancouver, BC, Canada
| | - Shawn C Chafe
- Department of Integrative Oncology, Bc Cancer Research Institute, Vancouver, BC, Canada
| | - Paul C McDonald
- Department of Integrative Oncology, Bc Cancer Research Institute, Vancouver, BC, Canada
| | - Shoukat Dedhar
- Department of Integrative Oncology, Bc Cancer Research Institute, Vancouver, BC, Canada.,Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
| | - Anne E G Lenferink
- Human Health Therapeutics Research Centre, National Research Council Canada, Montreal, Quebec, Canada
| |
Collapse
|
6
|
Lenferink AEG, McDonald PC, Cantin C, Grothé S, Gosselin M, Baardsnes J, Banville M, Lachance P, Robert A, Cepero-Donates Y, Radinovic S, Salois P, Parat M, Oamari H, Dulude A, Patel M, Lafrance M, Acel A, Bousquet-Gagnon N, L'Abbé D, Pelletier A, Malenfant F, Jaramillo M, O'Connor-Mccourt M, Wu C, Durocher Y, Duchesne M, Gadoury C, Marcil A, Fortin Y, Paul-Roc B, Acchione M, Chafe SC, Nemirovsky O, Lau J, Bénard F, Dedhar S. Isolation and characterization of monoclonal antibodies against human carbonic anhydrase-IX. MAbs 2021; 13:1999194. [PMID: 34806527 PMCID: PMC8632296 DOI: 10.1080/19420862.2021.1999194] [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] [Indexed: 12/19/2022] Open
Abstract
The architectural complexity and heterogeneity of the tumor microenvironment (TME) remains a substantial obstacle in the successful treatment of cancer. Hypoxia, caused by insufficient oxygen supply, and acidosis, resulting from the expulsion of acidic metabolites, are prominent features of the TME. To mitigate the consequences of the hostile TME, cancer cells metabolically rewire themselves and express a series of specific transporters and enzymes instrumental to this adaptation. One of these proteins is carbonic anhydrase (CA)IX, a zinc-containing extracellular membrane bound enzyme that has been shown to play a critical role in the maintenance of a neutral intracellular pH (pHi), allowing tumor cells to survive and thrive in these harsh conditions. Although CAIX has been considered a promising cancer target, only two antibody-based therapeutics have been clinically tested so far. To fill this gap, we generated a series of novel monoclonal antibodies (mAbs) that specifically recognize the extracellular domain (ECD) of human CAIX. Here we describe the biophysical and functional properties of a set of antibodies against the CAIX ECD domain and their applicability as: 1) suitable for development as an antibody-drug-conjugate, 2) an inhibitor of CAIX enzyme activity, or 3) an imaging/detection antibody. The results presented here demonstrate the potential of these specific hCAIX mAbs for further development as novel cancer therapeutic and/or diagnostic tools.
Collapse
Affiliation(s)
- Anne E G Lenferink
- Human Health Therapeutics Research Center, National Research Council of Canada, Montréal, Canada
| | - Paul C McDonald
- Department of Integrative Oncology, Bc Cancer Research Institute, Vancouver, Canada
| | - Christiane Cantin
- Human Health Therapeutics Research Center, National Research Council of Canada, Montréal, Canada
| | - Suzanne Grothé
- Human Health Therapeutics Research Center, National Research Council of Canada, Montréal, Canada
| | - Mylene Gosselin
- Human Health Therapeutics Research Center, National Research Council of Canada, Montréal, Canada
| | - Jason Baardsnes
- Human Health Therapeutics Research Center, National Research Council of Canada, Montréal, Canada
| | - Myriam Banville
- Human Health Therapeutics Research Center, National Research Council of Canada, Montréal, Canada
| | - Paul Lachance
- Human Health Therapeutics Research Center, National Research Council of Canada, Montréal, Canada
| | - Alma Robert
- Human Health Therapeutics Research Center, National Research Council of Canada, Montréal, Canada
| | - Yuneivy Cepero-Donates
- Human Health Therapeutics Research Center, National Research Council of Canada, Montréal, Canada
| | - Stevo Radinovic
- Human Health Therapeutics Research Center, National Research Council of Canada, Montréal, Canada
| | - Patrick Salois
- Human Health Therapeutics Research Center, National Research Council of Canada, Montréal, Canada
| | - Marie Parat
- Human Health Therapeutics Research Center, National Research Council of Canada, Montréal, Canada
| | - Hafida Oamari
- Human Health Therapeutics Research Center, National Research Council of Canada, Montréal, Canada
| | - Annie Dulude
- Human Health Therapeutics Research Center, National Research Council of Canada, Montréal, Canada
| | - Mehul Patel
- Human Health Therapeutics Research Center, National Research Council of Canada, Montréal, Canada
| | - Martin Lafrance
- Human Health Therapeutics Research Center, National Research Council of Canada, Montréal, Canada
| | - Andrea Acel
- Human Health Therapeutics Research Center, National Research Council of Canada, Montréal, Canada
| | - Nathalie Bousquet-Gagnon
- Human Health Therapeutics Research Center, National Research Council of Canada, Montréal, Canada
| | - Denis L'Abbé
- Human Health Therapeutics Research Center, National Research Council of Canada, Montréal, Canada
| | - Alex Pelletier
- Human Health Therapeutics Research Center, National Research Council of Canada, Montréal, Canada
| | - Félix Malenfant
- Human Health Therapeutics Research Center, National Research Council of Canada, Montréal, Canada
| | - Maria Jaramillo
- Human Health Therapeutics Research Center, National Research Council of Canada, Montréal, Canada
| | - Maureen O'Connor-Mccourt
- Human Health Therapeutics Research Center, National Research Council of Canada, Montréal, Canada
| | - Cunle Wu
- Human Health Therapeutics Research Center, National Research Council of Canada, Montréal, Canada
| | - Yves Durocher
- Human Health Therapeutics Research Center, National Research Council of Canada, Montréal, Canada
| | - Mélanie Duchesne
- Human Health Therapeutics Research Center, National Research Council of Canada, Montréal, Canada
| | - Christine Gadoury
- Human Health Therapeutics Research Center, National Research Council of Canada, Montréal, Canada
| | - Anne Marcil
- Human Health Therapeutics Research Center, National Research Council of Canada, Montréal, Canada
| | - Yves Fortin
- Human Health Therapeutics Research Center, National Research Council of Canada, Montréal, Canada
| | - Beatrice Paul-Roc
- Human Health Therapeutics Research Center, National Research Council of Canada, Montréal, Canada
| | - Maurizio Acchione
- Human Health Therapeutics Research Center, National Research Council of Canada, Montréal, Canada
| | - Shawn C Chafe
- Department of Integrative Oncology, Bc Cancer Research Institute, Vancouver, Canada
| | - Oksana Nemirovsky
- Department of Integrative Oncology, Bc Cancer Research Institute, Vancouver, Canada
| | - Joseph Lau
- Department of Molecular Oncology, Bc Cancer Research Institute, Vancouver, Canada
| | - Francois Bénard
- Department of Molecular Oncology, Bc Cancer Research Institute, Vancouver, Canada
| | - Shoukat Dedhar
- Department of Integrative Oncology, Bc Cancer Research Institute, Vancouver, Canada
| |
Collapse
|
7
|
Stravinskiene D, Sliziene A, Baranauskiene L, Petrikaite V, Zvirbliene A. Inhibitory Monoclonal Antibodies and Their Recombinant Derivatives Targeting Surface-Exposed Carbonic Anhydrase XII on Cancer Cells. Int J Mol Sci 2020; 21:ijms21249411. [PMID: 33321910 PMCID: PMC7763246 DOI: 10.3390/ijms21249411] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Accepted: 12/08/2020] [Indexed: 01/17/2023] Open
Abstract
Monoclonal and recombinant antibodies are widely used for the diagnostics and therapy of cancer. They are generated to interact with cell surface proteins which are usually involved in the development and progression of cancer. Carbonic anhydrase XII (CA XII) contributes to the survival of tumors under hypoxic conditions thus is considered a candidate target for antibody-based therapy. In this study, we have generated a novel collection of monoclonal antibodies (MAbs) against the recombinant extracellular domain of CA XII produced in HEK-293 cells. Eighteen out of 24 MAbs were reactive with cellular CA XII on the surface of live kidney and lung cancer cells as determined by flow cytometry. One MAb 14D6 also inhibited the enzymatic activity of recombinant CA XII as measured by the stopped-flow assay. MAb 14D6 showed the migrastatic effect on human lung carcinoma A549 and renal carcinoma A498 cell lines in a ‘wound healing’ assay. It did not reduce the growth of multicellular lung and renal cancer spheroids but reduced the cell viability by the ATP Bioluminescence assay. Epitope mapping revealed the surface-exposed amino acid sequence (35-FGPDGENS-42) close to the catalytic center of CA XII recognized by the MAb 14D6. The variable regions of the heavy and light chains of MAb 14D6 were sequenced and their complementarity-determining regions were defined. The obtained variable sequences were used to generate recombinant antibodies in two formats: single-chain fragment variable (scFv) expressed in E. coli and scFv fused to human IgG1 Fc fragment (scFv-Fc) expressed in Chinese Hamster Ovary (CHO) cells. Both recombinant antibodies maintained the same specificity for CA XII as the parental MAb 14D6. The novel antibodies may represent promising tools for CA XII-related cancer research and immunotherapy.
Collapse
Affiliation(s)
- Dovile Stravinskiene
- Department of Immunology and Cell Biology, Institute of Biotechnology, Life Sciences Center, Vilnius University, Sauletekio al. 7, LT-10257 Vilnius, Lithuania; (A.S.); (A.Z.)
- Correspondence:
| | - Aiste Sliziene
- Department of Immunology and Cell Biology, Institute of Biotechnology, Life Sciences Center, Vilnius University, Sauletekio al. 7, LT-10257 Vilnius, Lithuania; (A.S.); (A.Z.)
| | - Lina Baranauskiene
- Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences Center, Vilnius University, Sauletekio al. 7, LT-10257 Vilnius, Lithuania; (L.B.); (V.P.)
| | - Vilma Petrikaite
- Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences Center, Vilnius University, Sauletekio al. 7, LT-10257 Vilnius, Lithuania; (L.B.); (V.P.)
- Laboratory of Drug Targets Histopathology, Institute of Cardiology, Lithuanian University of Health Sciences, Sukileliu pr. 13, LT-50162 Kaunas, Lithuania
| | - Aurelija Zvirbliene
- Department of Immunology and Cell Biology, Institute of Biotechnology, Life Sciences Center, Vilnius University, Sauletekio al. 7, LT-10257 Vilnius, Lithuania; (A.S.); (A.Z.)
| |
Collapse
|
8
|
Krasavin M, Kalinin S, Sharonova T, Supuran CT. Inhibitory activity against carbonic anhydrase IX and XII as a candidate selection criterion in the development of new anticancer agents. J Enzyme Inhib Med Chem 2020; 35:1555-1561. [PMID: 32746643 PMCID: PMC7470080 DOI: 10.1080/14756366.2020.1801674] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Accepted: 07/21/2020] [Indexed: 12/19/2022] Open
Abstract
Analysis of the literature data reveals that while inhibition of cancer-related carbonic anhydrase IX and XII isoforms continues to be an important enrichment factor for designing anticancer agent development libraries, exclusive reliance on the in vitro inhibition of these two recombinant isozymes in nominating candidate compounds for evaluation of their effects on cancer cells may lead not only to identifying numerous compounds devoid of the desired cellular efficacy but also to overlooking many promising candidates which may not display the best potency in biochemical inhibition assay. However, SLC-0111, now in phase Ib/II clinical trials, was developed based on the excellent agreement between the in vitro, in vivo and more recently, in-patient data.
Collapse
Affiliation(s)
- Mikhail Krasavin
- Institute of Chemistry, St. Petersburg State University, St. Petersburg, Russia
| | - Stanislav Kalinin
- Institute of Chemistry, St. Petersburg State University, St. Petersburg, Russia
| | - Tatiana Sharonova
- Institute of Chemistry, St. Petersburg State University, St. Petersburg, Russia
| | - Claudiu T. Supuran
- Neurofarba Department, Section of Pharmaceutical Sciences, University of Florence, Florence, Italy
| |
Collapse
|
9
|
Abstract
Immuno-positron emission tomography (immunoPET) is a paradigm-shifting molecular imaging modality combining the superior targeting specificity of monoclonal antibody (mAb) and the inherent sensitivity of PET technique. A variety of radionuclides and mAbs have been exploited to develop immunoPET probes, which has been driven by the development and optimization of radiochemistry and conjugation strategies. In addition, tumor-targeting vectors with a short circulation time (e.g., Nanobody) or with an enhanced binding affinity (e.g., bispecific antibody) are being used to design novel immunoPET probes. Accordingly, several immunoPET probes, such as 89Zr-Df-pertuzumab and 89Zr-atezolizumab, have been successfully translated for clinical use. By noninvasively and dynamically revealing the expression of heterogeneous tumor antigens, immunoPET imaging is gradually changing the theranostic landscape of several types of malignancies. ImmunoPET is the method of choice for imaging specific tumor markers, immune cells, immune checkpoints, and inflammatory processes. Furthermore, the integration of immunoPET imaging in antibody drug development is of substantial significance because it provides pivotal information regarding antibody targeting abilities and distribution profiles. Herein, we present the latest immunoPET imaging strategies and their preclinical and clinical applications. We also emphasize current conjugation strategies that can be leveraged to develop next-generation immunoPET probes. Lastly, we discuss practical considerations to tune the development and translation of immunoPET imaging strategies.
Collapse
Affiliation(s)
- Weijun Wei
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, 1111 Highland Avenue, Room 7137, Madison, Wisconsin 53705, United States
| | - Zachary T Rosenkrans
- Department of Pharmaceutical Sciences, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
| | - Jianjun Liu
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Gang Huang
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai 201318, China
| | - Quan-Yong Luo
- Department of Nuclear Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Weibo Cai
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, 1111 Highland Avenue, Room 7137, Madison, Wisconsin 53705, United States
- Department of Pharmaceutical Sciences, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
- University of Wisconsin Carbone Cancer Center, Madison, Wisconsin 53705, United States
| |
Collapse
|
10
|
Ward C, Meehan J, Gray ME, Murray AF, Argyle DJ, Kunkler IH, Langdon SP. The impact of tumour pH on cancer progression: strategies for clinical intervention. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2020; 1:71-100. [PMID: 36046070 PMCID: PMC9400736 DOI: 10.37349/etat.2020.00005] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Accepted: 02/05/2020] [Indexed: 02/06/2023] Open
Abstract
Dysregulation of cellular pH is frequent in solid tumours and provides potential opportunities for therapeutic intervention. The acidic microenvironment within a tumour can promote migration, invasion and metastasis of cancer cells through a variety of mechanisms. Pathways associated with the control of intracellular pH that are under consideration for intervention include carbonic anhydrase IX, the monocarboxylate transporters (MCT, MCT1 and MCT4), the vacuolar-type H+-ATPase proton pump, and the sodium-hydrogen exchanger 1. This review will describe progress in the development of inhibitors to these targets.
Collapse
Affiliation(s)
- Carol Ward
- Cancer Research UK Edinburgh Centre and Edinburgh Pathology, Institute of Genetics and Molecular Medicine, University of Edinburgh, Crewe Road South, EH4 2XU Edinburgh, UK
| | - James Meehan
- Cancer Research UK Edinburgh Centre and Edinburgh Pathology, Institute of Genetics and Molecular Medicine, University of Edinburgh, Crewe Road South, EH4 2XU Edinburgh, UK
| | - Mark E Gray
- Royal (Dick) School of Veterinary Studies and Roslin Institute, University of Edinburgh, Easter Bush, EH25 9RG Midlothian, UK
| | - Alan F Murray
- School of Engineering, Institute for Integrated Micro and Nano Systems, EH9 3JL Edinburgh, UK
| | - David J Argyle
- Royal (Dick) School of Veterinary Studies and Roslin Institute, University of Edinburgh, Easter Bush, EH25 9RG Midlothian, UK
| | - Ian H Kunkler
- Cancer Research UK Edinburgh Centre and Edinburgh Pathology, Institute of Genetics and Molecular Medicine, University of Edinburgh, Crewe Road South, EH4 2XU Edinburgh, UK
| | - Simon P Langdon
- Cancer Research UK Edinburgh Centre and Edinburgh Pathology, Institute of Genetics and Molecular Medicine, University of Edinburgh, Crewe Road South, EH4 2XU Edinburgh, UK
| |
Collapse
|
11
|
Alterio V, Kellner M, Esposito D, Liesche-Starnecker F, Bua S, Supuran CT, Monti SM, Zeidler R, De Simone G. Biochemical and Structural Insights into Carbonic Anhydrase XII/Fab6A10 Complex. J Mol Biol 2019; 431:4910-4921. [DOI: 10.1016/j.jmb.2019.10.022] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 09/26/2019] [Accepted: 10/17/2019] [Indexed: 12/12/2022]
|
12
|
De Luca R, Gouyou B, Ongaro T, Villa A, Ziffels B, Sannino A, Buttinoni G, Galeazzi S, Mazzacuva M, Neri D. A Novel Fully-Human Potency-Matched Dual Cytokine-Antibody Fusion Protein Targets Carbonic Anhydrase IX in Renal Cell Carcinomas. Front Oncol 2019; 9:1228. [PMID: 31799191 PMCID: PMC6863974 DOI: 10.3389/fonc.2019.01228] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 10/28/2019] [Indexed: 12/12/2022] Open
Abstract
Certain cytokines synergize in activating anti-cancer immunity at the site of disease and it may be desirable to generate biopharmaceutical agents, capable of simultaneous delivery of cytokine pairs to the tumor. In this article, we have described the cloning, expression and characterization of IL2-XE114-TNFmut, a dual-cytokine biopharmaceutical featuring the sequential fusion of interleukin-2 (IL2) with the XE114 antibody in scFv format and a tumor necrosis factor mutant (TNFmut). The fusion protein recognized the cognate antigen (carbonic anhydrase IX, a marker of hypoxia and of renal cell carcinoma) with high affinity and specificity. IL2-XE114-TNFmut formed a stable non-covalent homotrimeric structure, displayed cytokine activity in in vitro tests and preferentially localized to solid tumors in vivo. The product exhibited a partial growth inhibition of murine CT26 tumors transfected for carbonic anhydrase IX. When administered to Cynomolgus monkey as intravenous injection, IL2-XE114-TNFmut showed the expected plasma concentration of ~1,500 ng/ml at early time points, indicating the absence of any in vivo trapping events, and a half-life of ~2 h. IL2-XE114-TNFmut may thus be considered as a promising biopharmaceutical for the treatment of metastatic clear-cell renal cell carcinoma, since these tumors are known to be sensitive to IL2 and to TNF.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Gianluca Buttinoni
- Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology (ETH Zürich), Zurich, Switzerland
| | | | | | - Dario Neri
- Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology (ETH Zürich), Zurich, Switzerland
| |
Collapse
|
13
|
New Monoclonal Antibodies for a Selective Detection of Membrane-Associated and Soluble Forms of Carbonic Anhydrase IX in Human Cell Lines and Biological Samples. Biomolecules 2019; 9:biom9080304. [PMID: 31349673 PMCID: PMC6723738 DOI: 10.3390/biom9080304] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 07/22/2019] [Accepted: 07/23/2019] [Indexed: 01/26/2023] Open
Abstract
Monoclonal antibodies (MAbs) selectively targeting tumor-associated antigens such as carbonic anhydrase IX (CA IX) can significantly contribute to research, diagnostics, and treatment of CA IX-related cancers. CA IX is overexpressed in numerous hypoxic cancers where it promotes tumor progression. Therefore, it is considered as a promising tumor biomarker. A novel collection of MAbs against recombinant CA IX was developed and evaluated in different immunoassays for studying CA IX expression. The reactivity of MAbs with native cell surface protein was confirmed by flow cytometry and the presence of hypoxia-inducible CA IX was investigated in several human cancer cell lines. In addition, the applicability of MAbs for visualization of CA IX-positive tumor cells by immunofluorescence microscopy was demonstrated. MAb H7 was identified as the most promising MAb for different immunoassays. It recognized a linear epitope covering CA IX sequence of 12 amino acid residues 55-GEDDPLGEEDLP-66 within the proteoglycan domain. The MAb H7 was the only one of the collection to immunoprecipitate CA IX protein from cell lysates and detect the denatured CA IX with near-infrared fluorescence Western blot. It was also employed in sandwich enzyme-linked immunosorbent assay to detect a soluble form of CA IX in growth medium of tumor cells and blood plasma samples. The diagnostic potential of the MAb H7 was confirmed on formalin-fixed and paraffin-embedded tissue specimen of cervical carcinoma in situ by immunohistochemistry. The generated MAbs, in particularly clone H7, have great potential in diagnostics and research of CA IX-related cancers.
Collapse
|
14
|
Abstract
Cancer development is a complex process that follows an intricate scenario with a dynamic interplay of selective and adaptive steps and an extensive cast of molecules and signaling pathways. Solid tumor initially grows as an avascular bulk of cells carrying oncogenic mutations until diffusion distances from the nearest functional blood vessels limit delivery of nutrients and oxygen on the one hand and removal of metabolic waste on the other one. These restrictions result in regional hypoxia and acidosis that select for adaptable tumor cells able to promote aberrant angiogenesis, remodel metabolism, acquire invasiveness and metastatic propensity, and gain therapeutic resistance. Tumor cells are thereby endowed with capability to survive and proliferate in hostile microenvironment, communicate with stroma, enter circulation, colonize secondary sites, and generate metastases. While the role of oncogenic mutations initializing and driving these processes is well established, a key contribution of non-genomic, landscaping molecular players is still less appreciated despite they can equally serve as viable targets of anticancer therapies. Carbonic anhydrase IX (CA IX) is one of these players: it is induced by hypoxia, functionally linked to acidosis, implicated in invasiveness, and correlated with therapeutic resistance. Here, we summarize the available experimental evidence supported by accumulating preclinical and clinical data that CA IX can contribute virtually to each step of cancer progression path via its enzyme activity and/or non-catalytic mechanisms. We also propose that targeting tumor cells that express CA IX may provide therapeutic benefits in various settings and combinations with both conventional and newly developed treatments.
Collapse
Affiliation(s)
- Silvia Pastorekova
- Department of Tumor Biology, Institute of Virology, Biomedical Research Center, University Science Park for Biomedicine, Slovak Academy of Sciences, Dúbravská cesta 9, 845 05, Bratislava, Slovakia.
| | - Robert J Gillies
- Department of Cancer Physiology, H. Lee Moffitt Cancer Center, 12902 Magnolia Avenue, Tampa, FL, 33612, USA
| |
Collapse
|
15
|
Nocentini A, Supuran CT. Carbonic anhydrase inhibitors as antitumor/antimetastatic agents: a patent review (2008-2018). Expert Opin Ther Pat 2018; 28:729-740. [PMID: 30074415 DOI: 10.1080/13543776.2018.1508453] [Citation(s) in RCA: 151] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Human carbonic anhydrases (CA, EC 4.2.1.1) IX and XII are tumor-associated proteins, being part of the molecular machinery that tumor cells build as adaptive responses to hypoxia and acidic conditions characteristic of the 'glycolytic shift' of many tumors. A wealth of research depicts CA IX and CA XII as biomarkers and therapeutic targets for various cancer types. AREAS COVERED The review presents an overview of the role of CA IX and CA XII in hypoxic tumors physio-pathology as well as the principal molecular, structural, and catalytic features of both isozymes. The review then covers the patent literature of medically relevant inhibitors of the tumor-associated CAs produced during the period 2008-2018. EXPERT OPINION A variety of approaches and design strategies were reported which afford CA IX/XII-specific inhibitors and avoid the compromising effects of isoforms-promiscuous compounds. Access to the crystal structures of human CAs isoforms have improved structure-based drug design campaigns related to zinc-binder chemotypes. Nevertheless, great potential still resides in non-classical CAIs that exhibit alternative binding mechanisms able to further distinguish the various active sites architecture. CA IX inhibitors hybrids/conjugates are increasingly emerging in the field as promising therapeutic tools to combine CA inhibition to the anticancer effects of other moieties or antitumor drugs.
Collapse
Affiliation(s)
- Alessio Nocentini
- a Department of NEUROFARBA, Section of Pharmaceutical and Nutraceutical Sciences , University of Florence , Sesto Fiorentino (Firenze) , Italy
| | - Claudiu T Supuran
- a Department of NEUROFARBA, Section of Pharmaceutical and Nutraceutical Sciences , University of Florence , Sesto Fiorentino (Firenze) , Italy
| |
Collapse
|
16
|
Lee SH, McIntyre D, Honess D, Hulikova A, Pacheco-Torres J, Cerdán S, Swietach P, Harris AL, Griffiths JR. Carbonic anhydrase IX is a pH-stat that sets an acidic tumour extracellular pH in vivo. Br J Cancer 2018; 119:622-630. [PMID: 30206370 PMCID: PMC6162214 DOI: 10.1038/s41416-018-0216-5] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Revised: 06/26/2018] [Accepted: 07/12/2018] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Tumour carbonic anhydrase IX (CAIX), a hypoxia-inducible tumour-associated cell surface enzyme, is thought to acidify the tumour microenvironment by hydrating CO2 to form protons and bicarbonate, but there is no definitive evidence for this in solid tumours in vivo. METHODS We used 1H magnetic resonance spectroscopic imaging (MRSI) of the extracellular pH probe imidazolyl succinic acid (ISUCA) to measure and spatially map extracellular pH in HCT116 tumours transfected to express CAIX and empty vector controls in SCID mice. We also measured intracellular pH in situ with 31P MRS and measured lactate in freeze-clamped tumours. RESULTS CAIX-expressing tumours had 0.15 pH-unit lower median extracellular pH than control tumours (pH 6.71 tumour vs pH 6.86 control, P = 0.01). Importantly, CAIX expression imposed an upper limit for tumour extracellular pH at 6.93. Despite the increased lactate concentration in CAIX-expressing tumours, 31P MRS showed no difference in intracellular pH, suggesting that CAIX acidifies only the tumour extracellular space. CONCLUSIONS CAIX acidifies the tumour microenvironment, and also provides an extracellular pH control mechanism. We propose that CAIX thus acts as an extracellular pH-stat, maintaining an acidic tumour extracellular pH that is tolerated by cancer cells and favours invasion and metastasis.
Collapse
Affiliation(s)
- Shen-Han Lee
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge, CB2 0RE, UK
- Singapore Health Services (SingHealth), Department of General Surgery, Singapore General Hospital, Academia, College Road, Singapore, 169856, Singapore
| | - Dominick McIntyre
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge, CB2 0RE, UK
| | - Davina Honess
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge, CB2 0RE, UK
| | - Alzbeta Hulikova
- Department of Physiology, Anatomy & Genetics, University of Oxford, Parks Road, Oxford, OX1 3PT, UK
| | - Jesús Pacheco-Torres
- Division of Cancer Imaging Research, The Johns Hopkins University School of Medicine, 720 Rutland Avenue, Traylor 211, Baltimore, MD, 21205, USA
| | - Sebastián Cerdán
- Instituto de Investigaciones Biomédicas "Alberto Sols", c/ Arturo Duperier 4, Madrid, 28029, Spain
| | - Pawel Swietach
- Department of Physiology, Anatomy & Genetics, University of Oxford, Parks Road, Oxford, OX1 3PT, UK
| | - Adrian L Harris
- Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DS, UK
| | - John R Griffiths
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge, CB2 0RE, UK.
| |
Collapse
|
17
|
Chen Y, Bian X, Aliru M, Deorukhkar AA, Ekpenyong O, Liang S, John J, Ma J, Gao X, Schwartz J, Singh P, Ye Y, Krishnan S, Xie H. Hypoxia-targeted gold nanorods for cancer photothermal therapy. Oncotarget 2018; 9:26556-26571. [PMID: 29899876 PMCID: PMC5995181 DOI: 10.18632/oncotarget.25492] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Accepted: 05/08/2018] [Indexed: 11/25/2022] Open
Abstract
Tumor hypoxia is a well-recognized driver of resistance to traditional cancer therapies such as chemotherapy and radiation therapy. We describe development of a new nanoconstruct composed of gold nanorods (GNRs) conjugated to carbonic anhydrase IX (CAIX) antibody that specifically binds to CAIX, a biomarker of hypoxia, to facilitate targeting tumor hypoxic areas for focused photothermal ablation. Physicochemical characterization studies confirmed the size, shape, monodispersity, surface charge, and serum stability of the GNRs. Enzyme-linked immunosorbent assays and cellular binding and uptake studies confirmed successful conjugation of antibody to the GNRs and specificity for CAIX. Near-infrared irradiation of CAIX-overexpressing cells treated with GNR/anti-CAIX resulted in significantly higher cell death than cells treated with control GNRs. In vivo biodistribution studies using hyperspectral imaging and inductively coupled plasma mass spectrometry confirmed intravenous administration results not only in greater accumulation of GNR/anti-CAIX in tumors than control GNRs but also greater penetration into hypoxic areas of tumors. Near-infrared ablation of these tumors showed no tumor regression in the sham-treated group, regression but recurrence in the non-targeted-GNR group, and complete tumor regression in the targeted-GNR group. GNR/anti-CAIX nanoconstructs show promise as hypoxia targeting and photothermal ablation agents for cancer treatment.
Collapse
Affiliation(s)
- Yuan Chen
- Department of Pharmaceutical and Environmental Sciences, College of Pharmacy and Health Sciences, Texas Southern University, Houston, TX, USA
| | - Xiaomei Bian
- Department of Pharmaceutical and Environmental Sciences, College of Pharmacy and Health Sciences, Texas Southern University, Houston, TX, USA
| | - Maureen Aliru
- Department of Radiation Oncology, Division of Radiation Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, TX, USA
| | - Amit A Deorukhkar
- Department of Radiation Oncology, Division of Radiation Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, TX, USA
| | - Oscar Ekpenyong
- Department of Pharmaceutical and Environmental Sciences, College of Pharmacy and Health Sciences, Texas Southern University, Houston, TX, USA
| | - Su Liang
- Department of Pharmaceutical and Environmental Sciences, College of Pharmacy and Health Sciences, Texas Southern University, Houston, TX, USA
| | - Jyothy John
- Department of Pharmaceutical and Environmental Sciences, College of Pharmacy and Health Sciences, Texas Southern University, Houston, TX, USA
| | - Jing Ma
- Department of Pharmaceutical and Environmental Sciences, College of Pharmacy and Health Sciences, Texas Southern University, Houston, TX, USA
| | - Xiuqing Gao
- Department of Pharmaceutical and Environmental Sciences, College of Pharmacy and Health Sciences, Texas Southern University, Houston, TX, USA
| | - Jon Schwartz
- Nanospectra Biosciences, Inc., Houston, Texas, USA
| | - Pankaj Singh
- Department of Radiation Oncology, Division of Radiation Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, TX, USA
| | - Yuanqing Ye
- Department of Epidemiology, Division of OVP, Cancer Prevention and Population Science, The University of Texas M. D. Anderson Cancer Center, Houston, TX, USA
| | - Sunil Krishnan
- Department of Radiation Oncology, Division of Radiation Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, TX, USA
| | - Huan Xie
- Department of Pharmaceutical and Environmental Sciences, College of Pharmacy and Health Sciences, Texas Southern University, Houston, TX, USA
| |
Collapse
|
18
|
Supuran CT, Alterio V, Di Fiore A, D' Ambrosio K, Carta F, Monti SM, De Simone G. Inhibition of carbonic anhydrase IX targets primary tumors, metastases, and cancer stem cells: Three for the price of one. Med Res Rev 2018; 38:1799-1836. [PMID: 29635752 DOI: 10.1002/med.21497] [Citation(s) in RCA: 201] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 01/22/2018] [Accepted: 03/02/2018] [Indexed: 12/12/2022]
Abstract
Human carbonic anhydrase (CA) IX is a tumor-associated protein, since it is scarcely present in normal tissues, but highly overexpressed in a large number of solid tumors, where it actively contributes to survival and metastatic spread of tumor cells. Due to these features, the characterization of its biochemical, structural, and functional features for drug design purposes has been extensively carried out, with consequent development of several highly selective small molecule inhibitors and monoclonal antibodies to be used for different purposes. Aim of this review is to provide a comprehensive state-of-the-art of studies performed on this enzyme, regarding structural, functional, and biomedical aspects, as well as the development of molecules with diagnostic and therapeutic applications for cancer treatment. A brief description of additional pharmacologic applications for CA IX inhibition in other diseases, such as arthritis and ischemia, is also provided.
Collapse
Affiliation(s)
- Claudiu T Supuran
- Dipartimento Neurofarba, Sezione di Scienze Farmaceutiche e Nutraceutiche, Università degli Studi di Firenze, Florence, Italy
| | | | - Anna Di Fiore
- Istituto di Biostrutture e Bioimmagini-CNR, Naples, Italy
| | | | - Fabrizio Carta
- Dipartimento Neurofarba, Sezione di Scienze Farmaceutiche e Nutraceutiche, Università degli Studi di Firenze, Florence, Italy
| | | | | |
Collapse
|
19
|
Abstract
Frequently observed phenotypes of tumours include high metabolic activity, hypoxia and poor perfusion; these act to produce an acidic microenvironment. Cellular function depends on pH homoeostasis, and thus, tumours become dependent on pH regulatory mechanisms. Many of the proteins involved in pH regulation are highly expressed in tumours, and their expression is often of prognostic significance. The more acidic tumour microenvironment also has important implications with regard to chemotherapeutic and radiotherapeutic interventions. In addition, we review pH-sensing mechanisms, the role of pH regulation in tumour phenotype and the use of pH regulatory mechanisms as therapeutic targets.
Collapse
Affiliation(s)
- Alan McIntyre
- Molecular Oncology Laboratories, Department of Medical Oncology, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Adrian L Harris
- Molecular Oncology Laboratories, Department of Medical Oncology, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK.
| |
Collapse
|
20
|
Rogez-Florent T, Goossens L, Drucbert AS, Duban-Deweer S, Six P, Depreux P, Danzé PM, Goossens JF, Foulon C. Amine coupling versus biotin capture for the assessment of sulfonamide as ligands of hCA isoforms. Anal Biochem 2016; 511:42-51. [DOI: 10.1016/j.ab.2016.07.029] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Revised: 07/08/2016] [Accepted: 07/29/2016] [Indexed: 12/11/2022]
|
21
|
Chang DK, Moniz RJ, Xu Z, Sun J, Signoretti S, Zhu Q, Marasco WA. Human anti-CAIX antibodies mediate immune cell inhibition of renal cell carcinoma in vitro and in a humanized mouse model in vivo. Mol Cancer 2015; 14:119. [PMID: 26062742 PMCID: PMC4464115 DOI: 10.1186/s12943-015-0384-3] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Accepted: 05/11/2015] [Indexed: 12/17/2022] Open
Abstract
Background Carbonic anhydrase (CA) IX is a surface-expressed protein that is upregulated by the hypoxia inducible factor (HIF) and represents a prototypic tumor-associated antigen that is overexpressed on renal cell carcinoma (RCC). Therapeutic approaches targeting CAIX have focused on the development of CAIX inhibitors and specific immunotherapies including monoclonal antibodies (mAbs). However, current in vivo mouse models used to characterize the anti-tumor properties of fully human anti-CAIX mAbs have significant limitations since the role of human effector cells in tumor cell killing in vivo is not directly evaluated. Methods The role of human anti-CAIX mAbs on CAIX+ RCC tumor cell killing by immunocytes or complement was tested in vitro by antibody-dependent cell-mediated cytotoxicity (ADCC), complement-dependent cytotoxicity (CDC) and antibody-dependent cellular phagocytosis (ADCP) as well as on CAIX+ RCC cellular motility, wound healing, migration and proliferation. The in vivo therapeutic activity mediated by anti-CAIX mAbs was determined by using a novel orthotopic RCC xenograft humanized animal model and analyzed by histology and FACS staining. Results Our studies demonstrate the capacity of human anti-CAIX mAbs that inhibit CA enzymatic activity to result in immune-mediated killing of RCC, including nature killer (NK) cell-mediated ADCC, CDC, and macrophage-mediated ADCP. The killing activity correlated positively with the level of CAIX expression on RCC tumor cell lines. In addition, Fc engineering of anti-CAIX mAbs was shown to enhance the ADCC activity against RCC. We also demonstrate that these anti-CAIX mAbs inhibit migration of RCC cells in vitro. Finally, through the implementation of a novel orthotopic RCC model utilizing allogeneic human peripheral blood mononuclear cells in NOD/SCID/IL2Rγ−/− mice, we show that anti-CAIX mAbs are capable of mediating human immune response in vivo including tumor infiltration of NK cells and activation of T cells, resulting in inhibition of CAIX+ tumor growth. Conclusions Our findings demonstrate that these novel human anti-CAIX mAbs have therapeutic potential in the unmet medical need of targeted killing of HIF-driven CAIX+RCC. The orthotopic tumor xenografted humanized mouse provides an improved model to evaluate the in vivo anti-tumor capabilities of fully human mAbs for RCC therapy. Electronic supplementary material The online version of this article (doi:10.1186/s12943-015-0384-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- De-Kuan Chang
- Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, 450 Brookline Ave., Boston, MA, USA. .,Department of Medicine, Harvard Medical School, 25 Shattuck Street, Boston, MA, USA.
| | - Raymond J Moniz
- Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, 450 Brookline Ave., Boston, MA, USA. .,Department of Medicine, Harvard Medical School, 25 Shattuck Street, Boston, MA, USA.
| | - Zhongyao Xu
- Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, 450 Brookline Ave., Boston, MA, USA.
| | - Jiusong Sun
- Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, 450 Brookline Ave., Boston, MA, USA. .,Department of Medicine, Harvard Medical School, 25 Shattuck Street, Boston, MA, USA.
| | - Sabina Signoretti
- Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Ave., Boston, MA, USA. .,Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Quan Zhu
- Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, 450 Brookline Ave., Boston, MA, USA. .,Department of Medicine, Harvard Medical School, 25 Shattuck Street, Boston, MA, USA.
| | - Wayne A Marasco
- Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, 450 Brookline Ave., Boston, MA, USA. .,Department of Medicine, Harvard Medical School, 25 Shattuck Street, Boston, MA, USA.
| |
Collapse
|
22
|
Uda NR, Seibert V, Stenner-Liewen F, Müller P, Herzig P, Gondi G, Zeidler R, van Dijk M, Zippelius A, Renner C. Esterase activity of carbonic anhydrases serves as surrogate for selecting antibodies blocking hydratase activity. J Enzyme Inhib Med Chem 2015; 30:955-60. [DOI: 10.3109/14756366.2014.1001754] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Affiliation(s)
| | - Volker Seibert
- 4-Antibody AG (Wholly-Owned Subsidiary of Agenus Inc., USA), Basel, Switzerland,
| | - Frank Stenner-Liewen
- Department of Biomedicine, University of Basel, Basel, Switzerland,
- Department Internal Medicine, University Hospital Basel, Basel, Switzerland,
| | - Philipp Müller
- Department of Biomedicine, University of Basel, Basel, Switzerland,
| | - Petra Herzig
- Department of Biomedicine, University of Basel, Basel, Switzerland,
| | - Gabor Gondi
- Department of Otorhinolaryngology, Klinikum der Universität München, Munich, Germany, and
- Helmholtz Zentrum München – German Research Center for Environmental Health, Research Unit Gene Vectors, Munich, Germany
| | - Reinhard Zeidler
- Department of Otorhinolaryngology, Klinikum der Universität München, Munich, Germany, and
- Helmholtz Zentrum München – German Research Center for Environmental Health, Research Unit Gene Vectors, Munich, Germany
| | - Marc van Dijk
- 4-Antibody AG (Wholly-Owned Subsidiary of Agenus Inc., USA), Basel, Switzerland,
| | - Alfred Zippelius
- Department of Biomedicine, University of Basel, Basel, Switzerland,
- Department Internal Medicine, University Hospital Basel, Basel, Switzerland,
| | - Christoph Renner
- Department of Biomedicine, University of Basel, Basel, Switzerland,
| |
Collapse
|
23
|
Mboge MY, McKenna R, Frost SC. Advances in Anti-Cancer Drug Development Targeting Carbonic Anhydrase IX and XII. TOPICS IN ANTI-CANCER RESEARCH 2015; 5:3-42. [PMID: 30272043 PMCID: PMC6162069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
The microenvironment within a solid tumor is heterogeneous with regions being both acidic and hypoxic. As a result of this, cancer cells upregulate genes that allow survival in such environments. Some of these genes are pH regulatory factors, including carbonic anhydrase IX (CA IX) and in some cases XII (CA XII). CA IX helps to maintain normal cytoplasmic pH (pHi) while simultaneously contributing to the extracellular pH (pHe). CA XII is also thought to be responsible for stabilizing pHe at physiological conditions. Extracellular acidification of the tumor microenvironment promotes local invasion and metastasis while decreasing the effectiveness of adjuvant therapies, thus contributing to poor cancer clinical outcomes. In this review, we describe the properties of CA IX and CA XII that substantiate their potential use as anticancer targets. We also discuss the current status of CA isoform-selective inhibitor development and patents of CA IX/XII targeted inhibitors that show potential for treating aggressive tumors. Some of the recently published patents discussed include sulfonamide-based small molecule inhibitors including derivatives of boron cluster compounds; metal complexes of poly(carboxyl)amine-containing ligands; nitroi-midazole-, ureidosulfonamide-, and coumarin-based compounds; as well as G250 and A610 monoclonal antibodies for cancer treatment.
Collapse
Affiliation(s)
- Mam Y. Mboge
- Corresponding authors Mam Y. Mboge and Susan C. Frost: University of Florida, College of Medicine, Department of Biochemistry and Molecular Biology, Box 100245, Gainesville, FL 32610, USA; Tel +1 352 294-8386, Fax +1 352 392-2953, ,
| | | | - Susan C. Frost
- Corresponding authors Mam Y. Mboge and Susan C. Frost: University of Florida, College of Medicine, Department of Biochemistry and Molecular Biology, Box 100245, Gainesville, FL 32610, USA; Tel +1 352 294-8386, Fax +1 352 392-2953, ,
| |
Collapse
|
24
|
Ilardi G, Zambrano N, Merolla F, Siano M, Varricchio S, Vecchione M, De Rosa G, Mascolo M, Staibano S. Histopathological determinants of tumor resistance: a special look to the immunohistochemical expression of carbonic anhydrase IX in human cancers. Curr Med Chem 2014; 21:1569-82. [PMID: 23992304 PMCID: PMC3979091 DOI: 10.2174/09298673113209990227] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2013] [Revised: 06/20/2013] [Accepted: 07/10/2013] [Indexed: 02/07/2023]
Abstract
Intrinsic and acquired drug resistance of tumor cells still causes the failure of treatment regimens in advanced
human cancers. It may be driven by intrinsic tumor cells features, or may also arise from micro environmental influences.
Hypoxia is a microenvironment feature associated with the aggressiveness and metastasizing ability of human solid cancers.
Hypoxic cancer cells overexpress Carbonic Anhydrase IX (CA IX). CA IX ensures a favorable tumor intracellular
pH, while contributing to stromal acidosis, which facilitates tumor invasion and metastasis. The overexpression of CA IX
is considered an epiphenomenon of the presence of hypoxic, aggressive tumor cells. Recently, a relationship between CA
IX overexpression and the cancer stem cells (CSCs) population has been hypothesized. CSCs are strictly regulated by tumor
hypoxia and drive a major non-mutational mechanism of cancer drug-resistance. We reviewed the current data concerning
the role of CA IX overexpression in human malignancies, extending such information to the expression of the
stem cells markers CD44 and nestin in solid cancers, to explore their relationship with the biological behavior of tumors.
CA IX is heavily expressed in advanced tumors. A positive trend of correlation between CA IX overexpression, tumor
stage/grade and poor outcome emerged. Moreover, stromal CA IX expression was associated with adverse events occurrence,
maybe signaling the direct action of CA IX in directing the mesenchymal changes that favor tumor invasion; in addition,
membranous/cytoplasmic co-overexpression of CA IX and stem cells markers were found in several aggressive
tumors. This suggests that CA IX targeting could indirectly deplete CSCs and counteract resistance of solid cancers in the
clinical setting.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - S Staibano
- Department of Advanced Biomedical Sciences, Pathology Section, School of Medicine and Surgery, University of Naples "Federico II", address: via S. Pansini, n.5, 80131, Naples, Italy.
| |
Collapse
|
25
|
ZATOVICOVA MIRIAM, JELENSKA LENKA, HULIKOVA ALZBETA, DITTE PETER, DITTE ZUZANA, CSADEROVA LUCIA, SVASTOVA ELISKA, SCHMALIX WOLFGANG, BOETTGER VOLKER, BEVAN PAUL, PASTOREK JAROMIR, PASTOREKOVA SILVIA. Monoclonal antibody G250 targeting CA IX: Binding specificity, internalization and therapeutic effects in a non-renal cancer model. Int J Oncol 2014; 45:2455-67. [DOI: 10.3892/ijo.2014.2658] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Accepted: 08/20/2014] [Indexed: 11/06/2022] Open
|
26
|
Pastorek J, Pastorekova S. Hypoxia-induced carbonic anhydrase IX as a target for cancer therapy: from biology to clinical use. Semin Cancer Biol 2014; 31:52-64. [PMID: 25117006 DOI: 10.1016/j.semcancer.2014.08.002] [Citation(s) in RCA: 230] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2014] [Revised: 07/30/2014] [Accepted: 08/04/2014] [Indexed: 12/12/2022]
Abstract
The tumor microenvironment includes a complicated network of physiological gradients contributing to plasticity of tumor cells and heterogeneity of tumor tissue. Hypoxia is a key component generating intratumoral oxygen gradients, which affect the cellular expression program and lead to therapy resistance and increased metastatic propensity of weakly oxygenated cell subpopulations. One of the adaptive responses of tumor cells to hypoxia involves the increased expression and functional activation of carbonic anhydrase IX (CA IX), a cancer-related cell surface enzyme catalyzing the reversible conversion of carbon dioxide to bicarbonate ion and proton. Via its catalytic activity, CA IX participates in regulation of intracellular and extracellular pH perturbations that result from hypoxia-induced changes in cellular metabolism producing excess of acid. Through the ability to regulate pH, CA IX also facilitates cell migration and invasion. In addition, CA IX has non-catalytic function in cell adhesion and spreading. Thus, CA IX endows tumor cells with survival advantages in hypoxia/acidosis and confers an increased ability to migrate, invade and metastasize. Accordingly, CA IX is expressed in a broad range of tumors, where it is associated with prognosis and therapy outcome. Its expression pattern and functional implications in tumor biology make CA IX a promising therapeutic target, which can be hit either by immunotherapy with monoclonal antibodies or with compounds inhibiting its enzyme activity. The first strategy has already reached the clinical trials, whereas the second one is still in preclinical testing. Both strategies indicate that CA IX can become a clinically useful anticancer target, but urge further efforts toward better selection of patients for immunotherapy and deeper understanding of tumor types, clinical situations and synthetic lethality interactions with other treatment approaches.
Collapse
Affiliation(s)
- Jaromir Pastorek
- Department of Molecular Medicine, Institute of Virology, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Silvia Pastorekova
- Department of Molecular Medicine, Institute of Virology, Slovak Academy of Sciences, Bratislava, Slovakia; Regional Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Brno, Czech Republic.
| |
Collapse
|
27
|
Abstract
Tumor microenvironment substantially influences the process of tumorigenesis. In many solid tumors, imbalance between the demand of rapidly proliferating cancer cells and the capabilities of the vascular system generates areas with insufficient oxygen supply. In response to tumor hypoxia, cancer cells modulate their gene expression pattern to match the requirements of the altered microenvironment. One of the most significant adaptations to this milieu is the shift towards anaerobic glycolysis to keep up the energy demands. This oncogenic metabolism is often maintained also in aerobic cells. Lactic acid, its metabolic end-product, accumulates hand-in-hand with carbon dioxide, leading to acidification of the extracellular environment. Carbonic anhydrase IX (CA IX) is the most widely expressed gene in response to hypoxia. Its crucial role in intracellular pH maintenance represents the means by which cancer cells adapt to the toxic conditions of the extracellular milieu. Furthermore, the activity of CA IX stimulates the migratory pathways of cancer cells and is connected with the increase of the aggressive/invasive phenotype of tumors. CA IX expression in many types of tumors indicates its relevance as a general marker of tumor hypoxia. Moreover, its expression is closely related to prognosis of the clinical outcome in several tumor types. All above mentioned facts support the strong position of CA IX as a potential drug therapy target. Here, we summarize the state-of-the-art knowledge on its regulation and role in cancer development.
Collapse
|
28
|
Tafreshi NK, Lloyd MC, Bui MM, Gillies RJ, Morse DL. Carbonic anhydrase IX as an imaging and therapeutic target for tumors and metastases. Subcell Biochem 2014; 75:221-54. [PMID: 24146382 DOI: 10.1007/978-94-007-7359-2_12] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Carbonic anhydrase IX (CAIX) which is a zinc containing metalloprotein, efficiently catalyzes the reversible hydration of carbon dioxide. It is constitutively up-regulated in several cancer types and has an important role in tumor progression, acidification and metastasis. High expression of CAIX generally correlates with poor prognosis and is related to a decrease in the disease-free interval following successful therapy. Therefore, it is considered as a prognostic indicator in oncology.In this review, we describe CAIX regulation and its role in tumor hypoxia, acidification and metastasis. In addition, the molecular imaging of CAIX and its potential for use in cancer detection, diagnosis, staging, and for use in following therapy response is discussed. Both antibodies and small molecular weight compounds have been used for targeted imaging of CAIX expression. The use of CAIX expression as an attractive and promising candidate marker for systemic anticancer therapy is also discussed.
Collapse
Affiliation(s)
- Narges K Tafreshi
- Department of Cancer Imaging and Metabolism, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA,
| | | | | | | | | |
Collapse
|
29
|
|
30
|
Gondi G, Mysliwietz J, Hulikova A, Jen JP, Swietach P, Kremmer E, Zeidler R. Antitumor efficacy of a monoclonal antibody that inhibits the activity of cancer-associated carbonic anhydrase XII. Cancer Res 2013; 73:6494-503. [PMID: 24030978 DOI: 10.1158/0008-5472.can-13-1110] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Carbonic anhydrase XII (CA XII) is a membrane-tethered cell surface enzyme that is highly expressed on many human tumor cells. Carbonic anhydrase members in this class of exofacial molecules facilitate tumor metabolism by facilitating CO2 venting and intracellular pH regulation. Accordingly, inhibition of exofacial CAs has been proposed as a general therapeutic strategy to target cancer. The recent characterization of 6A10, the first CA XII-specific inhibitory monoclonal antibody, offered an opportunity to evaluate this strategy with regard to CA XII-mediated catalysis. Using functional assays, we showed that 6A10 inhibited exofacial CA activity in CA XII-expressing cancer cells. 6A10 reduced spheroid growth in vitro under culture conditions where CA XII was active (i.e., alkaline pH) and where its catalytic activity was likely rate-limiting (i.e., restricted extracellular HCO3-supply). These in vitro results argued that the antibody exerted its growth-retarding effect by acting on the catalytic process, rather than on antigen binding per se. Notably, when administered in a mouse xenograft model of human cancer, 6A10 exerted a significant delay on tumor outgrowth. These results corroborate the notion that exofacial CA is critical for cancer cell physiology and they establish the immunotherapeutic efficacy of targeting CA XII using an inhibitory antibody.
Collapse
Affiliation(s)
- Gabor Gondi
- Authors' Affiliations: Research Unit Gene Vectors, and Institute of Molecular Immunology, Helmholtz Zentrum München-German Research Center for Environmental Health; Department of Otorhinolaryngology, Ludwig-Maximilians-Universität, Munich, Germany; Department Of Physiology, Anatomy and Genetics, Oxford University, Oxford, United Kingdom; Munich, Germany
| | | | | | | | | | | | | |
Collapse
|
31
|
Chadwick AL, Howell A, Sotgia F, Lisanti MP. Carbonic anhydrase 9 (CA9) and redox signaling in cancer-associated fibroblasts: therapeutic implications. Cell Cycle 2013; 12:2534-5. [PMID: 23907127 PMCID: PMC3865039 DOI: 10.4161/cc.25842] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Affiliation(s)
- Amy L Chadwick
- Manchester Breast Centre & Breakthrough Breast Cancer Research Unit, Faculty Institute of Cancer Sciences, University of Manchester, Manchester, UK
| | | | | | | |
Collapse
|
32
|
Molecular recognition moiety and its target biomolecule interact in switching enzyme activity. J Biosci Bioeng 2013; 115:639-44. [DOI: 10.1016/j.jbiosc.2012.12.019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2012] [Revised: 12/25/2012] [Accepted: 12/26/2012] [Indexed: 01/02/2023]
|
33
|
Application of monoclonal antibody G250 recognizing carbonic anhydrase IX in renal cell carcinoma. Int J Mol Sci 2013; 14:11402-23. [PMID: 23759990 PMCID: PMC3709739 DOI: 10.3390/ijms140611402] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2013] [Revised: 05/14/2013] [Accepted: 05/15/2013] [Indexed: 11/16/2022] Open
Abstract
Monoclonal antibody G250 (mAbG250) recognizes a determinant on carbonic anhydrase IX (CAIX). CAIX is expressed by virtually all renal cell carcinomas of the clear cell type (ccRCC), but expression in normal tissues is restricted. The homogeneous CAIX expression in ccRCC and excellent targeting capability of mAbG250 in animal models led to the initiation of the clinical evaluation of mAbG250 in (metastatic) RCC (mRCC) patients. Clinical studies confirmed the outstanding targeting ability of mAbG250 and cG250 PET imaging, as diagnostic modality holds great promise for the future, both in detecting localized and advanced disease. Confirmation of the results obtained in the non-randomized clinical trials with unmodified cG250 is needed to substantiate the value of cG250 treatment in mRCC. cG250-Based radio immuno-therapy (RIT) holds promise for treatment of patients with small-volume disease, and adjuvant treatment with unmodified cG250 may be of value in selected cases. In the upcoming years, ongoing clinical trials should provide evidence for these assumptions. Lastly, whether cG250-based RIT can be combined with tyrosine kinase inhibitors, which constitutes the current standard treatment for mRCC, needs to be established.
Collapse
|
34
|
Fiaschi T, Giannoni E, Taddei ML, Cirri P, Marini A, Pintus G, Nativi C, Richichi B, Scozzafava A, Carta F, Torre E, Supuran CT, Chiarugi P. Carbonic anhydrase IX from cancer-associated fibroblasts drives epithelial-mesenchymal transition in prostate carcinoma cells. Cell Cycle 2013; 12:1791-801. [PMID: 23656776 DOI: 10.4161/cc.24902] [Citation(s) in RCA: 115] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Extracellular acidification, a mandatory feature of several malignancies, has been mainly correlated with metabolic reprogramming of tumor cells toward Warburg metabolism, as well as to the expression of carbonic anydrases or proton pumps by malignant tumor cells. We report herein that for aggressive prostate carcinoma, acknowledged to be reprogrammed toward an anabolic phenotype and to upload lactate to drive proliferation, extracellular acidification is mainly mediated by stromal cells engaged in a molecular cross-talk circuitry with cancer cells. Indeed, cancer-associated fibroblasts, upon their activation by cancer delivered soluble factors, rapidly express carbonic anhydrase IX (CA IX). While expression of CAIX in cancer cells has already been correlated with poor prognosis in various human tumors, the novelty of our findings is the upregulation of CAIX in stromal cells upon activation. The de novo expression of CA IX, which is not addicted to hypoxic conditions, is driven by redox-based stabilization of hypoxia-inducible factor-1. Extracellular acidification due to carbonic anhydrase IX is mandatory to elicit activation of stromal fibroblasts delivered metalloprotease-2 and -9, driving in cancer cells the epithelial-mesenchymal transition epigenetic program, a key event associated with increased motility, survival and stemness. Both genetic silencing and pharmacological inhibition of CA IX (with sulfonamide/sulfamides potent inhibitors) or metalloprotease-9 are sufficient to impede epithelial-mesenchymal transition and invasiveness of prostate cancer cells induced by contact with cancer-associated fibroblasts. We also confirmed in vivo the upstream hierarchical role of stromal CA IX to drive successful metastatic spread of prostate carcinoma cells. These data include stromal cells, as cancer-associated fibroblasts as ideal targets for carbonic anhydrase IX-directed anticancer therapies.
Collapse
Affiliation(s)
- Tania Fiaschi
- Department of Biomedical, Experimental and Clinical Sciences, University of Florence, Tuscany, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
McDonald PC, Winum JY, Supuran CT, Dedhar S. Recent developments in targeting carbonic anhydrase IX for cancer therapeutics. Oncotarget 2012; 3:84-97. [PMID: 22289741 PMCID: PMC3292895 DOI: 10.18632/oncotarget.422] [Citation(s) in RCA: 333] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Carbonic anhydrase IX (CAIX) is a hypoxia-inducible enzyme that is overexpressed by cancer cells from many tumor types, and is a component of the pH regulatory system invoked by these cells to combat the deleterious effects of a high rate of glycolytic metabolism. CAIX functions to help produce and maintain an intracellular pH (pHi) favorable for tumor cell growth and survival, while at the same time participating in the generation of an increasingly acidic extracellular space, facilitating tumor cell invasiveness. Pharmacologic interference of CAIX catalytic activity using monoclonal antibodies or CAIX-specific small molecule inhibitors, consequently disrupting pH regulation by cancer cells, has been shown recently to impair primary tumor growth and metastasis. Many of these agents are in preclinical or clinical development and constitute a novel, targeted strategy for cancer therapy.
Collapse
Affiliation(s)
- Paul C McDonald
- Department of Integrative Oncology, British Columbia Cancer Research Centre and Cancer Agency, Vancouver, BC, Canada
| | | | | | | |
Collapse
|
36
|
Porporato PE, Dhup S, Dadhich RK, Copetti T, Sonveaux P. Anticancer targets in the glycolytic metabolism of tumors: a comprehensive review. Front Pharmacol 2011; 2:49. [PMID: 21904528 PMCID: PMC3161244 DOI: 10.3389/fphar.2011.00049] [Citation(s) in RCA: 332] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2011] [Accepted: 08/05/2011] [Indexed: 12/21/2022] Open
Abstract
CANCER IS A METABOLIC DISEASE AND THE SOLUTION OF TWO METABOLIC EQUATIONS: to produce energy with limited resources and to fulfill the biosynthetic needs of proliferating cells. Both equations are solved when glycolysis is uncoupled from oxidative phosphorylation in the tricarboxylic acid cycle, a process known as the glycolytic switch. This review addresses in a comprehensive manner the main molecular events accounting for high-rate glycolysis in cancer. It starts from modulation of the Pasteur Effect allowing short-term adaptation to hypoxia, highlights the key role exerted by the hypoxia-inducible transcription factor HIF-1 in long-term adaptation to hypoxia, and summarizes the current knowledge concerning the necessary involvement of aerobic glycolysis (the Warburg effect) in cancer cell proliferation. Based on the many observations positioning glycolysis as a central player in malignancy, the most advanced anticancer treatments targeting tumor glycolysis are briefly reviewed.
Collapse
Affiliation(s)
- Paolo E Porporato
- Pole of Pharmacology and Therapeutics, Institute of Experimental and Clinical Research, University of Louvain Medical School Brussels, Belgium
| | | | | | | | | |
Collapse
|