Radiopharmaceuticals for Oncological Diseases

A special issue of Cancers (ISSN 2072-6694).

Deadline for manuscript submissions: closed (30 November 2021) | Viewed by 43607

Special Issue Editor


E-Mail Website
Guest Editor
University Hospital Basel, University of Basel, Basel, Switzerland
Interests: targeted radiopharmaceuticals; molecular nuclear imaging; radionuclide therapy; peptides; radio-theranostics; radiometals

Special Issue Information

Dear Colleagues,

Cancer is a leading cause of death worldwide. Radiopharmaceuticals are able to deliver radiation to cancer cells, independent of their location in the body. Radioactivity is a particularly sensitive “antenna” with excellent tissue penetration. Thus, nuclear imaging via SPECT and PET has great advantages over other imaging modalities regarding sensitivity. In addition, targeted radiopharmaceuticals provide very effective systemic treatment options due to the specific delivery of lethal radioactive doses to cancer tissues, while sparing healthy tissues. Importantly, certain radiopharmaceutical designs are blending diagnostic and therapeutic properties within the same molecule (radio-theranostics). Radio-theranostics can be used for diagnosis, staging, patients’ stratification, and treatment, thus reinforcing precision medicine in oncology.

The incredible amount of innovation in the production of radionuclides with different physical and chemical properties, in the discovery of molecular targets expressed in cancer cells and in the cancer microenvironment, and in the development of chemically diverse vector molecules able to deliver radiation to these targets (e.g., small molecules, peptides, antibodies, engineered proteins, and others), has led to exciting advances in the last few years. Among them are bench-to-bedside translation of novel radio-theranostics in different oncological indications (e.g., radiolabeled PSMA inhibitors), new registrations (e.g., Lutathera), and substantial investments of global pharmaceutical companies.

This Special Issue titled “Radiopharmaceuticals for Oncological Diseases” aims to provide the state-of-the-art in this field. You are cordially invited to contribute with a review article or with an original article that explores novel radiopharmaceuticals for oncological indications or new oncological applications of known radiopharmaceuticals.

Prof. Dr. Melpomeni Fani
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

  • radiopharmaceuticals
  • theranostics
  • radionuclides
  • PET and SPECT tracers
  • endoradiotherapy
  • peptide receptor radionuclide therapy
  • radioligand therapy
  • molecular targets in cancer
  • nuclear oncology
  • clinical translation

Published Papers (12 papers)

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

Research

Jump to: Review, Other

18 pages, 1352 KiB  
Article
Auger Emitter Conjugated PARP Inhibitor for Therapy in Triple Negative Breast Cancers: A Comparative In-Vitro Study
by Ramya Ambur Sankaranarayanan, Jennifer Peil, Andreas T. J. Vogg, Carsten Bolm, Steven Terhorst, Arno Classen, Matthias Bauwens, Jochen Maurer, Felix Mottaghy and Agnieszka Morgenroth
Cancers 2022, 14(1), 230; https://doi.org/10.3390/cancers14010230 - 04 Jan 2022
Cited by 14 | Viewed by 2910 | Correction
Abstract
PARP1 inhibitors (PARPi) are currently approved for BRCAmut metastatic breast cancer, but they have shown limited response in triple negative breast cancer (TNBC) patients. Combination of an Auger emitter with PARPis enables PARP inhibition and DNA strand break induction simultaneously. This will [...] Read more.
PARP1 inhibitors (PARPi) are currently approved for BRCAmut metastatic breast cancer, but they have shown limited response in triple negative breast cancer (TNBC) patients. Combination of an Auger emitter with PARPis enables PARP inhibition and DNA strand break induction simultaneously. This will enhance cytotoxicity and additionally allow a theranostic approach. This study presents the radiosynthesis of the Auger emitter [125I] coupled olaparib derivative: [125I]-PARPi-01, and its therapeutic evaluation in a panel of TNBC cell lines. Specificity was tested by a blocking assay. DNA strand break induction was analysed by γH2AX immunofluorescence staining. Cell cycle analysis and apoptosis assays were studied using flow cytometry in TNBC cell lines (BRCAwt/mut). Anchorage independent growth potential was evaluated using soft agar assay. [125I]-PARPi-01 showed PARP1-specificity and higher cytotoxicity than olaparib in TNBC cell lines irrespective of BRCA their status. Cell lines harbouring DNA repair deficiency showed response to [125I]-PARPi-01 monotherapy. Combined treatment with Dox-NP further enhanced therapeutic efficiency in metastatic resistant BRCAwt cell lines. The clonogenic survival was significantly reduced after treatment with [125I]-PARPi-01 in all TNBC lines investigated. Therapeutic efficacy was further enhanced after combined treatment with chemotherapeutics. [125I]-PARPi-01 is a promising radiotherapeutic agent for low radiation dosages, and mono/combined therapies of TNBC. Full article
(This article belongs to the Special Issue Radiopharmaceuticals for Oncological Diseases)
Show Figures

Figure 1

Review

Jump to: Research, Other

19 pages, 2501 KiB  
Review
Targeted Dual-Modal PET/SPECT-NIR Imaging: From Building Blocks and Construction Strategies to Applications
by Syed Muhammad Usama, Sierra C. Marker, Servando Hernandez Vargas, Solmaz AghaAmiri, Sukhen C. Ghosh, Naruhiko Ikoma, Hop S. Tran Cao, Martin J. Schnermann and Ali Azhdarinia
Cancers 2022, 14(7), 1619; https://doi.org/10.3390/cancers14071619 - 23 Mar 2022
Cited by 8 | Viewed by 3579
Abstract
Molecular imaging is an emerging non-invasive method to qualitatively and quantitively visualize and characterize biological processes. Among the imaging modalities, PET/SPECT and near-infrared (NIR) imaging provide synergistic properties that result in deep tissue penetration and up to cell-level resolution. Dual-modal PET/SPECT-NIR agents are [...] Read more.
Molecular imaging is an emerging non-invasive method to qualitatively and quantitively visualize and characterize biological processes. Among the imaging modalities, PET/SPECT and near-infrared (NIR) imaging provide synergistic properties that result in deep tissue penetration and up to cell-level resolution. Dual-modal PET/SPECT-NIR agents are commonly combined with a targeting ligand (e.g., antibody or small molecule) to engage biomolecules overexpressed in cancer, thereby enabling selective multimodal visualization of primary and metastatic tumors. The use of such agents for (i) preoperative patient selection and surgical planning and (ii) intraoperative FGS could improve surgical workflow and patient outcomes. However, the development of targeted dual-modal agents is a chemical challenge and a topic of ongoing research. In this review, we define key design considerations of targeted dual-modal imaging from a topological perspective, list targeted dual-modal probes disclosed in the last decade, review recent progress in the field of NIR fluorescent probe development, and highlight future directions in this rapidly developing field. Full article
(This article belongs to the Special Issue Radiopharmaceuticals for Oncological Diseases)
Show Figures

Figure 1

14 pages, 1170 KiB  
Review
Radiolabeled Somatostatin Analogs—A Continuously Evolving Class of Radiopharmaceuticals
by Melpomeni Fani, Rosalba Mansi, Guillaume P. Nicolas and Damian Wild
Cancers 2022, 14(5), 1172; https://doi.org/10.3390/cancers14051172 - 24 Feb 2022
Cited by 26 | Viewed by 2959
Abstract
Somatostatin receptors (SSTs) are recognized as favorable molecular targets in neuroendocrine tumors (NETs) and neuroendocrine neoplasms (NENs), with subtype 2 (SST2) being the predominantly and most frequently expressed. PET/CT imaging with 68Ga-labeled SST agonists, e.g., 68Ga-DOTA-TOC (SomaKit TOC® [...] Read more.
Somatostatin receptors (SSTs) are recognized as favorable molecular targets in neuroendocrine tumors (NETs) and neuroendocrine neoplasms (NENs), with subtype 2 (SST2) being the predominantly and most frequently expressed. PET/CT imaging with 68Ga-labeled SST agonists, e.g., 68Ga-DOTA-TOC (SomaKit TOC®) or 68Ga-DOTA-TATE (NETSPOT®), plays an important role in staging and restaging these tumors and can identify patients who qualify and would potentially benefit from peptide receptor radionuclide therapy (PRRT) with the therapeutic counterparts 177Lu-DOTA-TOC or 177Lu-DOTA-TATE (Lutathera®). This is an important feature of SST targeting, as it allows a personalized treatment approach (theranostic approach). Today, new developments hold promise for enhancing diagnostic accuracy and therapeutic efficacy. Among them, the use of SST2 antagonists, such as JR11 and LM3, has shown certain advantages in improving image sensitivity and tumor radiation dose, and there is evidence that they may find application in other oncological indications beyond NETs and NENs. In addition, PRRT performed with more cytotoxic α-emitters, such as 225Ac, or β- and Auger electrons, such as 161Tb, presents higher efficacy. It remains to be seen if any of these new developments will overpower the established radiolabeled SST analogs and PRRT with β--emitters. Full article
(This article belongs to the Special Issue Radiopharmaceuticals for Oncological Diseases)
Show Figures

Figure 1

26 pages, 4080 KiB  
Review
DNA Repair Enzyme Poly(ADP-Ribose) Polymerase 1/2 (PARP1/2)-Targeted Nuclear Imaging and Radiotherapy
by Nghia T. Nguyen, Anna Pacelli, Michael Nader and Susanne Kossatz
Cancers 2022, 14(5), 1129; https://doi.org/10.3390/cancers14051129 - 23 Feb 2022
Cited by 6 | Viewed by 3007
Abstract
Since it was discovered that many tumor types are vulnerable to inhibition of the DNA repair machinery, research towards efficient and selective inhibitors has accelerated. Amongst other enzymes, poly(ADP-ribose)-polymerase 1 (PARP1) was identified as a key player in this process, which resulted in [...] Read more.
Since it was discovered that many tumor types are vulnerable to inhibition of the DNA repair machinery, research towards efficient and selective inhibitors has accelerated. Amongst other enzymes, poly(ADP-ribose)-polymerase 1 (PARP1) was identified as a key player in this process, which resulted in the development of selective PARP inhibitors (PARPi) as anti-cancer drugs. Most small molecule PARPi’s exhibit high affinity for both PARP1 and PARP2. PARPi are under clinical investigation for mono- and combination therapy in several cancer types and five PARPi are now clinically approved. In parallel, radiolabeled PARPi have emerged for non-invasive imaging of PARP1 expression. PARP imaging agents have been suggested as companion diagnostics, patient selection, and treatment monitoring tools to improve the outcome of PARPi therapy, but also as stand-alone diagnostics. We give a comprehensive overview over the preclinical development of PARP imaging agents, which are mostly based on the PARPi olaparib, rucaparib, and recently also talazoparib. We also report on the current status of clinical translation, which involves a growing number of early phase trials. Additionally, this work provides an insight into promising approaches of PARP-targeted radiotherapy based on Auger and α-emitting isotopes. Furthermore, the review covers synthetic strategies for PARP-targeted imaging and therapy agents that are compatible with large scale production and clinical translation. Full article
(This article belongs to the Special Issue Radiopharmaceuticals for Oncological Diseases)
Show Figures

Figure 1

24 pages, 467 KiB  
Review
Radiolabeled PSMA Inhibitors
by Oliver C. Neels, Klaus Kopka, Christos Liolios and Ali Afshar-Oromieh
Cancers 2021, 13(24), 6255; https://doi.org/10.3390/cancers13246255 - 13 Dec 2021
Cited by 21 | Viewed by 4867
Abstract
PSMA has shown to be a promising target for diagnosis and therapy (theranostics) of prostate cancer. We have reviewed developments in the field of radio- and fluorescence-guided surgery and targeted photodynamic therapy as well as multitargeting PSMA inhibitors also addressing albumin, GRPr and [...] Read more.
PSMA has shown to be a promising target for diagnosis and therapy (theranostics) of prostate cancer. We have reviewed developments in the field of radio- and fluorescence-guided surgery and targeted photodynamic therapy as well as multitargeting PSMA inhibitors also addressing albumin, GRPr and integrin αvβ3. An overview of the regulatory status of PSMA-targeting radiopharmaceuticals in the USA and Europe is also provided. Technical and quality aspects of PSMA-targeting radiopharmaceuticals are described and new emerging radiolabeling strategies are discussed. Furthermore, insights are given into the production, application and potential of alternatives beyond the commonly used radionuclides for radiolabeling PSMA inhibitors. An additional refinement of radiopharmaceuticals is required in order to further improve dose-limiting factors, such as nephrotoxicity and salivary gland uptake during endoradiotherapy. The improvement of patient treatment achieved by the advantageous combination of radionuclide therapy with alternative therapies is also a special focus of this review. Full article
(This article belongs to the Special Issue Radiopharmaceuticals for Oncological Diseases)
15 pages, 3519 KiB  
Review
New Radionuclides and Technological Advances in SPECT and PET Scanners
by Nicholas P. van der Meulen, Klaus Strobel and Thiago Viana Miranda Lima
Cancers 2021, 13(24), 6183; https://doi.org/10.3390/cancers13246183 - 08 Dec 2021
Cited by 17 | Viewed by 4677
Abstract
Developments throughout the history of nuclear medicine have involved improvements in both instrumentation and radionuclides, which have been intertwined. Instrumentation developments always occurred during the search to improving devices’ sensitivity and included advances in detector technology (with the introduction of cadmium zinc telluride [...] Read more.
Developments throughout the history of nuclear medicine have involved improvements in both instrumentation and radionuclides, which have been intertwined. Instrumentation developments always occurred during the search to improving devices’ sensitivity and included advances in detector technology (with the introduction of cadmium zinc telluride and digital Positron Emission Tomography—PET-devices with silicon photomultipliers), design (total body PET) and configuration (ring-shaped, Single-Photon Emission Computed Tomography (SPECT), Compton camera). In the field of radionuclide development, we observed the continual changing of clinically used radionuclides, which is sometimes influenced by instrumentation technology but also driven by availability, patient safety and clinical questions. Some areas, such as tumour imaging, have faced challenges when changing radionuclides based on availability, when this produced undesirable clinical findings with the introduction of unclear focal uptakes and unspecific uptakes. On the other end of spectrum, further developments of PET technology have seen a resurgence in its use in nuclear cardiology, with rubidium-82 from strontium-82/rubidium-82 generators being the radionuclide of choice, moving away from SPECT nuclides thallium-201 and technetium-99m. These continuing improvements in both instrumentation and radionuclide development have helped the growth of nuclear medicine and its importance in the ever-evolving range of patient care options. Full article
(This article belongs to the Special Issue Radiopharmaceuticals for Oncological Diseases)
Show Figures

Figure 1

18 pages, 3942 KiB  
Review
It’s Time to Shift the Paradigm: Translation and Clinical Application of Non-αvβ3 Integrin Targeting Radiopharmaceuticals
by Susanne Kossatz, Ambros Johannes Beer and Johannes Notni
Cancers 2021, 13(23), 5958; https://doi.org/10.3390/cancers13235958 - 26 Nov 2021
Cited by 6 | Viewed by 3099
Abstract
For almost the entire period of the last two decades, translational research in the area of integrin-targeting radiopharmaceuticals was strongly focused on the subtype αvβ3, owing to its expression on endothelial cells and its well-established role as a biomarker for, and promoter of, [...] Read more.
For almost the entire period of the last two decades, translational research in the area of integrin-targeting radiopharmaceuticals was strongly focused on the subtype αvβ3, owing to its expression on endothelial cells and its well-established role as a biomarker for, and promoter of, angiogenesis. Despite a large number of translated tracers and clinical studies, a clinical value of αvβ3-integrin imaging could not be defined yet. The focus of research has, thus, been moving slowly but steadily towards other integrin subtypes which are involved in a large variety of tumorigenic pathways. Peptidic and non-peptidic radioligands for the integrins α5β1, αvβ6, αvβ8, α6β1, α6β4, α3β1, α4β1, and αMβ2 were first synthesized and characterized preclinically. Some of these compounds, targeting the subtypes αvβ6, αvβ8, and α6β1/β4, were subsequently translated into humans during the last few years. αvβ6-Integrin has arguably attracted most attention because it is expressed by some of the cancers with the worst prognosis (above all, pancreatic ductal adenocarcinoma), which substantiates a clinical need for the respective theranostic agents. The receptor furthermore represents a biomarker for malignancy and invasiveness of carcinomas, as well as for fibrotic diseases, such as idiopathic pulmonary fibrosis (IPF), and probably even for Sars-CoV-2 (COVID-19) related syndromes. Accordingly, the largest number of recent first-in-human applications has been reported for radiolabeled compounds targeting αvβ6-integrin. The results indicate a substantial clinical value, which might lead to a paradigm change and trigger the replacement of αvβ3 by αvβ6 as the most popular integrin in theranostics. Full article
(This article belongs to the Special Issue Radiopharmaceuticals for Oncological Diseases)
Show Figures

Figure 1

17 pages, 2152 KiB  
Review
In Vivo Targeting of CXCR4—New Horizons
by Margret Schottelius, Ken Herrmann and Constantin Lapa
Cancers 2021, 13(23), 5920; https://doi.org/10.3390/cancers13235920 - 25 Nov 2021
Cited by 21 | Viewed by 2707
Abstract
Given its pre-eminent role in the context of tumor cell growth as well as metastasis, the C-X-C motif chemokine receptor 4 (CXCR4) has attracted a lot of interest in the field of nuclear oncology, and clinical evidence on the high potential of CXCR4-targeted [...] Read more.
Given its pre-eminent role in the context of tumor cell growth as well as metastasis, the C-X-C motif chemokine receptor 4 (CXCR4) has attracted a lot of interest in the field of nuclear oncology, and clinical evidence on the high potential of CXCR4-targeted theranostics is constantly accumulating. Additionally, since CXCR4 also represents a key player in the orchestration of inflammatory responses to inflammatory stimuli, based on its expression on a variety of pro- and anti-inflammatory immune cells (e.g., macrophages and T-cells), CXCR4-targeted inflammation imaging has recently gained considerable attention. Therefore, after briefly summarizing the current clinical status quo of CXCR4-targeted theranostics in cancer, this review primarily focuses on imaging of a broad spectrum of inflammatory diseases via the quantification of tissue infiltration with CXCR4-expressing immune cells. An up-to-date overview of the ongoing preclinical and clinical efforts to visualize inflammation and its resolution over time is provided, and the predictive value of the CXCR4-associated imaging signal for disease outcome is discussed. Since the sensitivity and specificity of CXCR4-targeted immune cell imaging greatly relies on the availability of suitable, tailored imaging probes, recent developments in the field of CXCR4-targeted imaging agents for various applications are also addressed. Full article
(This article belongs to the Special Issue Radiopharmaceuticals for Oncological Diseases)
Show Figures

Figure 1

17 pages, 4668 KiB  
Review
Update on Preclinical Development and Clinical Translation of Cholecystokinin-2 Receptor Targeting Radiopharmaceuticals
by Elisabeth von Guggenberg, Petra Kolenc, Christof Rottenburger, Renata Mikołajczak and Alicja Hubalewska-Dydejczyk
Cancers 2021, 13(22), 5776; https://doi.org/10.3390/cancers13225776 - 18 Nov 2021
Cited by 12 | Viewed by 2808
Abstract
The cholecystokinin-2 receptor (CCK2R) has been a target of interest for molecular imaging and targeted radionuclide therapy for two decades. However, so far CCK2R targeted imaging and therapy has not been introduced in clinical practice. Within this review the recent radiopharmaceutical development of [...] Read more.
The cholecystokinin-2 receptor (CCK2R) has been a target of interest for molecular imaging and targeted radionuclide therapy for two decades. However, so far CCK2R targeted imaging and therapy has not been introduced in clinical practice. Within this review the recent radiopharmaceutical development of CCK2R targeting compounds and the ongoing clinical trials are presented. Currently, new gastrin derivatives as well as nonpeptidic substances are being developed to improve the properties for clinical use. A team of specialists from the field of radiopharmacy and nuclear medicine reviewed the available literature and summarized their own experiences in the development and clinical testing of CCK2R targeting radiopharmaceuticals. The recent clinical trials with novel radiolabeled minigastrin analogs demonstrate the potential for both applications, imaging as well as targeted radiotherapy, and reinforce the clinical applicability within a theranostic concept. The intense efforts in optimizing CCK2R targeting radiopharmaceuticals has led to new substances for clinical use, as shown in first imaging studies in patients with advanced medullary thyroid cancer. The first clinical results suggest that the wider clinical implication of CCK2R-targeted radiopharmaceuticals is reasonable. Full article
(This article belongs to the Special Issue Radiopharmaceuticals for Oncological Diseases)
Show Figures

Graphical abstract

20 pages, 3144 KiB  
Review
Radiolabeled Bombesin Analogs
by Rosalba Mansi, Berthold A. Nock, Simone U. Dalm, Martijn B. Busstra, Wytske M. van Weerden and Theodosia Maina
Cancers 2021, 13(22), 5766; https://doi.org/10.3390/cancers13225766 - 17 Nov 2021
Cited by 36 | Viewed by 2791
Abstract
The gastrin-releasing peptide receptor (GRPR) is expressed in high numbers in a variety of human tumors, including the frequently occurring prostate and breast cancers, and therefore provides the rationale for directing diagnostic or therapeutic radionuclides on cancer lesions after administration of anti-GRPR peptide [...] Read more.
The gastrin-releasing peptide receptor (GRPR) is expressed in high numbers in a variety of human tumors, including the frequently occurring prostate and breast cancers, and therefore provides the rationale for directing diagnostic or therapeutic radionuclides on cancer lesions after administration of anti-GRPR peptide analogs. This concept has been initially explored with analogs of the frog 14-peptide bombesin, suitably modified at the N-terminus with a number of radiometal chelates. Radiotracers that were selected for clinical testing revealed inherent problems associated with these GRPR agonists, related to low metabolic stability, unfavorable abdominal accumulation, and adverse effects. A shift toward GRPR antagonists soon followed, with safer analogs becoming available, whereby, metabolic stability and background clearance issues were gradually improved. Clinical testing of three main major antagonist types led to promising outcomes, but at the same time brought to light several limitations of this concept, partly related to the variation of GRPR expression levels across cancer types, stages, previous treatments, and other factors. Currently, these parameters are being rigorously addressed by cell biologists, chemists, nuclear medicine physicians, and other discipline practitioners in a common effort to make available more effective and safe state-of-the-art molecular tools to combat GRPR-positive tumors. In the present review, we present the background, current status, and future perspectives of this endeavor. Full article
(This article belongs to the Special Issue Radiopharmaceuticals for Oncological Diseases)
Show Figures

Figure 1

12 pages, 1810 KiB  
Review
Radioligands Targeting Fibroblast Activation Protein (FAP)
by Thomas Lindner, Frederik L. Giesel, Clemens Kratochwil and Sebastian E. Serfling
Cancers 2021, 13(22), 5744; https://doi.org/10.3390/cancers13225744 - 16 Nov 2021
Cited by 30 | Viewed by 7466
Abstract
Targeting fibroblast activation protein (FAP) in cancer-associated fibroblasts (CAFs) has attracted significant attention in nuclear medicine. Since these cells are present in most cancerous tissues and FAP is rarely expressed in healthy tissues, anti-FAP tracers have a potential as pan-tumor agents. Compared to [...] Read more.
Targeting fibroblast activation protein (FAP) in cancer-associated fibroblasts (CAFs) has attracted significant attention in nuclear medicine. Since these cells are present in most cancerous tissues and FAP is rarely expressed in healthy tissues, anti-FAP tracers have a potential as pan-tumor agents. Compared to the standard tumor tracer [18F]FDG, these tracers show better tumor-to-background ratios (TBR) in many indications. Unlike [18F]FDG, FAP-targeted tracers do not require exhausting preparations, such as dietary restrictions on the part of the patient, and offer the possibility of radioligand therapy (RLT) in a theragnostic approach. Although a radiolabeled antibody was clinically investigated as early as the 1990s, the breakthrough event for FAP-targeting in nuclear medicine was the introduction and clinical application of the so-called FAPI-tracers in 2018. From then, the development and application of FAP-targeted tracers became hot topics for the radiopharmaceutical and nuclear medicine community, and attracted the interest of pharmaceutical companies. The aim of this review is to provide a comprehensive overview of the development of FAP-targeted radiopharmaceuticals and their application in nuclear medicine. Full article
(This article belongs to the Special Issue Radiopharmaceuticals for Oncological Diseases)
Show Figures

Figure 1

Other

Jump to: Research, Review

1 pages, 165 KiB  
Correction
Correction: Sankaranarayanan et al. Auger Emitter Conjugated PARP Inhibitor for Therapy in Triple Negative Breast Cancers: A Comparative In-Vitro Study. Cancers 2022, 14, 230
by Ramya Ambur Sankaranarayanan, Jennifer Peil, Andreas T. J. Vogg, Carsten Bolm, Steven Terhorst, Arno Classen, Matthias Bauwens, Jochen Maurer, Felix Mottaghy and Agnieszka Morgenroth
Cancers 2023, 15(9), 2641; https://doi.org/10.3390/cancers15092641 - 06 May 2023
Viewed by 839
Abstract
The authors wish to replace the ‘Author Contributions’ statement and the affiliation for Jochen Maurer of this article [...] Full article
(This article belongs to the Special Issue Radiopharmaceuticals for Oncological Diseases)
Back to TopTop