Molecular Imaging in Oncology: Recent Advances

A special issue of Cancers (ISSN 2072-6694). This special issue belongs to the section "Methods and Technologies Development".

Deadline for manuscript submissions: 15 September 2024 | Viewed by 15802

Special Issue Editor


E-Mail Website
Guest Editor
Department of Radiology, Section of Nuclear Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
Interests: nuclear medicine; biomedical imaging; cancer pathogenesis and therapy
Special Issues, Collections and Topics in MDPI journals

Special Issue Information

Dear Colleagues, 

Nuclear medicine and molecular imaging thrive under continuous change. Molecular imaging is a technique that facilitates the non-invasive imaging of molecular targets. As an imaging technique, PET has two unique characteristics: a very high sensitivity (at the picomolar level) and the possibility of visualizing and quantifying molecular processes and targets under investigation. Primarily, however, PET offers the opportunity to obtain pathophysiological information from living patients in a non-invasive and quantified way. In the current decade, a wave of new theranostic compounds, targeting the same receptor for imaging and therapy, will change the oncology therapeutic arsenal. The rapid development of quantitative molecular imaging (PET and SPECT), combined with high-level abilities in cross-sectional morphological imaging (CT and MRI), has excavated exciting opportunities. This way, the field of nuclear medicine has rejuvenated itself repeatedly. This Special Issue highlights the hot topics of the molecular imaging tracer principle, which provides rich soil for scientific discovery, clinical diagnosis, treatment response monitoring, image-guided decision making, precision medicine and personalized medicine, non-invasive tissue characterization, the development of new tracers and imaging biomarkers, the development of (radiomics-based) prediction tools, artificial intelligence, deep learning, Big Data, targeted radionuclide therapies, and personalized dosimetry.

Prof. Dr. Lioe-Fee de Geus-Oei
Guest Editor

Manuscript Submission Information

Manuscripts should be submitted online at www.mdpi.com by registering and logging in to this website. Once you are registered, click here to go to the submission form. Manuscripts can be submitted until the deadline. All submissions that pass pre-check are peer-reviewed. Accepted papers will be published continuously in the journal (as soon as accepted) and will be listed together on the special issue website. Research articles, review articles as well as short communications are invited. For planned papers, a title and short abstract (about 100 words) can be sent to the Editorial Office for announcement on this website.

Submitted manuscripts should not have been published previously, nor be under consideration for publication elsewhere (except conference proceedings papers). All manuscripts are thoroughly refereed through a single-blind peer-review process. A guide for authors and other relevant information for submission of manuscripts is available on the Instructions for Authors page. Cancers is an international peer-reviewed open access semimonthly journal published by MDPI.

Please visit the Instructions for Authors page before submitting a manuscript. The Article Processing Charge (APC) for publication in this open access journal is 2900 CHF (Swiss Francs). Submitted papers should be well formatted and use good English. Authors may use MDPI's English editing service prior to publication or during author revisions.

Keywords

  • nuclear medicine
  • molecular imaging
  • PET
  • oncology
  • cancer
  • SPECT
  • precision medicine
  • personalized medicine
  • biomarker
  • targeted radionuclide therapies
  • personalized dosimetry

Published Papers (11 papers)

Order results
Result details
Select all
Export citation of selected articles as:

Research

Jump to: Review, Other

13 pages, 4124 KiB  
Article
Clinical [18F]FSPG Positron Emission Tomography Imaging Reveals Heterogeneity in Tumor-Associated System xc Activity
by Amy R. Sharkey, Norman Koglin, Erik S. Mittra, Sangwon Han, Gary J. R. Cook and Timothy H. Witney
Cancers 2024, 16(7), 1437; https://doi.org/10.3390/cancers16071437 - 08 Apr 2024
Viewed by 612
Abstract
Background: (4S)-4-(3-[18F]fluoropropyl)-L-glutamic acid ([18F]FSPG) positron emission tomography/computed tomography (PET/CT) provides a readout of system xc transport activity and has been used for cancer detection in clinical studies of different cancer types. As system xc [...] Read more.
Background: (4S)-4-(3-[18F]fluoropropyl)-L-glutamic acid ([18F]FSPG) positron emission tomography/computed tomography (PET/CT) provides a readout of system xc transport activity and has been used for cancer detection in clinical studies of different cancer types. As system xc provides the rate-limiting precursor for glutathione biosynthesis, an abundant antioxidant, [18F]FSPG imaging may additionally provide important prognostic information. Here, we performed an analysis of [18F]FSPG radiotracer distribution between primary tumors, metastases, and normal organs from cancer patients. We further assessed the heterogeneity of [18F]FSPG retention between cancer types, and between and within individuals. Methods: This retrospective analysis of prospectively collected data compared [18F]FSPG PET/CT in subjects with head and neck squamous cell cancer (HNSCC, n = 5) and non-small-cell lung cancer (NSCLC, n = 10), scanned at different institutions. Using semi-automated regions of interest drawn around tumors and metastases, the maximum standardized uptake value (SUVmax), SUVmean, SUV standard deviation and SUVpeak were measured. [18F]FSPG time–activity curves (TACs) for normal organs, primary tumors and metastases were subsequently compared to 18F-2-fluoro-2-deoxy-D-glucose ([18F]FDG) PET/CT at 60 min post injection (p.i.). Results: The mean administered activity of [18F]FSPG was 309.3 ± 9.1 MBq in subjects with NSCLC and 285.1 ± 11.3 MBq in those with HNSCC. The biodistribution of [18F]FSPG in both cohorts showed similar TACs in healthy organs from cancer patients. There was no statistically significant overall difference in the average SUVmax of tumor lesions at 60 min p.i. for NSCLC (8.1 ± 7.1) compared to HNSCC (6.0 ± 4.1; p = 0.29) for [18F]FSPG. However, there was heterogeneous retention between and within cancer types; the SUVmax at 60 min p.i. ranged from 1.4 to 23.7 in NSCLC and 3.1–12.1 in HNSCC. Conclusion: [18F]FSPG PET/CT imaging from both NSCLC and HNSCC cohorts showed the same normal-tissue biodistribution, but marked tumor heterogeneity across subjects and between lesions. Despite rapid elimination through the urinary tract and low normal-background tissue retention, the diagnostic potential of [18F]FSPG was limited by variability in tumor retention. As [18F]FSPG retention is mediated by the tumor’s antioxidant capacity and response to oxidative stress, this heterogeneity may provide important insights into an individual tumor’s response or resistance to therapy. Full article
(This article belongs to the Special Issue Molecular Imaging in Oncology: Recent Advances)
Show Figures

Figure 1

12 pages, 3038 KiB  
Article
89Zr-Immuno-PET with Immune Checkpoint Inhibitors: Measuring Target Engagement in Healthy Organs
by Iris H. C. Miedema, Jessica E. Wijngaarden, Johanna E. E. Pouw, Gerben J. C. Zwezerijnen, Hylke J. Sebus, Egbert Smit, Adrianus J. de Langen, Idris Bahce, Andrea Thiele, Daniëlle J. Vugts, Ronald Boellaard, Marc C. Huisman and C. Willemien Menke-van der Houven van Oordt
Cancers 2023, 15(23), 5546; https://doi.org/10.3390/cancers15235546 - 23 Nov 2023
Cited by 2 | Viewed by 998
Abstract
Introduction: 89Zr-immuno-PET (positron emission tomography with zirconium-89-labeled monoclonal antibodies ([89Zr]Zr-mAbs)) can be used to study the biodistribution of mAbs targeting the immune system. The measured uptake consists of target-specific and non-specific components, and it can be influenced by plasma availability [...] Read more.
Introduction: 89Zr-immuno-PET (positron emission tomography with zirconium-89-labeled monoclonal antibodies ([89Zr]Zr-mAbs)) can be used to study the biodistribution of mAbs targeting the immune system. The measured uptake consists of target-specific and non-specific components, and it can be influenced by plasma availability of the tracer. To find evidence for target-specific uptake, i.e., target engagement, we studied five immune-checkpoint-targeting [89Zr]Zr-mAbs to (1) compare the uptake with previously reported baseline values for non-specific organ uptake (ns-baseline) and (2) look for saturation effects of increasing mass doses. Method: 89Zr-immuno-PET data from five [89Zr]Zr-mAbs, i.e., nivolumab and pembrolizumab (anti-PD-1), durvalumab (anti-PD-L1), BI 754,111 (anti-LAG-3), and ipilimumab (anti-CTLA-4), were analysed. For each mAb, 2–3 different mass doses were evaluated. PET scans and blood samples from at least two time points 24 h post injection were available. In 35 patients, brain, kidneys, liver, spleen, lungs, and bone marrow were delineated. Patlak analysis was used to account for differences in plasma activity concentration and to quantify irreversible uptake (Ki). To identify target engagement, Ki values were compared to ns-baseline Ki values previously reported, and the effect of increasing mass doses on Ki was investigated. Results: All mAbs, except ipilimumab, showed Ki values in spleen above the ns-baseline for the lowest administered mass dose, in addition to decreasing Ki values with higher mass doses, both indicative of target engagement. For bone marrow, no ns-baseline was established previously, but a similar pattern was observed. For kidneys, most mAbs showed Ki values within the ns-baseline for both low and high mass doses. However, with high mass doses, some saturation effects were seen, suggestive of a lower ns-baseline value. Ki values were near zero in brain tissue for all mass doses of all mAbs. Conclusion: Using Patlak analysis and the established ns-baseline values, evidence for target engagement in (lymphoid) organs for several immune checkpoint inhibitors could be demonstrated. A decrease in the Ki values with increasing mass doses supports the applicability of Patlak analysis for the assessment of target engagement for PET ligands with irreversible uptake behavior. Full article
(This article belongs to the Special Issue Molecular Imaging in Oncology: Recent Advances)
Show Figures

Graphical abstract

16 pages, 2246 KiB  
Article
89Zr-Trastuzumab PET/CT Imaging of HER2-Positive Breast Cancer for Predicting Pathological Complete Response after Neoadjuvant Systemic Therapy: A Feasibility Study
by D. G. J. Linders, M. M. Deken, M. A. van Dam, M. N. J. M. Wasser, E. M. C. Voormolen, J. R. Kroep, G. A. M. S. van Dongen, D. Vugts, H. M. Oosterkamp, M. E. Straver, C. J. H. van de Velde, D. Cohen, P. Dibbets-Schneider, F. H. P. van Velden, L. M. Pereira Arias-Bouda, A. L. Vahrmeijer, G. J. Liefers, L. F. de Geus-Oei and D. E. Hilling
Cancers 2023, 15(20), 4980; https://doi.org/10.3390/cancers15204980 - 13 Oct 2023
Viewed by 1220
Abstract
Background: Approximately 20% of invasive ductal breast malignancies are human epidermal growth factor receptor 2 (HER2)-positive. These patients receive neoadjuvant systemic therapy (NAT) including HER2-targeting therapies. Up to 65% of patients achieve a pathological complete response (pCR). These patients might not have needed [...] Read more.
Background: Approximately 20% of invasive ductal breast malignancies are human epidermal growth factor receptor 2 (HER2)-positive. These patients receive neoadjuvant systemic therapy (NAT) including HER2-targeting therapies. Up to 65% of patients achieve a pathological complete response (pCR). These patients might not have needed surgery. However, accurate preoperative identification of a pCR remains challenging. A radiologic complete response (rCR) on MRI corresponds to a pCR in only 73% of patients. The current feasibility study investigates if HER2-targeted PET/CT-imaging using Zirconium-89 (89Zr)-radiolabeled trastuzumab can be used for more accurate NAT response evaluation. Methods: HER2-positive breast cancer patients scheduled to undergo NAT and subsequent surgery received a 89Zr-trastuzumab PET/CT both before (PET/CT-1) and after (PET/CT-2) NAT. Qualitative and quantitative response evaluation was performed. Results: Six patients were enrolled. All primary tumors could be identified on PET/CT-1. Four patients had a pCR and two a pathological partial response (pPR) in the primary tumor. Qualitative assessment of PET/CT resulted in an accuracy of 66.7%, compared to 83.3% of the standard-of-care MRI. Quantitative assessment showed a difference between the SUVR on PET/CT-1 and PET/CT-2 (ΔSUVR) in patients with a pPR and pCR of −48% and −90% (p = 0.133), respectively. The difference in tumor-to-blood ratio on PET/CT-1 and PET/CT-2 (ΔTBR) in patients with pPR and pCR was −79% and −94% (p = 0.133), respectively. Three patients had metastatic lymph nodes at diagnosis that were all identified on PET/CT-1. All three patients achieved a nodal pCR. Qualitative assessment of the lymph nodes with PET/CT resulted in an accuracy of 66.7%, compared to 50% of the MRI. Conclusions: NAT response evaluation using 89Zr-trastuzumab PET/CT is feasible. In the current study, qualitative assessment of the PET/CT images is not superior to standard-of-care MRI. Our results suggest that quantitative assessment of 89Zr-trastuzumab PET/CT has potential for a more accurate response evaluation of the primary tumor after NAT in HER2-positive breast cancer. Full article
(This article belongs to the Special Issue Molecular Imaging in Oncology: Recent Advances)
Show Figures

Figure 1

12 pages, 1139 KiB  
Article
Prognostic Value of [18F]FDG PET Radiomics to Detect Peritoneal and Distant Metastases in Locally Advanced Gastric Cancer—A Side Study of the Prospective Multicentre PLASTIC Study
by Lieke C. E. Pullen, Wyanne A. Noortman, Lianne Triemstra, Cas de Jongh, Fenna J. Rademaker, Romy Spijkerman, Gijsbert M. Kalisvaart, Emma C. Gertsen, Lioe-Fee de Geus-Oei, Nelleke Tolboom, Wobbe O. de Steur, Maura Dantuma, Riemer H. J. A. Slart, Richard van Hillegersberg, Peter D. Siersema, Jelle P. Ruurda, Floris H. P. van Velden, Erik Vegt and on behalf of the PLASTIC Study Group
Cancers 2023, 15(11), 2874; https://doi.org/10.3390/cancers15112874 - 23 May 2023
Cited by 1 | Viewed by 1347
Abstract
Aim: To improve identification of peritoneal and distant metastases in locally advanced gastric cancer using [18F]FDG-PET radiomics. Methods: [18F]FDG-PET scans of 206 patients acquired in 16 different Dutch hospitals in the prospective multicentre PLASTIC-study were analysed. Tumours were delineated [...] Read more.
Aim: To improve identification of peritoneal and distant metastases in locally advanced gastric cancer using [18F]FDG-PET radiomics. Methods: [18F]FDG-PET scans of 206 patients acquired in 16 different Dutch hospitals in the prospective multicentre PLASTIC-study were analysed. Tumours were delineated and 105 radiomic features were extracted. Three classification models were developed to identify peritoneal and distant metastases (incidence: 21%): a model with clinical variables, a model with radiomic features, and a clinicoradiomic model, combining clinical variables and radiomic features. A least absolute shrinkage and selection operator (LASSO) regression classifier was trained and evaluated in a 100-times repeated random split, stratified for the presence of peritoneal and distant metastases. To exclude features with high mutual correlations, redundancy filtering of the Pearson correlation matrix was performed (r = 0.9). Model performances were expressed by the area under the receiver operating characteristic curve (AUC). In addition, subgroup analyses based on Lauren classification were performed. Results: None of the models could identify metastases with low AUCs of 0.59, 0.51, and 0.56, for the clinical, radiomic, and clinicoradiomic model, respectively. Subgroup analysis of intestinal and mixed-type tumours resulted in low AUCs of 0.67 and 0.60 for the clinical and radiomic models, and a moderate AUC of 0.71 in the clinicoradiomic model. Subgroup analysis of diffuse-type tumours did not improve the classification performance. Conclusion: Overall, [18F]FDG-PET-based radiomics did not contribute to the preoperative identification of peritoneal and distant metastases in patients with locally advanced gastric carcinoma. In intestinal and mixed-type tumours, the classification performance of the clinical model slightly improved with the addition of radiomic features, but this slight improvement does not outweigh the laborious radiomic analysis. Full article
(This article belongs to the Special Issue Molecular Imaging in Oncology: Recent Advances)
Show Figures

Figure 1

11 pages, 480 KiB  
Article
Development and External Validation of a PET Radiomic Model for Prognostication of Head and Neck Cancer
by Wyanne A. Noortman, Nicolas Aide, Dennis Vriens, Lisa S. Arkes, Cornelis H. Slump, Ronald Boellaard, Jelle J. Goeman, Christophe M. Deroose, Jean-Pascal Machiels, Lisa F. Licitra, Renaud Lhommel, Alessandra Alessi, Erwin Woff, Karolien Goffin, Christophe Le Tourneau, Jocelyn Gal, Stéphane Temam, Jean-Pierre Delord, Floris H. P. van Velden and Lioe-Fee de Geus-Oei
Cancers 2023, 15(10), 2681; https://doi.org/10.3390/cancers15102681 - 09 May 2023
Viewed by 1661
Abstract
Aim: To build and externally validate an [18F]FDG PET radiomic model to predict overall survival in patients with head and neck squamous cell carcinoma (HNSCC). Methods: Two multicentre datasets of patients with operable HNSCC treated with preoperative afatinib who underwent a [...] Read more.
Aim: To build and externally validate an [18F]FDG PET radiomic model to predict overall survival in patients with head and neck squamous cell carcinoma (HNSCC). Methods: Two multicentre datasets of patients with operable HNSCC treated with preoperative afatinib who underwent a baseline and evaluation [18F]FDG PET/CT scan were included (EORTC: n = 20, Unicancer: n = 34). Tumours were delineated, and radiomic features were extracted. Each cohort served once as a training and once as an external validation set for the prediction of overall survival. Supervised feature selection was performed using variable hunting with variable importance, selecting the top two features. A Cox proportional hazards regression model using selected radiomic features and clinical characteristics was fitted on the training dataset and validated in the external validation set. Model performances are expressed by the concordance index (C-index). Results: In both models, the radiomic model surpassed the clinical model with validation C-indices of 0.69 and 0.79 vs. 0.60 and 0.67, respectively. The model that combined the radiomic features and clinical variables performed best, with validation C-indices of 0.71 and 0.82. Conclusion: Although assessed in two small but independent cohorts, an [18F]FDG-PET radiomic signature based on the evaluation scan seems promising for the prediction of overall survival for HNSSC treated with preoperative afatinib. The robustness and clinical applicability of this radiomic signature should be assessed in a larger cohort. Full article
(This article belongs to the Special Issue Molecular Imaging in Oncology: Recent Advances)
Show Figures

Figure 1

13 pages, 9340 KiB  
Article
Prostate-Specific Membrane Antigen Targeted Pet/CT Imaging in Patients with Colon, Gastric and Pancreatic Cancer
by Floris A. Vuijk, Fleur Kleiburg, Wyanne A. Noortman, Linda Heijmen, Shirin Feshtali Shahbazi, Floris H. P. van Velden, Victor M. Baart, Shadhvi S. Bhairosingh, Bert D. Windhorst, Lukas J. A. C. Hawinkels, Petra Dibbets-Schneider, Neanke Bouwman, Stijn A. L. P. Crobach, Arantza Fariña-Sarasqueta, Andreas W. K. S. Marinelli, Daniela E. Oprea-Lager, Rutger-Jan Swijnenburg, Frits Smit, Alexander L. Vahrmeijer, Lioe-Fee de Geus-Oei, Denise E. Hilling and Marije Slingerlandadd Show full author list remove Hide full author list
Cancers 2022, 14(24), 6209; https://doi.org/10.3390/cancers14246209 - 15 Dec 2022
Cited by 4 | Viewed by 2538
Abstract
Current imaging modalities frequently misjudge disease stage in colorectal, gastric and pancreatic cancer. As treatment decisions are dependent on disease stage, incorrect staging has serious consequences. Previous preclinical research and case reports indicate that prostate-specific membrane antigen (PSMA)-targeted PET/CT imaging might provide a [...] Read more.
Current imaging modalities frequently misjudge disease stage in colorectal, gastric and pancreatic cancer. As treatment decisions are dependent on disease stage, incorrect staging has serious consequences. Previous preclinical research and case reports indicate that prostate-specific membrane antigen (PSMA)-targeted PET/CT imaging might provide a solution to some of these challenges. This prospective clinical study aims to assess the feasibility of [18F]DCFPyL PET/CT imaging to target and visualize primary colon, gastric and pancreatic cancer. In this prospective clinical trial, patients with colon, gastric and pancreatic cancer were included and underwent both [18F]DCFPyL and [18F]FDG PET/CT scans prior to surgical resection or (for gastric cancer) neoadjuvant therapy. Semiquantitative analysis of immunohistochemical PSMA staining was performed on the surgical resection specimens, and the results were correlated to imaging parameters. The results of this study demonstrate detection of the primary tumor by [18F]DCFPyL PET/CT in 7 out of 10 patients with colon, gastric and pancreatic cancer, with a mean tumor-to-blood pool ratio (TBR) of 3.3 and mean SUVmax of 3.6. However, due to the high surrounding uptake, visual distinction of these tumors was difficult, and the SUVmax and TBR on [18F]FDG PET/CT were significantly higher than on [18F]DCFPyL PET/CT. In addition, no correlation between PSMA expression in the resection specimen and SUVmax on [18F]DCFPyL PET/CT was found. In conclusion, the detection of several gastrointestinal cancers using [18F]DCFPyL PET/CT is feasible. However, low tumor expression and high uptake physiologically in organs/background hamper the clear distinction of the tumor. As a result, [18F]FDG PET/CT was superior in detecting colon, gastric and pancreatic cancers. Full article
(This article belongs to the Special Issue Molecular Imaging in Oncology: Recent Advances)
Show Figures

Figure 1

Review

Jump to: Research, Other

23 pages, 3424 KiB  
Review
ImmunoPET Targeting Receptor Tyrosine Kinase: Clinical Applications
by Flavia Linguanti, Elisabetta Maria Abenavoli, Raffaella Calabretta, Valentina Berti and Egesta Lopci
Cancers 2023, 15(24), 5886; https://doi.org/10.3390/cancers15245886 - 18 Dec 2023
Cited by 1 | Viewed by 849
Abstract
Receptor tyrosine kinases, or RTKs, are one large family of cell surface receptors involved in signal transduction, which represent an integral part of the signaling pathways. They play a crucial role in most important cellular processes, starting with the cell cycle, proliferation and [...] Read more.
Receptor tyrosine kinases, or RTKs, are one large family of cell surface receptors involved in signal transduction, which represent an integral part of the signaling pathways. They play a crucial role in most important cellular processes, starting with the cell cycle, proliferation and differentiation, as well as cell migration, metabolism and survival. The introduction of ImmunoPET evaluating the expression of RTKs by specific monoclonal antibodies (mAbs) or antibody fragments is regarded as a promising tool for imaging treatment efficacy and developing anticancer therapeutics. Our review focuses mainly on the current clinical research regarding ImmunoPET targeting RTKs, with particular interest in the epidermal growth factor family, or HER family, and vascular endothelial-derived growth factor/receptor. Full article
(This article belongs to the Special Issue Molecular Imaging in Oncology: Recent Advances)
Show Figures

Figure 1

15 pages, 1681 KiB  
Review
Is System xc a Suitable Target for Tumour Detection and Response Assessment with Imaging?
by Amy R. Sharkey, Timothy H. Witney and Gary J. R. Cook
Cancers 2023, 15(23), 5573; https://doi.org/10.3390/cancers15235573 - 24 Nov 2023
Cited by 1 | Viewed by 1069
Abstract
System xc is upregulated in cancer cells and can be imaged using novel radiotracers, most commonly with (4S)-4-(3-[18F]fluoropropyl)-L-glutamic acid (18F-FSPG). The aim of this review was to summarise the use of 18F-FSPG in humans, explore the [...] Read more.
System xc is upregulated in cancer cells and can be imaged using novel radiotracers, most commonly with (4S)-4-(3-[18F]fluoropropyl)-L-glutamic acid (18F-FSPG). The aim of this review was to summarise the use of 18F-FSPG in humans, explore the benefits and limitations of 18F-FSPG, and assess the potential for further use of 18F-FSPG in cancer patients. To date, ten papers have described the use of 18F-FSPG in human cancers. These studies involved small numbers of patients (range 1–26) and assessed the use of 18F-FSPG as a general oncological diagnostic agent across different cancer types. These clinical trials were contrasting in their findings, limiting the scope of 18F-FSPG PET/CT as a purely diagnostic agent, primarily due to heterogeneity of 18F-FSPG retention both between cancer types and patients. Despite these limitations, a potential further application for 18F-FSPG is in the assessment of early treatment response and prediction of treatment resistance. Animal models of cancer have shown that changes in 18F-FSPG retention following effective therapy precede glycolytic changes, as indicated by 18F-FDG, and changes in tumour volume, as measured by CT. If these results could be replicated in human clinical trials, imaging with 18F-FSPG PET/CT would offer an exciting route towards addressing the currently unmet clinical needs of treatment resistance prediction and early imaging assessment of therapy response. Full article
(This article belongs to the Special Issue Molecular Imaging in Oncology: Recent Advances)
Show Figures

Figure 1

21 pages, 5658 KiB  
Review
Current and Future Use of Long Axial Field-of-View Positron Emission Tomography/Computed Tomography Scanners in Clinical Oncology
by Mostafa Roya, Samaneh Mostafapour, Philipp Mohr, Laura Providência, Zekai Li, Johannes H. van Snick, Adrienne H. Brouwers, Walter Noordzij, Antoon T. M. Willemsen, Rudi A. J. O. Dierckx, Adriaan A. Lammertsma, Andor W. J. M. Glaudemans, Charalampos Tsoumpas, Riemer H. J. A. Slart and Joyce van Sluis
Cancers 2023, 15(21), 5173; https://doi.org/10.3390/cancers15215173 - 27 Oct 2023
Cited by 3 | Viewed by 1341
Abstract
The latest technical development in the field of positron emission tomography/computed tomography (PET/CT) imaging has been the extension of the PET axial field-of-view. As a result of the increased number of detectors, the long axial field-of-view (LAFOV) PET systems are not only characterized [...] Read more.
The latest technical development in the field of positron emission tomography/computed tomography (PET/CT) imaging has been the extension of the PET axial field-of-view. As a result of the increased number of detectors, the long axial field-of-view (LAFOV) PET systems are not only characterized by a larger anatomical coverage but also by a substantially improved sensitivity, compared with conventional short axial field-of-view PET systems. In clinical practice, this innovation has led to the following optimization: (1) improved overall image quality, (2) decreased duration of PET examinations, (3) decreased amount of radioactivity administered to the patient, or (4) a combination of any of the above. In this review, novel applications of LAFOV PET in oncology are highlighted and future directions are discussed. Full article
(This article belongs to the Special Issue Molecular Imaging in Oncology: Recent Advances)
Show Figures

Figure 1

22 pages, 5895 KiB  
Review
Smart Polymeric Micelles for Anticancer Hydrophobic Drugs
by Andy Guzmán Rodríguez, Marquiza Sablón Carrazana, Chrislayne Rodríguez Tanty, Martijn J. A. Malessy, Gastón Fuentes and Luis J. Cruz
Cancers 2023, 15(1), 4; https://doi.org/10.3390/cancers15010004 - 20 Dec 2022
Cited by 10 | Viewed by 2100
Abstract
Cancer has become one of the deadliest diseases in our society. Surgery accompanied by subsequent chemotherapy is the treatment most used to prolong or save the patient’s life. Still, it carries secondary risks such as infections and thrombosis and causes cytotoxic effects in [...] Read more.
Cancer has become one of the deadliest diseases in our society. Surgery accompanied by subsequent chemotherapy is the treatment most used to prolong or save the patient’s life. Still, it carries secondary risks such as infections and thrombosis and causes cytotoxic effects in healthy tissues. Using nanocarriers such as smart polymer micelles is a promising alternative to avoid or minimize these problems. These nanostructured systems will be able to encapsulate hydrophilic and hydrophobic drugs through modified copolymers with various functional groups such as carboxyls, amines, hydroxyls, etc. The release of the drug occurs due to the structural degradation of these copolymers when they are subjected to endogenous (pH, redox reactions, and enzymatic activity) and exogenous (temperature, ultrasound, light, magnetic and electric field) stimuli. We did a systematic review of the efficacy of smart polymeric micelles as nanocarriers for anticancer drugs (doxorubicin, paclitaxel, docetaxel, lapatinib, cisplatin, adriamycin, and curcumin). For this reason, we evaluate the influence of the synthesis methods and the physicochemical properties of these systems that subsequently allow an effective encapsulation and release of the drug. On the other hand, we demonstrate how computational chemistry will enable us to guide and optimize the design of these micelles to carry out better experimental work. Full article
(This article belongs to the Special Issue Molecular Imaging in Oncology: Recent Advances)
Show Figures

Figure 1

Other

Jump to: Research, Review

12 pages, 1089 KiB  
Commentary
Integrating [18F]-Fluorodeoxyglucose Positron Emission Tomography with Computed Tomography with Radiation Therapy and Immunomodulation in Precision Therapy for Solid Tumors
by Conor M. Prendergast, Egesta Lopci, Romain-David Seban, Dorine De Jong, Samy Ammari, Sanjay Aneja, Antonin Lévy, Abin Sajan, Mary M. Salvatore, Kathleen M. Cappacione, Lawrence H. Schwartz, Eric Deutsch and Laurent Dercle
Cancers 2023, 15(21), 5179; https://doi.org/10.3390/cancers15215179 - 27 Oct 2023
Cited by 1 | Viewed by 971
Abstract
[18F]-FDG positron emission tomography with computed tomography (PET/CT) imaging is widely used to enhance the quality of care in patients diagnosed with cancer. Furthermore, it holds the potential to offer insight into the synergic effect of combining radiation therapy (RT) with [...] Read more.
[18F]-FDG positron emission tomography with computed tomography (PET/CT) imaging is widely used to enhance the quality of care in patients diagnosed with cancer. Furthermore, it holds the potential to offer insight into the synergic effect of combining radiation therapy (RT) with immuno-oncological (IO) agents. This is achieved by evaluating treatment responses both at the RT and distant tumor sites, thereby encompassing the phenomenon known as the abscopal effect. In this context, PET/CT can play an important role in establishing timelines for RT/IO administration and monitoring responses, including novel patterns such as hyperprogression, oligoprogression, and pseudoprogression, as well as immune-related adverse events. In this commentary, we explore the incremental value of PET/CT to enhance the combination of RT with IO in precision therapy for solid tumors, by offering supplementary insights to recently released joint guidelines. Full article
(This article belongs to the Special Issue Molecular Imaging in Oncology: Recent Advances)
Show Figures

Figure 1

Planned Papers

The below list represents only planned manuscripts. Some of these manuscripts have not been received by the Editorial Office yet. Papers submitted to MDPI journals are subject to peer-review.

Title: 99mTc-sestamibi quantification using prone SPECT/CT for therapy monitoring in locally advanced breast cancer
Authors: Christinne Corion; Lenka M. Pereira Arias-Bouda
Affiliation: Leids Universitair Medisch Centrum, Leiden, Netherlands

Back to TopTop