1
|
Pangua C, Espuelas S, Simón JA, Álvarez S, Martínez-Ohárriz C, Collantes M, Peñuelas I, Calvo A, Irache JM. Enhancing bevacizumab efficacy in a colorectal tumor mice model using dextran-coated albumin nanoparticles. Drug Deliv Transl Res 2024:10.1007/s13346-024-01734-3. [PMID: 39455507 DOI: 10.1007/s13346-024-01734-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/18/2024] [Indexed: 10/28/2024]
Abstract
Bevacizumab is a monoclonal antibody (mAb) that prevents the growth of new blood vessels and is currently employed in the treatment of colorectal cancer (CRC). However, like other mAb, bevacizumab shows a limited penetration in the tumors, hampering their effectiveness and inducing adverse reactions. The aim of this work was to design and evaluate albumin-based nanoparticles, coated with dextran, as carriers for bevacizumab in order to promote its accumulation in the tumor and, thus, improve its antiangiogenic activity. These nanoparticles (B-NP-DEX50) displayed a mean size of about 250 nm and a payload of about 110 µg/mg. In a CRC mice model, these nanoparticles significantly reduced tumor growth and increased tumor doubling time, tumor necrosis and apoptosis more effectively than free bevacizumab. At the end of study, bevacizumab plasma levels were higher in the free drug group, while tumor levels were higher in the B-NP-DEX50 group (2.5-time higher). In line with this, the biodistribution study revealed that nanoparticles accumulated in the tumor core, potentially improving therapeutic efficacy while reducing systemic exposure. In summary, B-NP-DEX can be an adequate alternative to improve the therapeutic efficiency of biologically active molecules, offering a more specific biodistribution to the site of action.
Collapse
Affiliation(s)
- Cristina Pangua
- NANO-VAC Research Group, Department of Pharmaceutical Sciences, School of Pharmacy and Nutrition, University of Navarra, C/ Irunlarrea 1, Pamplona, 31008, Spain
| | - Socorro Espuelas
- NANO-VAC Research Group, Department of Pharmaceutical Sciences, School of Pharmacy and Nutrition, University of Navarra, C/ Irunlarrea 1, Pamplona, 31008, Spain
- Institute for Health Research (IdiSNA), Pamplona, 31008, Spain
| | - Jon Ander Simón
- Program in Solid Tumors, CIMA of the University of Navarra, Pamplona, 31008, Spain
| | - Samuel Álvarez
- NANO-VAC Research Group, Department of Pharmaceutical Sciences, School of Pharmacy and Nutrition, University of Navarra, C/ Irunlarrea 1, Pamplona, 31008, Spain
| | | | - María Collantes
- Radiopharmacy Unit, Clinica Universidad de Navarra, Pamplona, 31008, Spain
- Institute for Health Research (IdiSNA), Pamplona, 31008, Spain
| | - Iván Peñuelas
- Radiopharmacy Unit, Clinica Universidad de Navarra, Pamplona, 31008, Spain
- Translational Molecular Imaging Unit (UNIMTRA), Department of Nuclear Medicine, Clinica Universidad de Navarra, Pamplona, 31008, Spain
- Institute for Health Research (IdiSNA), Pamplona, 31008, Spain
| | - Alfonso Calvo
- Program in Solid Tumors, CIMA of the University of Navarra, Pamplona, 31008, Spain
- Institute for Health Research (IdiSNA), Pamplona, 31008, Spain
| | - Juan M Irache
- NANO-VAC Research Group, Department of Pharmaceutical Sciences, School of Pharmacy and Nutrition, University of Navarra, C/ Irunlarrea 1, Pamplona, 31008, Spain.
- Institute for Health Research (IdiSNA), Pamplona, 31008, Spain.
| |
Collapse
|
2
|
Huynh BN, Groendahl AR, Tomic O, Liland KH, Knudtsen IS, Hoebers F, van Elmpt W, Dale E, Malinen E, Futsaether CM. Deep learning with uncertainty estimation for automatic tumor segmentation in PET/CT of head and neck cancers: impact of model complexity, image processing and augmentation. Biomed Phys Eng Express 2024; 10:055038. [PMID: 39127060 DOI: 10.1088/2057-1976/ad6dcd] [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: 04/29/2024] [Accepted: 08/09/2024] [Indexed: 08/12/2024]
Abstract
Objective.Target volumes for radiotherapy are usually contoured manually, which can be time-consuming and prone to inter- and intra-observer variability. Automatic contouring by convolutional neural networks (CNN) can be fast and consistent but may produce unrealistic contours or miss relevant structures. We evaluate approaches for increasing the quality and assessing the uncertainty of CNN-generated contours of head and neck cancers with PET/CT as input.Approach.Two patient cohorts with head and neck squamous cell carcinoma and baseline18F-fluorodeoxyglucose positron emission tomography and computed tomography images (FDG-PET/CT) were collected retrospectively from two centers. The union of manual contours of the gross primary tumor and involved nodes was used to train CNN models for generating automatic contours. The impact of image preprocessing, image augmentation, transfer learning and CNN complexity, architecture, and dimension (2D or 3D) on model performance and generalizability across centers was evaluated. A Monte Carlo dropout technique was used to quantify and visualize the uncertainty of the automatic contours.Main results. CNN models provided contours with good overlap with the manually contoured ground truth (median Dice Similarity Coefficient: 0.75-0.77), consistent with reported inter-observer variations and previous auto-contouring studies. Image augmentation and model dimension, rather than model complexity, architecture, or advanced image preprocessing, had the largest impact on model performance and cross-center generalizability. Transfer learning on a limited number of patients from a separate center increased model generalizability without decreasing model performance on the original training cohort. High model uncertainty was associated with false positive and false negative voxels as well as low Dice coefficients.Significance.High quality automatic contours can be obtained using deep learning architectures that are not overly complex. Uncertainty estimation of the predicted contours shows potential for highlighting regions of the contour requiring manual revision or flagging segmentations requiring manual inspection and intervention.
Collapse
Affiliation(s)
- Bao Ngoc Huynh
- Faculty of Science and Technology, Norwegian University of Life Sciences, Ås, Norway
| | - Aurora Rosvoll Groendahl
- Faculty of Science and Technology, Norwegian University of Life Sciences, Ås, Norway
- Section of Oncology, Vestre Viken Hospital Trust, Drammen, Norway
| | - Oliver Tomic
- Faculty of Science and Technology, Norwegian University of Life Sciences, Ås, Norway
| | - Kristian Hovde Liland
- Faculty of Science and Technology, Norwegian University of Life Sciences, Ås, Norway
| | - Ingerid Skjei Knudtsen
- Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway
| | - Frank Hoebers
- Department of Radiation Oncology (MAASTRO), GROW School for Oncology and Reproduction, Maastricht, Netherlands
| | - Wouter van Elmpt
- Department of Radiation Oncology (MAASTRO), GROW School for Oncology and Reproduction, Maastricht, Netherlands
| | - Einar Dale
- Department of Oncology, Oslo University Hospital, Oslo, Norway
| | - Eirik Malinen
- Department of Medical Physics, Oslo University Hospital, Oslo, Norway
- Department of Physics, University of Oslo, Oslo, Norway
| | | |
Collapse
|
3
|
de Jong D, Desperito E, Al Feghali KA, Dercle L, Seban RD, Das JP, Ma H, Sajan A, Braumuller B, Prendergast C, Liou C, Deng A, Roa T, Yeh R, Girard A, Salvatore MM, Capaccione KM. Advances in PET/CT Imaging for Breast Cancer. J Clin Med 2023; 12:4537. [PMID: 37445572 PMCID: PMC10342839 DOI: 10.3390/jcm12134537] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Revised: 06/26/2023] [Accepted: 06/30/2023] [Indexed: 07/15/2023] Open
Abstract
One out of eight women will be affected by breast cancer during her lifetime. Imaging plays a key role in breast cancer detection and management, providing physicians with information about tumor location, heterogeneity, and dissemination. In this review, we describe the latest advances in PET/CT imaging of breast cancer, including novel applications of 18F-FDG PET/CT and the development and testing of new agents for primary and metastatic breast tumor imaging and therapy. Ultimately, these radiopharmaceuticals may guide personalized approaches to optimize treatment based on the patient's specific tumor profile, and may become a new standard of care. In addition, they may enhance the assessment of treatment efficacy and lead to improved outcomes for patients with a breast cancer diagnosis.
Collapse
Affiliation(s)
- Dorine de Jong
- Center for Cell Engineering, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA;
| | - Elise Desperito
- Department of Radiology, Columbia University Irving Medical Center, New York, NY 10032, USA; (E.D.); (L.D.); (H.M.); (A.S.); (B.B.); (C.P.); (C.L.); (T.R.); (M.M.S.)
| | | | - Laurent Dercle
- Department of Radiology, Columbia University Irving Medical Center, New York, NY 10032, USA; (E.D.); (L.D.); (H.M.); (A.S.); (B.B.); (C.P.); (C.L.); (T.R.); (M.M.S.)
| | - Romain-David Seban
- Department of Nuclear Medicine and Endocrine Oncology, Institut Curie, 92210 Saint-Cloud, France;
- Laboratory of Translational Imaging in Oncology, Paris Sciences et Lettres (PSL) Research University, Institut Curie, 91401 Orsay, France
| | - Jeeban P. Das
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; (J.P.D.); (R.Y.)
| | - Hong Ma
- Department of Radiology, Columbia University Irving Medical Center, New York, NY 10032, USA; (E.D.); (L.D.); (H.M.); (A.S.); (B.B.); (C.P.); (C.L.); (T.R.); (M.M.S.)
| | - Abin Sajan
- Department of Radiology, Columbia University Irving Medical Center, New York, NY 10032, USA; (E.D.); (L.D.); (H.M.); (A.S.); (B.B.); (C.P.); (C.L.); (T.R.); (M.M.S.)
| | - Brian Braumuller
- Department of Radiology, Columbia University Irving Medical Center, New York, NY 10032, USA; (E.D.); (L.D.); (H.M.); (A.S.); (B.B.); (C.P.); (C.L.); (T.R.); (M.M.S.)
| | - Conor Prendergast
- Department of Radiology, Columbia University Irving Medical Center, New York, NY 10032, USA; (E.D.); (L.D.); (H.M.); (A.S.); (B.B.); (C.P.); (C.L.); (T.R.); (M.M.S.)
| | - Connie Liou
- Department of Radiology, Columbia University Irving Medical Center, New York, NY 10032, USA; (E.D.); (L.D.); (H.M.); (A.S.); (B.B.); (C.P.); (C.L.); (T.R.); (M.M.S.)
| | - Aileen Deng
- Department of Hematology and Oncology, Novant Health, 170 Medical Park Road, Mooresville, NC 28117, USA;
| | - Tina Roa
- Department of Radiology, Columbia University Irving Medical Center, New York, NY 10032, USA; (E.D.); (L.D.); (H.M.); (A.S.); (B.B.); (C.P.); (C.L.); (T.R.); (M.M.S.)
| | - Randy Yeh
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; (J.P.D.); (R.Y.)
| | - Antoine Girard
- Department of Nuclear Medicine, Centre Eugène Marquis, Université Rennes 1, 35000 Rennes, France;
| | - Mary M. Salvatore
- Department of Radiology, Columbia University Irving Medical Center, New York, NY 10032, USA; (E.D.); (L.D.); (H.M.); (A.S.); (B.B.); (C.P.); (C.L.); (T.R.); (M.M.S.)
| | - Kathleen M. Capaccione
- Department of Radiology, Columbia University Irving Medical Center, New York, NY 10032, USA; (E.D.); (L.D.); (H.M.); (A.S.); (B.B.); (C.P.); (C.L.); (T.R.); (M.M.S.)
| |
Collapse
|
4
|
Recent Development of Radiofluorination of Boron Agents for Boron Neutron Capture Therapy of Tumor: Creation of 18F-Labeled C-F and B-F Linkages. Pharmaceuticals (Basel) 2023; 16:ph16010093. [PMID: 36678590 PMCID: PMC9866017 DOI: 10.3390/ph16010093] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 12/27/2022] [Accepted: 01/03/2023] [Indexed: 01/11/2023] Open
Abstract
Boron neutron capture therapy (BNCT) is a binary therapeutic technique employing a boron agent to be delivered to the tumor site followed by the irradiation of neutrons. Biofunctional molecules/nanoparticles labeled with F-18 can provide an initial pharmacokinetic profile of patients to guide the subsequent treatment planning procedure of BNCT. Borono phenylalanine (BPA), recognized by the l-type amino acid transporter, can cross the blood-brain barrier and be accumulated in gliomas. The radiofluoro BNCT agents are reviewed by considering (1) less cytotoxicity, (2) diagnosing and therapeutic purposes, (3) aqueous solubility and extraction route, as well as (4), the trifluoroborate effect. A trifluoroborate-containing amino acid such as fluoroboronotyrosine (FBY) represents an example with both functionalities of imaging and therapeutics. Comparing with the insignificant cytotoxicity of clinical BPA with IC50 > 500 μM, FBY also shows minute toxicity with IC50 > 500 μM. [18F]FBY is a potential diagnostic agent for its tumor to normal accumulation (T/N) ratio, which ranges from 2.3 to 24.5 from positron emission tomography, whereas the T/N ratio of FBPA is greater than 2.5. Additionally, in serving as a BNCT therapeutic agent, the boron concentration of FBY accumulated in gliomas remains uncertain. The solubility of 3-BPA is better than that of BPA, as evidenced by the cerebral dose of 3.4%ID/g vs. 2.2%ID/g, respectively. While the extraction route of d-BPA differs from that of BPA, an impressive T/N ratio of 6.9 vs. 1.5 is noted. [18F]FBPA, the most common clinical boron agent, facilitates the application of BPA in clinical BNCT. In addition to [18F]FBY, [18F] trifluoroborated nucleoside analog obtained through 1,3-dipolar cycloaddition shows marked tumoral uptake of 1.5%ID/g. Other examples using electrophilic and nucleophilic fluorination on the boron compounds are also reviewed, including diboronopinacolone phenylalanine and nonsteroidal anti-inflammatory agents.
Collapse
|
5
|
Dobre EG, Surcel M, Constantin C, Ilie MA, Caruntu A, Caruntu C, Neagu M. Skin Cancer Pathobiology at a Glance: A Focus on Imaging Techniques and Their Potential for Improved Diagnosis and Surveillance in Clinical Cohorts. Int J Mol Sci 2023; 24:1079. [PMID: 36674595 PMCID: PMC9866322 DOI: 10.3390/ijms24021079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Revised: 01/02/2023] [Accepted: 01/03/2023] [Indexed: 01/08/2023] Open
Abstract
Early diagnosis is essential for completely eradicating skin cancer and maximizing patients' clinical benefits. Emerging optical imaging modalities such as reflectance confocal microscopy (RCM), optical coherence tomography (OCT), magnetic resonance imaging (MRI), near-infrared (NIR) bioimaging, positron emission tomography (PET), and their combinations provide non-invasive imaging data that may help in the early detection of cutaneous tumors and surgical planning. Hence, they seem appropriate for observing dynamic processes such as blood flow, immune cell activation, and tumor energy metabolism, which may be relevant for disease evolution. This review discusses the latest technological and methodological advances in imaging techniques that may be applied for skin cancer detection and monitoring. In the first instance, we will describe the principle and prospective clinical applications of the most commonly used imaging techniques, highlighting the challenges and opportunities of their implementation in the clinical setting. We will also highlight how imaging techniques may complement the molecular and histological approaches in sharpening the non-invasive skin characterization, laying the ground for more personalized approaches in skin cancer patients.
Collapse
Affiliation(s)
- Elena-Georgiana Dobre
- Faculty of Biology, University of Bucharest, Splaiul Independentei 91-95, 050095 Bucharest, Romania
| | - Mihaela Surcel
- Immunology Department, “Victor Babes” National Institute of Pathology, 050096 Bucharest, Romania
| | - Carolina Constantin
- Immunology Department, “Victor Babes” National Institute of Pathology, 050096 Bucharest, Romania
- Department of Pathology, Colentina University Hospital, 020125 Bucharest, Romania
| | | | - Ana Caruntu
- Department of Oral and Maxillofacial Surgery, “Carol Davila” Central Military Emergency Hospital, 010825 Bucharest, Romania
- Department of Oral and Maxillofacial Surgery, Faculty of Dental Medicine, “Titu Maiorescu” University, 031593 Bucharest, Romania
| | - Constantin Caruntu
- Department of Physiology, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Department of Dermatology, “Prof. N.C. Paulescu” National Institute of Diabetes, Nutrition and Metabolic Diseases, 011233 Bucharest, Romania
| | - Monica Neagu
- Faculty of Biology, University of Bucharest, Splaiul Independentei 91-95, 050095 Bucharest, Romania
- Immunology Department, “Victor Babes” National Institute of Pathology, 050096 Bucharest, Romania
- Department of Pathology, Colentina University Hospital, 020125 Bucharest, Romania
| |
Collapse
|
6
|
Godet I, Doctorman S, Wu F, Gilkes DM. Detection of Hypoxia in Cancer Models: Significance, Challenges, and Advances. Cells 2022; 11:686. [PMID: 35203334 PMCID: PMC8869817 DOI: 10.3390/cells11040686] [Citation(s) in RCA: 51] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 02/07/2022] [Accepted: 02/09/2022] [Indexed: 02/06/2023] Open
Abstract
The rapid proliferation of cancer cells combined with deficient vessels cause regions of nutrient and O2 deprivation in solid tumors. Some cancer cells can adapt to these extreme hypoxic conditions and persist to promote cancer progression. Intratumoral hypoxia has been consistently associated with a worse patient prognosis. In vitro, 3D models of spheroids or organoids can recapitulate spontaneous O2 gradients in solid tumors. Likewise, in vivo murine models of cancer reproduce the physiological levels of hypoxia that have been measured in human tumors. Given the potential clinical importance of hypoxia in cancer progression, there is an increasing need to design methods to measure O2 concentrations. O2 levels can be directly measured with needle-type probes, both optical and electrochemical. Alternatively, indirect, noninvasive approaches have been optimized, and include immunolabeling endogenous or exogenous markers. Fluorescent, phosphorescent, and luminescent reporters have also been employed experimentally to provide dynamic measurements of O2 in live cells or tumors. In medical imaging, modalities such as MRI and PET are often the method of choice. This review provides a comparative overview of the main methods utilized to detect hypoxia in cell culture and preclinical models of cancer.
Collapse
Affiliation(s)
- Inês Godet
- The Sidney Kimmel Comprehensive Cancer Center, Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA;
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD 21218, USA; (S.D.); (F.W.)
- Johns Hopkins Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - Steven Doctorman
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD 21218, USA; (S.D.); (F.W.)
| | - Fan Wu
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD 21218, USA; (S.D.); (F.W.)
| | - Daniele M. Gilkes
- The Sidney Kimmel Comprehensive Cancer Center, Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA;
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD 21218, USA; (S.D.); (F.W.)
- Johns Hopkins Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore, MD 21218, USA
- Cellular and Molecular Medicine Program, The Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| |
Collapse
|
7
|
Shuvalov O, Daks A, Fedorova O, Petukhov A, Barlev N. Linking Metabolic Reprogramming, Plasticity and Tumor Progression. Cancers (Basel) 2021; 13:cancers13040762. [PMID: 33673109 PMCID: PMC7917602 DOI: 10.3390/cancers13040762] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 02/03/2021] [Accepted: 02/07/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary In the present review, we discuss the role of metabolic reprogramming which occurs in malignant cells. The process of metabolic reprogramming is also known as one of the “hallmarks of cancer”. Due to several reasons, including the origin of cancer, tumor microenvironment, and the tumor progression stage, metabolic reprogramming can be heterogeneous and dynamic. In this review, we provide evidence that the usage of metabolic drugs is a promising approach to treat cancer. However, because these drugs can damage not only malignant cells but also normal rapidly dividing cells, it is important to understand the exact metabolic changes which are elicited by particular drivers in concrete tissue and are specific for each stage of cancer development, including metastases. Finally, the review highlights new promising targets for the development of new metabolic drugs. Abstract The specific molecular features of cancer cells that distinguish them from the normal ones are denoted as “hallmarks of cancer”. One of the critical hallmarks of cancer is an altered metabolism which provides tumor cells with energy and structural resources necessary for rapid proliferation. The key feature of a cancer-reprogrammed metabolism is its plasticity, allowing cancer cells to better adapt to various conditions and to oppose different therapies. Furthermore, the alterations of metabolic pathways in malignant cells are heterogeneous and are defined by several factors including the tissue of origin, driving mutations, and microenvironment. In the present review, we discuss the key features of metabolic reprogramming and plasticity associated with different stages of tumor, from primary tumors to metastases. We also provide evidence of the successful usage of metabolic drugs in anticancer therapy. Finally, we highlight new promising targets for the development of new metabolic drugs.
Collapse
Affiliation(s)
- Oleg Shuvalov
- Institute of Cytology RAS, 194064 St-Petersburg, Russia; (O.S.); (A.D.); (O.F.); (A.P.)
| | - Alexandra Daks
- Institute of Cytology RAS, 194064 St-Petersburg, Russia; (O.S.); (A.D.); (O.F.); (A.P.)
| | - Olga Fedorova
- Institute of Cytology RAS, 194064 St-Petersburg, Russia; (O.S.); (A.D.); (O.F.); (A.P.)
| | - Alexey Petukhov
- Institute of Cytology RAS, 194064 St-Petersburg, Russia; (O.S.); (A.D.); (O.F.); (A.P.)
- Almazov National Medical Research Center, 197341 St-Petersburg, Russia
| | - Nickolai Barlev
- Institute of Cytology RAS, 194064 St-Petersburg, Russia; (O.S.); (A.D.); (O.F.); (A.P.)
- MIPT, 141701 Dolgoprudny, Moscow Region, Russia
- Orekhovich IBMC, 119435 Moscow, Russia
- Correspondence: ; Tel.: +7-812-297-4519
| |
Collapse
|
8
|
Sadeghzadeh M, Wenzel B, Gündel D, Deuther-Conrad W, Toussaint M, Moldovan RP, Fischer S, Ludwig FA, Teodoro R, Jonnalagadda S, Jonnalagadda SK, Schüürmann G, Mereddy VR, Drewes LR, Brust P. Development of Novel Analogs of the Monocarboxylate Transporter Ligand FACH and Biological Validation of One Potential Radiotracer for Positron Emission Tomography (PET) Imaging. Molecules 2020; 25:molecules25102309. [PMID: 32423056 PMCID: PMC7288138 DOI: 10.3390/molecules25102309] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 05/08/2020] [Accepted: 05/11/2020] [Indexed: 12/14/2022] Open
Abstract
Monocarboxylate transporters 1-4 (MCT1-4) are involved in several metabolism-related diseases, especially cancer, providing the chance to be considered as relevant targets for diagnosis and therapy. [18F]FACH was recently developed and showed very promising preclinical results as a potential positron emission tomography (PET) radiotracer for imaging of MCTs. Given that [18F]FACH did not show high blood-brain barrier permeability, the current work is aimed to investigate whether more lipophilic analogs of FACH could improve brain uptake for imaging of gliomas, while retaining binding to MCTs. The 2-fluoropyridinyl-substituted analogs 1 and 2 were synthesized and their MCT1 inhibition was estimated by [14C]lactate uptake assay on rat brain endothelial-4 (RBE4) cells. While compounds 1 and 2 showed lower MCT1 inhibitory potencies than FACH (IC50 = 11 nM) by factors of 11 and 25, respectively, 1 (IC50 = 118 nM) could still be a suitable PET candidate. Therefore, 1 was selected for radiosynthesis of [18F]1 and subsequent biological evaluation for imaging of the MCT expression in mouse brain. Regarding lipophilicity, the experimental log D7.4 result for [18F]1 agrees pretty well with its predicted value. In vivo and in vitro studies revealed high uptake of the new radiotracer in kidney and other peripheral MCT-expressing organs together with significant reduction by using specific MCT1 inhibitor α-cyano-4-hydroxycinnamic acid. Despite a higher lipophilicity of [18F]1 compared to [18F]FACH, the in vivo brain uptake of [18F]1 was in a similar range, which is reflected by calculated BBB permeabilities as well through similar transport rates by MCTs on RBE4 cells. Further investigation is needed to clarify the MCT-mediated transport mechanism of these radiotracers in brain.
Collapse
Affiliation(s)
- Masoud Sadeghzadeh
- Department of Neuroradiopharmaceuticals, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Permoserstraße 15, 04318 Leipzig, Germany; (B.W.); (D.G.); (W.D.-C.); (M.T.); (R.-P.M.); (S.F.); (F.-A.L.); (R.T.); (P.B.)
- Correspondence: ; Tel.: +49-341-2341794630; Fax: +49-341-2341794699
| | - Barbara Wenzel
- Department of Neuroradiopharmaceuticals, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Permoserstraße 15, 04318 Leipzig, Germany; (B.W.); (D.G.); (W.D.-C.); (M.T.); (R.-P.M.); (S.F.); (F.-A.L.); (R.T.); (P.B.)
| | - Daniel Gündel
- Department of Neuroradiopharmaceuticals, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Permoserstraße 15, 04318 Leipzig, Germany; (B.W.); (D.G.); (W.D.-C.); (M.T.); (R.-P.M.); (S.F.); (F.-A.L.); (R.T.); (P.B.)
| | - Winnie Deuther-Conrad
- Department of Neuroradiopharmaceuticals, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Permoserstraße 15, 04318 Leipzig, Germany; (B.W.); (D.G.); (W.D.-C.); (M.T.); (R.-P.M.); (S.F.); (F.-A.L.); (R.T.); (P.B.)
| | - Magali Toussaint
- Department of Neuroradiopharmaceuticals, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Permoserstraße 15, 04318 Leipzig, Germany; (B.W.); (D.G.); (W.D.-C.); (M.T.); (R.-P.M.); (S.F.); (F.-A.L.); (R.T.); (P.B.)
| | - Rareş-Petru Moldovan
- Department of Neuroradiopharmaceuticals, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Permoserstraße 15, 04318 Leipzig, Germany; (B.W.); (D.G.); (W.D.-C.); (M.T.); (R.-P.M.); (S.F.); (F.-A.L.); (R.T.); (P.B.)
| | - Steffen Fischer
- Department of Neuroradiopharmaceuticals, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Permoserstraße 15, 04318 Leipzig, Germany; (B.W.); (D.G.); (W.D.-C.); (M.T.); (R.-P.M.); (S.F.); (F.-A.L.); (R.T.); (P.B.)
| | - Friedrich-Alexander Ludwig
- Department of Neuroradiopharmaceuticals, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Permoserstraße 15, 04318 Leipzig, Germany; (B.W.); (D.G.); (W.D.-C.); (M.T.); (R.-P.M.); (S.F.); (F.-A.L.); (R.T.); (P.B.)
| | - Rodrigo Teodoro
- Department of Neuroradiopharmaceuticals, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Permoserstraße 15, 04318 Leipzig, Germany; (B.W.); (D.G.); (W.D.-C.); (M.T.); (R.-P.M.); (S.F.); (F.-A.L.); (R.T.); (P.B.)
| | - Shirisha Jonnalagadda
- Department of Chemistry and Biochemistry, Department of Pharmacy Practice & Pharmaceutical Sciences, University of Minnesota, Duluth, MN 55812, USA; (S.J.); (S.K.J.); (V.R.M.)
| | - Sravan K. Jonnalagadda
- Department of Chemistry and Biochemistry, Department of Pharmacy Practice & Pharmaceutical Sciences, University of Minnesota, Duluth, MN 55812, USA; (S.J.); (S.K.J.); (V.R.M.)
| | - Gerrit Schüürmann
- UFZ Department of Ecological Chemistry, Helmholtz Centre for Environmental Research, Permoserstraße 15, 04318 Leipzig, Germany;
- Institute of Organic Chemistry, Technical University Bergakademie Freiberg, Leipziger Straße 29, 09599 Freiberg, Germany
| | - Venkatram R. Mereddy
- Department of Chemistry and Biochemistry, Department of Pharmacy Practice & Pharmaceutical Sciences, University of Minnesota, Duluth, MN 55812, USA; (S.J.); (S.K.J.); (V.R.M.)
| | - Lester R. Drewes
- Department of Biomedical Sciences, University of Minnesota Medical School Duluth, 251 SMed, 1035 University Drive, Duluth, MN 55812, USA;
| | - Peter Brust
- Department of Neuroradiopharmaceuticals, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Permoserstraße 15, 04318 Leipzig, Germany; (B.W.); (D.G.); (W.D.-C.); (M.T.); (R.-P.M.); (S.F.); (F.-A.L.); (R.T.); (P.B.)
| |
Collapse
|
9
|
Sattler B, Kranz M, Wenzel B, Jain NT, Moldovan RP, Toussaint M, Deuther-Conrad W, Ludwig FA, Teodoro R, Sattler T, Sadeghzadeh M, Sabri O, Brust P. Preclinical Incorporation Dosimetry of [ 18F]FACH-A Novel 18F-Labeled MCT1/MCT4 Lactate Transporter Inhibitor for Imaging Cancer Metabolism with PET. Molecules 2020; 25:E2024. [PMID: 32357571 PMCID: PMC7248880 DOI: 10.3390/molecules25092024] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 04/18/2020] [Accepted: 04/20/2020] [Indexed: 02/08/2023] Open
Abstract
Overexpression of monocarboxylate transporters (MCTs) has been shown for a variety of human cancers (e.g., colon, brain, breast, and kidney) and inhibition resulted in intracellular lactate accumulation, acidosis, and cell death. Thus, MCTs are promising targets to investigate tumor cancer metabolism with positron emission tomography (PET). Here, the organ doses (ODs) and the effective dose (ED) of the first 18F-labeled MCT1/MCT4 inhibitor were estimated in juvenile pigs. Whole-body dosimetry was performed in three piglets (age: ~6 weeks, weight: ~13-15 kg). The animals were anesthetized and subjected to sequential hybrid Positron Emission Tomography and Computed Tomography (PET/CT) up to 5 h after an intravenous (iv) injection of 156 ± 54 MBq [18F]FACH. All relevant organs were defined by volumes of interest. Exponential curves were fitted to the time-activity data. Time and mass scales were adapted to the human order of magnitude and the ODs calculated using the ICRP 89 adult male phantom with OLINDA 2.1. The ED was calculated using tissue weighting factors as published in Publication 103 of the International Commission of Radiation Protection (ICRP103). The highest organ dose was received by the urinary bladder (62.6 ± 28.9 µSv/MBq), followed by the gall bladder (50.4 ± 37.5 µSv/MBq) and the pancreas (30.5 ± 27.3 µSv/MBq). The highest contribution to the ED was by the urinary bladder (2.5 ± 1.1 µSv/MBq), followed by the red marrow (1.7 ± 0.3 µSv/MBq) and the stomach (1.3 ± 0.4 µSv/MBq). According to this preclinical analysis, the ED to humans is 12.4 µSv/MBq when applying the ICRP103 tissue weighting factors. Taking into account that preclinical dosimetry underestimates the dose to humans by up to 40%, the conversion factor applied for estimation of the ED to humans would rise to 20.6 µSv/MBq. In this case, the ED to humans upon an iv application of ~300 MBq [18F]FACH would be about 6.2 mSv. This risk assessment encourages the translation of [18F]FACH into clinical study phases and the further investigation of its potential as a clinical tool for cancer imaging with PET.
Collapse
Affiliation(s)
- Bernhard Sattler
- Department of Nuclear Medicine, University Hospital Leipzig, 04103 Leipzig, Germany
| | - Mathias Kranz
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Department of Neuroradiopharmaceuticals, 04318 Leipzig, Germany
- Tromsø PET Center, University Hospital of North Norway, 9009 Tromsø, Norway
- Nuclear Medicine and Radiation Biology Research Group, The Arctic University of Norway, 9009 Tromsø, Norway
| | - Barbara Wenzel
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Department of Neuroradiopharmaceuticals, 04318 Leipzig, Germany
| | - Nalin T. Jain
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Department of Neuroradiopharmaceuticals, 04318 Leipzig, Germany
| | - Rareş-Petru Moldovan
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Department of Neuroradiopharmaceuticals, 04318 Leipzig, Germany
| | - Magali Toussaint
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Department of Neuroradiopharmaceuticals, 04318 Leipzig, Germany
| | - Winnie Deuther-Conrad
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Department of Neuroradiopharmaceuticals, 04318 Leipzig, Germany
| | - Friedrich-Alexander Ludwig
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Department of Neuroradiopharmaceuticals, 04318 Leipzig, Germany
| | - Rodrigo Teodoro
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Department of Neuroradiopharmaceuticals, 04318 Leipzig, Germany
| | - Tatjana Sattler
- Department of Claw Animals, University of Leipzig, 04103 Leipzig, Germany
| | - Masoud Sadeghzadeh
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Department of Neuroradiopharmaceuticals, 04318 Leipzig, Germany
| | - Osama Sabri
- Department of Nuclear Medicine, University Hospital Leipzig, 04103 Leipzig, Germany
| | - Peter Brust
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Department of Neuroradiopharmaceuticals, 04318 Leipzig, Germany
| |
Collapse
|
10
|
Special Issue "Anticancer Drugs". Pharmaceuticals (Basel) 2019; 12:ph12030134. [PMID: 31527393 PMCID: PMC6789469 DOI: 10.3390/ph12030134] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 09/06/2019] [Indexed: 12/23/2022] Open
Abstract
The focus of this Special Issue of Pharmaceuticals is on the design, synthesis, and molecular mechanism of action of novel antitumor, drugs with a special emphasis on the relationship between the chemical structure and the biological activity of the molecules. This Special Issue also provides an understanding of the biologic and genotypic context in which targets are selected for oncology drug discovery, thus providing a rationalization for the biological activity of these drugs and guiding the design of more effective agents. In this Special Issue of Pharmaceuticals dedicated to anticancer drugs, we present a selection of preclinical research papers including both traditional chemotherapeutic agents and newer more targeted therapies and biological agents. We have included articles that report the design of small molecules with promising anticancer activity as tubulin inhibitors, vascular targeting agents, and topoisomerase targeting agents, alongside a comprehensive review of clinically successful antibody-drug conjugates used in cancer treatment.
Collapse
|
11
|
Liu F, Zhao B, Xia XT, Yan JR, Yu FQ, Yan GP, Hu J, Chen S, Wang YF, Liu H, Lan XL, Zhang YX. Al 18 F labeled sulfonamide-conjugated positron emission tomography tracer in vivo tumor-targeted imaging. J Cell Biochem 2019; 120:17006-17014. [PMID: 31131464 DOI: 10.1002/jcb.28961] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 03/06/2019] [Indexed: 01/01/2023]
Abstract
An ideal positron emission tomography (PET) tracer should be highly extractable by the tumor tissue or organ that contains low toxicity and can provide high-resolution images in vivo. In this work, the aim was to evaluate the application of Al18 F-labeled 1,4,7-triazacyclononane-1,4,7-triacetic acid containing sulfonamide group (18 F-Al-NOTA-SN) as a potential tumor-targeting signal-enhanced radioactive tracer in PET. SN as a tumor-targeting group was incorporated to NOTA to make a ligand. Subsequently, this ligand reacted with Na18 F and AlCl3 to produce a compound 18 F-Al-NOTA-SN. This compound was further characterized and its property in regard to cell cytotoxicity assay, microPET imaging, biodistribution, cell uptake assay, and tumor selectivity in vitro and in vivo, was also investigated. 18 F-Al-NOTA-SN possessed low cell cytotoxicity and uptake to COS-7 and 293T healthy cells and high cell cytotoxicity and uptake to MDA-MB-231, HepG2, and HeLa tumor cells in vitro. Moreover, 18 F-Al-NOTA-SN showed good tumor-targeting property and high PET signal enhancement of HeLa tumors, liver, and kidneys in mice, as well as the uptake ratios of tumor to blood and tumor to muscle, were 4.98 and 3.87, respectively. 18 F-Al-NOTA-SN can be accepted to be kidney and liver eliminated earlier and show a potential tumor-targeting signal-enhanced radioactive tracer in PET.
Collapse
Affiliation(s)
- Fan Liu
- School of Chemical Engineering and Pharmacy, Wuhan Institute of Technology, Wuhan, China.,School of Chemistry, Physics, and Mechanical Engineering, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Biao Zhao
- School of Materials Science and Engineering, Wuhan Institute of Technology, Wuhan, China
| | - Xiao-Tian Xia
- Center for PET of Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jia-Rui Yan
- School of Materials Science and Engineering, Wuhan Institute of Technology, Wuhan, China
| | - Fa-Quan Yu
- School of Chemical Engineering and Pharmacy, Wuhan Institute of Technology, Wuhan, China
| | - Guo-Ping Yan
- School of Materials Science and Engineering, Wuhan Institute of Technology, Wuhan, China
| | - Jia Hu
- Center for PET of Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Si Chen
- School of Materials Science and Engineering, Wuhan Institute of Technology, Wuhan, China
| | - Yu-Fang Wang
- School of Materials Science and Engineering, Wuhan Institute of Technology, Wuhan, China
| | - Hui Liu
- School of Materials Science and Engineering, Wuhan Institute of Technology, Wuhan, China
| | - Xiao-Li Lan
- Center for PET of Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yong-Xue Zhang
- Center for PET of Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|