1
|
Nemakhavhani L, Abrahamse H, Kumar SSD. A review on dendrimer-based nanoconjugates and their intracellular trafficking in cancer photodynamic therapy. ARTIFICIAL CELLS, NANOMEDICINE, AND BIOTECHNOLOGY 2024; 52:384-398. [PMID: 39101753 DOI: 10.1080/21691401.2024.2368033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 06/03/2024] [Accepted: 06/10/2024] [Indexed: 08/06/2024]
Abstract
Nanotechnology-based cancer treatment has received considerable attention, and these treatments generally use drug-loaded nanoparticles (NPs) to target and destroy cancer cells. Nanotechnology combined with photodynamic therapy (PDT) has demonstrated positive outcomes in cancer therapy. Combining nanotechnology and PDT is effective in targeting metastatic cancer cells. Nanotechnology can also increase the effectiveness of PDT by targeting cells at a molecular level. Dendrimer-based nanoconjugates (DBNs) are highly stable and biocompatible, making them suitable for drug delivery applications. Moreover, the hyperbranched structures in DBNs have the capacity to load hydrophobic compounds, such as photosensitizers (PSs) and chemotherapy drugs, and deliver them efficiently to tumour cells. This review primarily focuses on DBNs and their potential applications in cancer treatment. We discuss the chemical design, mechanism of action, and targeting efficiency of DBNs in tumour metastasis, intracellular trafficking in cancer treatment, and DBNs' biocompatibility, biodegradability and clearance properties. Overall, this study will provide the most recent insights into the application of DBNs and PDT in cancer therapy.
Collapse
Affiliation(s)
- Lufuno Nemakhavhani
- Laser Research Centre, University of Johannesburg, Johannesburg, South Africa
| | - Heidi Abrahamse
- Laser Research Centre, University of Johannesburg, Johannesburg, South Africa
| | | |
Collapse
|
2
|
Cosmi V, Wang B, Goorden MC, Beekman FJ. NaI gamma camera performance for high energies: Effects of crystal thickness, photomultiplier tube geometry and light guide thickness. Med Phys 2024; 51:4696-4708. [PMID: 38569052 DOI: 10.1002/mp.17043] [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: 06/28/2023] [Revised: 12/18/2023] [Accepted: 03/12/2024] [Indexed: 04/05/2024] Open
Abstract
BACKGROUND Gamma camera imaging, including single photon emission computed tomography (SPECT), is crucial for research, diagnostics, and radionuclide therapy. Gamma cameras are predominantly based on arrays of photon multipliers tubes (PMTs) that read out NaI(Tl) scintillation crystals. In this way, standard gamma cameras can localize ɣ-rays with energies typically ranging from 30 to 360 keV. In the last decade, there has been an increasing interest towards gamma imaging outside this conventional clinical energy range, for example, for theragnostic applications and preclinical multi-isotope positron emission tomography (PET) and PET-SPECT. However, standard gamma cameras are typically equipped with 9.5 mm thick NaI(Tl) crystals which can result in limited sensitivity for these higher energies. PURPOSE Here we investigate to what extent thicker scintillators can improve the photopeak sensitivity for higher energy isotopes while attempting to maintain spatial resolution. METHODS Using Monte Carlo simulations, we analyzed multiple PMT-based configurations of gamma detectors with monolithic NaI (Tl) crystals of 20 and 40 mm thickness. Optimized light guide thickness together with 2-inch round, 3-inch round, 60 × 60 mm2 square, and 76 × 76 mm2 square PMTs were tested. For each setup, we assessed photopeak sensitivity, energy resolution, spatial, and depth-of-interaction (DoI) resolution for conventional (140 keV) and high (511 keV) energy ɣ using a maximum-likelihood algorithm. These metrics were compared to those of a "standard" 9.5 mm-thick crystal detector with 3-inch round PMTs. RESULTS Estimated photopeak sensitivities for 511 keV were 27% and 53% for 20 and 40 mm thick scintillators, which is respectively, 2.2 and 4.4 times higher than for 9.5 mm thickness. In most cases, energy resolution benefits from using square PMTs instead of round ones, regardless of their size. Lateral and DoI spatial resolution are best for smaller PMTs (2-inch round and 60 × 60 mm2 square) which outperform the more cost-effective larger PMT setups (3-inch round and 76 × 76 mm2 square), while PMT layout and shape have negligible (< 10%) effect on resolution. Best spatial resolution was obtained with 60 × 60 mm2 PMTs; for 140 keV, lateral resolution was 3.5 mm irrespective of scintillator thickness, improving to 2.8 and 2.9 mm for 511 keV with 20 and 40 mm thick crystals, respectively. Using the 3-inch round PMTs, lateral resolutions of 4.5 and 3.9 mm for 140 keV and of 3.5 and 3.7 mm for 511 keV were obtained with 20 and 40 mm thick crystals respectively, indicating a moderate performance degradation compared to the 3.5 and 2.9 mm resolution obtained by the standard detector for 140 and 511 keV. Additionally, DoI resolution for 511 keV was 7.0 and 5.6 mm with 20 and 40 mm crystals using 60 × 60 mm2 square PMTs, while with 3-inch round PMTs 12.1 and 5.9 mm were obtained. CONCLUSION Depending on PMT size and shape, the use of thicker scintillator crystals can substantially improve detector sensitivity at high gamma energies, while spatial resolution is slightly improved or mildly degraded compared to standard crystals.
Collapse
Affiliation(s)
- Valerio Cosmi
- Department of Radiation Science and Technology, Delft University of Technology, Delft, The Netherlands
| | - Beien Wang
- Department of Radiation Science and Technology, Delft University of Technology, Delft, The Netherlands
| | - Marlies C Goorden
- Department of Radiation Science and Technology, Delft University of Technology, Delft, The Netherlands
| | - Freek J Beekman
- Department of Radiation Science and Technology, Delft University of Technology, Delft, The Netherlands
- Free Bee International, Gouda, The Netherlands
| |
Collapse
|
3
|
Al Zorgani MM, Ugail H, Pors K, Dauda AM. Deep Transfer Learning-Based Approach for Glucose Transporter-1 (GLUT1) Expression Assessment. J Digit Imaging 2023; 36:2367-2381. [PMID: 37670181 PMCID: PMC10584776 DOI: 10.1007/s10278-023-00859-0] [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: 11/21/2021] [Revised: 05/20/2023] [Accepted: 05/22/2023] [Indexed: 09/07/2023] Open
Abstract
Glucose transporter-1 (GLUT-1) expression level is a biomarker of tumour hypoxia condition in immunohistochemistry (IHC)-stained images. Thus, the GLUT-1 scoring is a routine procedure currently employed for predicting tumour hypoxia markers in clinical practice. However, visual assessment of GLUT-1 scores is subjective and consequently prone to inter-pathologist variability. Therefore, this study proposes an automated method for assessing GLUT-1 scores in IHC colorectal carcinoma images. For this purpose, we leverage deep transfer learning methodologies for evaluating the performance of six different pre-trained convolutional neural network (CNN) architectures: AlexNet, VGG16, GoogleNet, ResNet50, DenseNet-201 and ShuffleNet. The target CNNs are fine-tuned as classifiers or adapted as feature extractors with support vector machine (SVM) to classify GLUT-1 scores in IHC images. Our experimental results show that the winning model is the trained SVM classifier on the extracted deep features fusion Feat-Concat from DenseNet201, ResNet50 and GoogLeNet extractors. It yields the highest prediction accuracy of 98.86%, thus outperforming the other classifiers on our dataset. We also conclude, from comparing the methodologies, that the off-the-shelf feature extraction is better than the fine-tuning model in terms of time and resources required for training.
Collapse
Affiliation(s)
- Maisun Mohamed Al Zorgani
- Faculty of Engineering and Informatics, School of Media, Design and Technology, University of Bradford, Richmond Road, Bradford, BD7 1DP, UK.
| | - Hassan Ugail
- Faculty of Engineering and Informatics, School of Media, Design and Technology, University of Bradford, Richmond Road, Bradford, BD7 1DP, UK
| | - Klaus Pors
- Institute of Cancer Therapeutics, University of Bradford, Richmond Road, Bradford, BD7 1DP, UK
| | - Abdullahi Magaji Dauda
- Institute of Cancer Therapeutics, University of Bradford, Richmond Road, Bradford, BD7 1DP, UK
| |
Collapse
|
4
|
Bhise K, Gavande NS, Iyer AK. Leveraging hypoxia in triple-negative breast cancer as a promising treatment strategy. Drug Discov Today 2023; 28:103761. [PMID: 37660983 DOI: 10.1016/j.drudis.2023.103761] [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: 05/11/2023] [Revised: 08/08/2023] [Accepted: 08/29/2023] [Indexed: 09/05/2023]
Abstract
Current treatment strategies for triple-negative breast cancer (TNBC) are based upon conventional chemotherapy, immunotherapy, or a combination of both. The treatment regimen for chemotherapy is often a combination of two or more drugs, either dose dense or low dose for synergy. Anthracyclines, alkylating agents, antimicrotubule agents, and antimetabolites for early-stage TNBC; and antimetabolites, non-taxane microtubule inhibitors, and cross-linker platinums for late-stage TNBC are usually administered in the clinical setting. Newer options for patients with advanced TNBC, such as poly (ADP-ribose) polymerase (PARP) inhibitors and immune checkpoint inhibitors, have recently emerged for cases where surgery is not a viable option and the disease has metastasized. This review outlines the current trends in hypoxia-inspired treatment strategies for TNBC with a focus on clinical trials.
Collapse
Affiliation(s)
- Ketki Bhise
- Use-inspired Biomaterials & Integrated Nano Delivery (U-BiND) Systems Laboratory, Department of Pharmaceutical Sciences, Wayne State University, Detroit, MI, USA
| | - Navnath S Gavande
- Department of Pharmaceutical Sciences, Wayne State University, Detroit, MI, USA; Molecular Therapeutics Program, Karmanos Cancer Institute, Detroit, MI, USA
| | - Arun K Iyer
- Use-inspired Biomaterials & Integrated Nano Delivery (U-BiND) Systems Laboratory, Department of Pharmaceutical Sciences, Wayne State University, Detroit, MI, USA; Molecular Imaging Program, Karmanos Cancer Institute, Detroit, MI, USA.
| |
Collapse
|
5
|
Boreel DF, Span PN, Kip A, Boswinkel M, Peters JPW, Adema GJ, Bussink J, Heskamp S. Quantitative Imaging of Hypoxic CAIX-Positive Tumor Areas with Low Immune Cell Infiltration in Syngeneic Mouse Tumor Models. Mol Pharm 2023; 20:2245-2255. [PMID: 36882391 PMCID: PMC10074386 DOI: 10.1021/acs.molpharmaceut.3c00045] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/09/2023]
Abstract
Limited diffusion of oxygen in combination with increased oxygen consumption leads to chronic hypoxia in most solid malignancies. This scarcity of oxygen is known to induce radioresistance and leads to an immunosuppressive microenvironment. Carbonic anhydrase IX (CAIX) is an enzyme functioning as a catalyzer for acid export in hypoxic cells and is an endogenous biomarker for chronic hypoxia. The aim of this study is to develop a radiolabeled antibody that recognizes murine CAIX to visualize chronic hypoxia in syngeneic tumor models and to study the immune cell population in these hypoxic areas. An anti-mCAIX antibody (MSC3) was conjugated to diethylenetriaminepentaacetic acid (DTPA) and radiolabeled with indium-111 (111In). CAIX expression on murine tumor cells was determined using flow cytometry, and in vitro affinity of [111In]In-MSC3 was analyzed in a competitive binding assay. Ex vivo biodistribution studies were performed to determine in vivo radiotracer distribution. CAIX+ tumor fractions were determined by mCAIX microSPECT/CT, and the tumor microenvironment was analyzed using immunohistochemistry and autoradiography. We showed that [111In]In-MSC3 binds to CAIX-expressing (CAIX+) murine cells in vitro and accumulates in CAIX+ areas in vivo. We optimized the use of [111In]In-MSC3 for preclinical imaging such that it can be applied in syngeneic mouse models and showed that we can quantitatively distinguish between tumor models with varying CAIX+ fractions by ex vivo analyses and in vivo mCAIX microSPECT/CT. Analysis of the tumor microenvironment identified these CAIX+ areas as less infiltrated by immune cells. Together these data demonstrate that mCAIX microSPECT/CT is a sensitive technique to visualize hypoxic CAIX+ tumor areas that exhibit reduced infiltration of immune cells in syngeneic mouse models. In the future, this technique may enable visualization of CAIX expression before or during hypoxia-targeted or hypoxia-reducing treatments. Thereby, it will help optimize immuno- and radiotherapy efficacy in translationally relevant syngeneic mouse tumor models.
Collapse
Affiliation(s)
- Daan F Boreel
- Radiotherapy and OncoImmunology Laboratory, Radiation Oncology, Radboud University Medical Center, Geert Grooteplein Zuid 32, 6525GA Nijmegen, The Netherlands.,Department of Medical Imaging, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525GA Nijmegen, The Netherlands
| | - Paul N Span
- Radiotherapy and OncoImmunology Laboratory, Radiation Oncology, Radboud University Medical Center, Geert Grooteplein Zuid 32, 6525GA Nijmegen, The Netherlands
| | - Annemarie Kip
- Department of Medical Imaging, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525GA Nijmegen, The Netherlands
| | - Milou Boswinkel
- Department of Medical Imaging, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525GA Nijmegen, The Netherlands
| | - Johannes P W Peters
- Radiotherapy and OncoImmunology Laboratory, Radiation Oncology, Radboud University Medical Center, Geert Grooteplein Zuid 32, 6525GA Nijmegen, The Netherlands
| | - Gosse J Adema
- Radiotherapy and OncoImmunology Laboratory, Radiation Oncology, Radboud University Medical Center, Geert Grooteplein Zuid 32, 6525GA Nijmegen, The Netherlands
| | - Johan Bussink
- Radiotherapy and OncoImmunology Laboratory, Radiation Oncology, Radboud University Medical Center, Geert Grooteplein Zuid 32, 6525GA Nijmegen, The Netherlands
| | - Sandra Heskamp
- Department of Medical Imaging, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525GA Nijmegen, The Netherlands
| |
Collapse
|
6
|
Grajek J, Kather JN, Poleszczuk J. An in silico model to study the impact of carbonic anhydrase IX expression on tumour growth and anti-PD-1 therapy. J R Soc Interface 2023; 20:20220654. [PMID: 36695125 PMCID: PMC9874981 DOI: 10.1098/rsif.2022.0654] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 01/04/2023] [Indexed: 01/26/2023] Open
Abstract
Immune checkpoint inhibitors (ICIs) are revolutionary cancer treatments. However, the mechanisms behind their effectiveness are not yet fully understood. Here, we aimed to investigate the role of the pH-regulatory enzyme carbonic anhydrase IX (CAIX) in ICI success. Consequently, we developed an in silico model of the tumour microenvironment. The hybrid model consists of an agent-based model of tumour-immune cell interactions, coupled with a set of diffusion-reaction equations describing substances in the environment. It is calibrated with data from the literature, enabling the study of its qualitative behaviour. In our model, CAIX-expressing tumours acidified their neighbourhood, thereby reducing immune infiltration by 90% (p < 0.001) and resulting in a 25% increase in tumour burden (p < 0.001). Moreover, suppression of CAIX improved the response to anti-PD-1 (23% tumour reduction in CAIX knockouts and 6% in CAIX-expressing tumours, p < 0.001), independently of initial PD-L1 expression. Our simulations suggest that patients with CAIX-expressing tumours could respond favourably to combining ICIs with CAIX suppression, even in the absence of pre-treatment PD-L1 expression. Furthermore, when calibrated with tumour-type-specific data, our model could serve as a high-throughput tool for testing the effectiveness of such a combinatorial approach.
Collapse
Affiliation(s)
- Julia Grajek
- Nalecz Institute of Biocybernetics and Biomedical Engineering, Polish Academy of Sciences, Warsaw 02-109, Poland
| | - Jakob Nikolas Kather
- Else Kroener Fresenius Center for Digital Health, Technical University Dresden, Dresden 01309, Germany
| | - Jan Poleszczuk
- Nalecz Institute of Biocybernetics and Biomedical Engineering, Polish Academy of Sciences, Warsaw 02-109, Poland
| |
Collapse
|
7
|
Promise of hypoxia-targeted tracers in metastatic lymph node imaging. Eur J Nucl Med Mol Imaging 2022; 49:4293-4297. [PMID: 35994060 DOI: 10.1007/s00259-022-05938-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
|
8
|
van der Heide CD, Dalm SU. Radionuclide imaging and therapy directed towards the tumor microenvironment: a multi-cancer approach for personalized medicine. Eur J Nucl Med Mol Imaging 2022; 49:4616-4641. [PMID: 35788730 PMCID: PMC9606105 DOI: 10.1007/s00259-022-05870-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 06/09/2022] [Indexed: 12/19/2022]
Abstract
Targeted radionuclide theranostics is becoming more and more prominent in clinical oncology. Currently, most nuclear medicine compounds researched for cancer theranostics are directed towards targets expressed in only a small subset of cancer types, limiting clinical applicability. The identification of cancer-specific targets that are (more) universally expressed will allow more cancer patients to benefit from these personalized nuclear medicine–based interventions. A tumor is not merely a collection of cancer cells, it also comprises supporting stromal cells embedded in an altered extracellular matrix (ECM), together forming the tumor microenvironment (TME). Since the TME is less genetically unstable than cancer cells, and TME phenotypes can be shared between cancer types, it offers targets that are more universally expressed. The TME is characterized by the presence of altered processes such as hypoxia, acidity, and increased metabolism. Next to the ECM, the TME consists of cancer-associated fibroblasts (CAFs), macrophages, endothelial cells forming the neo-vasculature, immune cells, and cancer-associated adipocytes (CAAs). Radioligands directed at the altered processes, the ECM, and the cellular components of the TME have been developed and evaluated in preclinical and clinical studies for targeted radionuclide imaging and/or therapy. In this review, we provide an overview of the TME targets and their corresponding radioligands. In addition, we discuss what developments are needed to further explore the TME as a target for radionuclide theranostics, with the hopes of stimulating the development of novel TME radioligands with multi-cancer, or in some cases even pan-cancer, application.
Collapse
Affiliation(s)
| | - Simone U Dalm
- Department of Radiology & Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands.
| |
Collapse
|
9
|
Chen KT, Seimbille Y. New Developments in Carbonic Anhydrase IX-Targeted Fluorescence and Nuclear Imaging Agents. Int J Mol Sci 2022; 23:ijms23116125. [PMID: 35682802 PMCID: PMC9181387 DOI: 10.3390/ijms23116125] [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: 05/05/2022] [Revised: 05/27/2022] [Accepted: 05/27/2022] [Indexed: 02/04/2023] Open
Abstract
Carbonic anhydrase IX (CAIX) is a tumor-specific and hypoxia-induced biomarker for the molecular imaging of solid malignancies. The nuclear- and optical-imaging of CAIX-expressing tumors have received great attention due to their potential for clinical applications. Nuclear imaging is a powerful tool for the non-invasive diagnosis of primary and metastatic CAIX-positive tumors and for the assessment of responses to antineoplastic treatment. Intraoperative optical fluorescence imaging provides improved visualization for surgeons to increase the discrimination of tumor lesions, allowing for safer surgical treatment. Over the past decades, many CAIX-targeted molecular imaging probes, based on monoclonal antibodies, antibody fragments, peptides, and small molecules, have been reported. In this review, we outline the recent development of CAIX-targeted probes for single-photon emission computerized tomography (SPECT), positron emission tomography (PET), and near-infrared fluorescence imaging (NIRF), and we discuss issues yet to be addressed.
Collapse
Affiliation(s)
- Kuo-Ting Chen
- Department of Chemistry, National Dong Hwa University, Hualien 974301, Taiwan
- Correspondence: ; Tel.: +886-3-8903603
| | - Yann Seimbille
- Department of Radiology and Nuclear Medicine, Erasmus MC, University Medical Center Rotterdam, Wytemaweg 80, 3015 CN Rotterdam, The Netherlands;
- Erasmus MC Cancer Institute, Erasmus University Medical Center, Doctor Molewaterplein 40, 3015 GD Rotterdam, The Netherlands
| |
Collapse
|
10
|
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
|
11
|
van Lith SAM, Huizing FJ, Franssen GM, Hoeben BAW, Lok J, Doulkeridou S, Boerman OC, Gotthardt M, van Bergen En Henegouwen PMP, Bussink J, Heskamp S. Novel VHH-Based Tracers with Variable Plasma Half-Lives for Imaging of CAIX-Expressing Hypoxic Tumor Cells. Mol Pharm 2022; 19:3511-3520. [PMID: 35044182 PMCID: PMC9533306 DOI: 10.1021/acs.molpharmaceut.1c00841] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
![]()
Hypoxic areas are
present in the majority of solid tumors, and
hypoxia is associated with resistance to therapies and poor outcomes.
A transmembrane protein that is upregulated by tumor cells that have
adapted to hypoxic conditions is carbonic anhydrase IX (CAIX). Therefore,
noninvasive imaging of CAIX could be of prognostic value, and it could
steer treatment strategies. The aim of this study was to compare variants
of CAIX-binding VHH B9, with and without a C-terminal albumin-binding
domain with varying affinity (ABDlow and ABDhigh), for SPECT imaging of CAIX expression. The binding affinity and
internalization of the various B9-variants were analyzed using SK-RC-52
cells. Biodistribution studies were performed in mice with subcutaneous
SCCNij153 human head and neck cancer xenografts. Tracer uptake was
determined by ex vivo radioactivity counting and
visualized by SPECT/CT imaging. Furthermore, autoradiography images
of tumor sections were spatially correlated with CAIX immunohistochemistry.
B9-variants demonstrated a similar moderate affinity for CAIX in vitro. Maximal tumor uptake and acceptable tumor-to-blood
ratios were found in the SCCNij153 model at 4 h post injection for
[111In]In-DTPA-B9 (0.51 ± 0.08%ID/g and 8.1 ±
0.85, respectively), 24 h post injection for [111In]In-DTPA-B9-ABDlow (2.39 ± 0.44%ID/g and 3.66 ± 0.81, respectively)
and at 72 h post injection for [111In]In-DTPA-B9-ABDhigh (8.7 ± 1.34%ID/g and 2.43 ± 0.15, respectively). An excess of unlabeled monoclonal anti-CAIX antibody efficiently
inhibited tumor uptake of [111In]In-DTPA-B9, while only
a partial reduction of [111In]In-DTPA-B9-ABDlow and [111In]In-DTPA-B9-ABDhigh uptake was found.
Immunohistochemistry and autoradiography images showed colocalization
of all B9-variants with CAIX expression; however, [111In]In-DTPA-B9-ABDlow and [111In]In-DTPA-B9-ABDhigh also
accumulated in non-CAIX expressing regions. Tumor uptake of [111In]In-DTPA-B9-ABDlow and [111In]In-DTPA-B9-ABDhigh, but not of [111In]In-DTPA-B9, could be visualized
with SPECT/CT imaging. In conclusion, [111In]In-DTPA-B9
has a high affinity to CAIX and shows specific targeting to CAIX in
head and neck cancer xenografts. The addition of ABD prolonged plasma
half-life, increased tumor uptake, and enabled SPECT/CT imaging. This
uptake was, however, partly CAIX- independent, precluding the ABD-tracers
for use in hypoxia quantification in this tumor type.
Collapse
Affiliation(s)
- Sanne A M van Lith
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen 6500 HB, The Netherlands
| | - Fokko J Huizing
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen 6500 HB, The Netherlands.,Department of Radiation Oncology, Radboud University Medical Center, Nijmegen 6500 HB, The Netherlands
| | - Gerben M Franssen
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen 6500 HB, The Netherlands
| | - Bianca A W Hoeben
- Department of Radiation Oncology, Radboud University Medical Center, Nijmegen 6500 HB, The Netherlands.,Department of Radiation Oncology, University Medical Center Utrecht, Utrecht 3508 GA, The Netherlands
| | - Jasper Lok
- Department of Radiation Oncology, Radboud University Medical Center, Nijmegen 6500 HB, The Netherlands
| | - Sofia Doulkeridou
- Department of Cell Biology, University of Utrecht, Utrecht, 3584 GE, The Netherlands
| | - Otto C Boerman
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen 6500 HB, The Netherlands
| | - Martin Gotthardt
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen 6500 HB, The Netherlands
| | | | - Johan Bussink
- Department of Radiation Oncology, Radboud University Medical Center, Nijmegen 6500 HB, The Netherlands
| | - Sandra Heskamp
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen 6500 HB, The Netherlands
| |
Collapse
|
12
|
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
|
13
|
Boreel DF, Span PN, Heskamp S, Adema GJ, Bussink J. Targeting Oxidative Phosphorylation to Increase the Efficacy of Radio- and Immune-Combination Therapy. Clin Cancer Res 2021; 27:2970-2978. [PMID: 33419779 DOI: 10.1158/1078-0432.ccr-20-3913] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Revised: 11/25/2020] [Accepted: 01/06/2021] [Indexed: 11/16/2022]
Abstract
As tumors grow, they upregulate glycolytic and oxidative metabolism to support their increased and altered energetic demands. These metabolic changes have major effects on the tumor microenvironment. One of the properties leading to this aberrant metabolism is hypoxia, which occurs when tumors outgrow their often-chaotic vasculature. This scarcity of oxygen is known to induce radioresistance but can also have a disrupting effect on the antitumor immune response. Hypoxia inhibits immune effector cell function, while immune cells with a more suppressing phenotype become more active. Therefore, hypoxia strongly affects the efficacy of both radiotherapy and immunotherapy, as well as this therapy combination. Inhibition of oxidative phosphorylation (OXPHOS) is gaining interest for its ability to combat tumor hypoxia, and there are strong indications that this results in a reactivation of the immune response. This strategy decreases oxygen consumption, leading to better oxygenation of hypoxic tumor areas and eventually an increase in immunogenic cell death induced by radio-immunotherapy combinations. Promising preclinical improvements in radio- and immunotherapy efficacy have been observed by the hypoxia-reducing effect of OXPHOS inhibitors and several compounds are currently in clinical trials for their anticancer properties. Here, we will review the pharmacologic attenuation of tumor hypoxia using OXPHOS inhibitors, with emphasis on their impact on the intrinsic antitumor immune response and how this affects the efficacy of (combined) radio- and immunotherapy.
Collapse
Affiliation(s)
- Daan F Boreel
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, the Netherlands. .,Department of Medical Imaging, Nuclear Medicine, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Paul N Span
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Sandra Heskamp
- Department of Medical Imaging, Nuclear Medicine, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Gosse J Adema
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Johan Bussink
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, the Netherlands
| |
Collapse
|
14
|
Shukla M, Forghani R, Agarwal M. Patient-Centric Head and Neck Cancer Radiation Therapy: Role of Advanced Imaging. Neuroimaging Clin N Am 2020; 30:341-357. [PMID: 32600635 DOI: 10.1016/j.nic.2020.04.005] [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: 12/24/2022]
Abstract
The traditional 'one-size-fits-all' approach to H&N cancer therapy is archaic. Advanced imaging can identify radioresistant areas by using biomarkers that detect tumor hypoxia, hypercellularity etc. Highly conformal radiotherapy can target resistant areas with precision. The critical information that can be gleaned about tumor biology from these advanced imaging modalities facilitates individualized radiotherapy. The tumor imaging world is pushing its boundaries. Molecular imaging can now detect protein expression and genotypic variations across tumors that can be exploited for tailoring treatment. The exploding field of radiomics and radiogenomics extracts quantitative, biologic and genetic information and further expands the scope of personalized therapy.
Collapse
Affiliation(s)
- Monica Shukla
- Department of Radiation Oncology, Froedtert and Medical College of Wisconsin, 9200 W. Wisconsin Avenue, Milwaukee, WI 53226, USA
| | - Reza Forghani
- Augmented Intelligence & Precision Health Laboratory, Department of Radiology, Research Institute of McGill University Health Centre, 1001 Decarie Boulevard, Montreal, Quebec H4A 3J1, Canada
| | - Mohit Agarwal
- Department of Radiology, Section of Neuroradiology, Froedtert and Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA.
| |
Collapse
|
15
|
Janoniene A, Petrikaite V. In Search of Advanced Tumor Diagnostics and Treatment: Achievements and Perspectives of Carbonic Anhydrase IX Targeted Delivery. Mol Pharm 2020; 17:1800-1815. [PMID: 32374612 DOI: 10.1021/acs.molpharmaceut.0c00180] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The research of how cells sense and adapt the oxygen deficiency has been recognized as worth winning a Nobel Prize in 2019. Understanding hypoxia-driven molecular machinery paved a path for novel strategies in fighting hypoxia-related diseases including cancer. The oxygen depletion inside the tumor provokes HIF-1 dependent gene and protein expression which helps the tumor to survive. For this reason, tumor related molecules are in the spotlight for scientists developing anticancer agents. One such target is carbonic anhydrase IX (CA IX)-a protein located on the outer cell membrane of most hypoxic tumor cells. This offers the opportunity to exploit it as a target for delivery of cytotoxic drugs, dyes, or radioisotopes to cancer cells. Therefore, researchers investigate CA IX specific small molecules and antibodies as tumor-targeting moieties in nanosystems and conjugates which are expected to overcome the limitations of some existing diagnostic and treatment strategies. This review covers the vast majority of CA IX-targeted systems (nanoparticle and conjugate based) for both therapeutic and imaging purposes published up to now. Furthermore, it shows their stage of development and gives an assessment of their clinical translation possibilities.
Collapse
Affiliation(s)
- Agne Janoniene
- Vilnius University Life Science Center, Institute of Biotechnology, LT-10257 Vilnius, Lithuania
| | - Vilma Petrikaite
- Vilnius University Life Science Center, Institute of Biotechnology, LT-10257 Vilnius, Lithuania.,Lithuanian University of Health Sciences, Institute of Cardiology, LT-50162 Kaunas, Lithuania
| |
Collapse
|
16
|
Huizing FJ, Garousi J, Lok J, Franssen G, Hoeben BAW, Frejd FY, Boerman OC, Bussink J, Tolmachev V, Heskamp S. CAIX-targeting radiotracers for hypoxia imaging in head and neck cancer models. Sci Rep 2019; 9:18898. [PMID: 31827111 PMCID: PMC6906415 DOI: 10.1038/s41598-019-54824-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 11/12/2019] [Indexed: 12/13/2022] Open
Abstract
Hypoxia-induced carbonic anhydrase IX (CAIX) expression is a prognostic marker in solid tumors. In recent years many radiotracers have been developed, but a fair comparison of these compounds is not possible because of the diversity in tumor models and other experimental parameters. In this study we performed a direct in vivo comparison of three promising CAIX targeting radiotracers in xenografted head and neck cancer models. The biodistribution of [111In]In-DOTA-ZCAIX:2 was directly compared with [111In]In-DTPA-G250-F(ab′)2 and [111In] In-DTPA-G250 in female BALB/C nu/nu mice bearing two HNSCC xenografts with different levels of CAIX expression. In vivo biodistribution was quantified by means of microSPECT/CT scans and ex vivo biodistribution was determined with the use of a γ-counter. Tumors were snap frozen and sections were stained for CAIX expression, vessels, hypoxia (pimonidazole) and tumor blood perfusion. Tracer uptake was significantly higher in SSCNij153 tumors compared to SCCNij185 tumors for [111In]In-DOTA-HE3-ZCAIX:2: 0.32 ± 0.03 versus 0.18 ± 0.01%ID/g,(p = 0.003) 4 h p.i., for [111In]In-DTPA-girentuximab-F(ab′)2: 3.0 ± 0.5%ID/g and 1.2 ± 0.1%ID/g (p = 0.03), 24 h p.i. and for [111In]In-DTPA-girentuximab: 30 ± 2.1%ID/g and 7.0 ± 1.0%ID/g (p = 0.0002) 72 h p.i. SPECT imaging with both [111In]In-DTPA-girentuximab-F(ab′)2 and [111In]In-DTPA-girentuximab showed a clear difference in tracer distribution between the two tumor models. The whole IgG, i.e. [111In]In-DTPA-girentuximab, showed the highest tumor-to-muscle ratio. We showed that different CAIX-targeting radiotracers can discriminate a low CAIX-expressing tumor from a high CAIX-expressing head and neck cancer xenografts model. In these hypoxic head and neck xenograft models [111In]In-DTPA-girentuximab showed the most promising results.
Collapse
Affiliation(s)
- Fokko J Huizing
- Departments of Radiation Oncology, Radboud University Medical Center, Nijmegen, The Netherlands.
| | - Javad Garousi
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Jasper Lok
- Departments of Radiation Oncology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Gerben Franssen
- Departments of Radiology and Nuclear Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Bianca A W Hoeben
- Departments of Radiation Oncology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Fredrik Y Frejd
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden.,Affibody AB, Solna, Sweden
| | - Otto C Boerman
- Departments of Radiology and Nuclear Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Johan Bussink
- Departments of Radiation Oncology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Vladimir Tolmachev
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Sandra Heskamp
- Departments of Radiology and Nuclear Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
17
|
Garousi J, Huizing FJ, Vorobyeva A, Mitran B, Andersson KG, Leitao CD, Frejd FY, Löfblom J, Bussink J, Orlova A, Heskamp S, Tolmachev V. Comparative evaluation of affibody- and antibody fragments-based CAIX imaging probes in mice bearing renal cell carcinoma xenografts. Sci Rep 2019; 9:14907. [PMID: 31624303 PMCID: PMC6797765 DOI: 10.1038/s41598-019-51445-w] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 09/25/2019] [Indexed: 12/11/2022] Open
Abstract
Carbonic anhydrase IX (CAIX) is a cancer-associated molecular target for several classes of therapeutics. CAIX is overexpressed in a large fraction of renal cell carcinomas (RCC). Radionuclide molecular imaging of CAIX-expression might offer a non-invasive methodology for stratification of patients with disseminated RCC for CAIX-targeting therapeutics. Radiolabeled monoclonal antibodies and their fragments are actively investigated for imaging of CAIX expression. Promising alternatives are small non-immunoglobulin scaffold proteins, such as affibody molecules. A CAIX-targeting affibody ZCAIX:2 was re-designed with the aim to decrease off-target interactions and increase imaging contrast. The new tracer, DOTA-HE3-ZCAIX:2, was labeled with 111In and characterized in vitro. Tumor-targeting properties of [111In]In-DOTA-HE3-ZCAIX:2 were compared head-to-head with properties of the parental variant, [99mTc]Tc(CO)3-HE3-ZCAIX:2, and the most promising antibody fragment-based tracer, [111In]In-DTPA-G250(Fab’)2, in the same batch of nude mice bearing CAIX-expressing RCC xenografts. Compared to the 99mTc-labeled parental variant, [111In]In-DOTA-HE3-ZCAIX:2 provides significantly higher tumor-to-lung, tumor-to-bone and tumor-to-liver ratios, which is essential for imaging of CAIX expression in the major metastatic sites of RCC. [111In]In-DOTA-HE3-ZCAIX:2 offers significantly higher tumor-to-organ ratios compared with [111In]In-G250(Fab’)2. In conclusion, [111In]In-DOTA-HE3-ZCAIX:2 can be considered as a highly promising tracer for imaging of CAIX expression in RCC metastases based on our results and literature data.
Collapse
Affiliation(s)
- Javad Garousi
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Fokko J Huizing
- Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Anzhelika Vorobyeva
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Bogdan Mitran
- Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Ken G Andersson
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Charles Dahlsson Leitao
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Fredrik Y Frejd
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - John Löfblom
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Johan Bussink
- Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Anna Orlova
- Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Sandra Heskamp
- Department of Radiology and Nuclear medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Vladimir Tolmachev
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden.
| |
Collapse
|