Targeted Radionuclide Tumor Therapy

A special issue of Pharmaceutics (ISSN 1999-4923).

Deadline for manuscript submissions: closed (30 September 2019) | Viewed by 51507

Special Issue Editors


E-Mail Website
Guest Editor
Department of Immunology, Genetics and Pathology (IGP), Dag Hammarskjölds väg 20 Uppsala University, SE-751 85 Uppsala, Sweden
Interests: radionuclide; targeting; imaging; therapy; scaffold proteins; antibody; peptide; pretargeting
Special Issues, Collections and Topics in MDPI journals

E-Mail Website
Guest Editor
Division of Oncology and Pathology, Department of Clinical Sciences, Kamprad Laboratory, Lund University, 22243 Lund, Sweden
Interests: targeted radionuclide therapy; radionuclide molecular imaging; radiolabelling chemistry; scaffold proteins; antibody
Special Issues, Collections and Topics in MDPI journals

Special Issue Information

Dear Colleagues,

In the era of personalized cancer treatment there has been a great emphasis on targeted therapy. Targeted radionuclide therapy represents a promising therapeutic strategy for the treatment of patients with disseminated cancer. The success of targeted radionuclide therapy in malignant lymphomas has proven that this approach is feasible, effective and clinically translatable. However, the full translation of this approach to solid tumours was restricted by low tumour accumulation, normal tissue toxicity and an intrinsically greater radioresistance. There has recently been progress in the treatment of solid tumours with targeted radionuclide therapy, particularly in patients with somatostatin receptor-positive neuroendocrine tumours. This has opened the door to the development of new radionuclide-based targeting agents with therapeutic potential. As stated, the development of new targeted radionuclide therapies and their translation into clinical practice represents a great challenge in the field. This Special Issue has the aim of highlighting current progress in the development of potential targeting agents for targeted radionuclide therapy.

Prof. Vladimir Tolmachev
Dr. Mohamed Altai
Guest Editors

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. Pharmaceutics is an international peer-reviewed open access monthly 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

  • Radionuclide therapy
  • Labeling chemistry
  • Theranostics
  • Dosimetry
  • In vitro and in vivo characterization

Published Papers (11 papers)

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

Research

Jump to: Review

15 pages, 3428 KiB  
Article
A Proof-of-Concept Study on the Therapeutic Potential of Au Nanoparticles Radiolabeled with the Alpha-Emitter Actinium-225
by Evangelia-Alexandra Salvanou, Dimitris Stellas, Charalampos Tsoukalas, Barbara Mavroidi, Maria Paravatou-Petsotas, Nikolaos Kalogeropoulos, Stavros Xanthopoulos, Franck Denat, Gautier Laurent, Rana Bazzi, Stephane Roux and Penelope Bouziotis
Pharmaceutics 2020, 12(2), 188; https://doi.org/10.3390/pharmaceutics12020188 - 21 Feb 2020
Cited by 41 | Viewed by 4193
Abstract
Actinium-225 (225Ac) is receiving increased attention for its application in targeted radionuclide therapy, due to the short range of its emitted alpha particles in conjunction with their high linear energy transfer, which lead to the eradication of tumor cells while sparing [...] Read more.
Actinium-225 (225Ac) is receiving increased attention for its application in targeted radionuclide therapy, due to the short range of its emitted alpha particles in conjunction with their high linear energy transfer, which lead to the eradication of tumor cells while sparing neighboring healthy tissue. The objective of our study was the evaluation of a gold nanoparticle radiolabeled with 225Ac as an injectable radiopharmaceutical form of brachytherapy for local radiation treatment of cancer. Au@TADOTAGA was radiolabeled with 225Ac at pH 5.6 (30 min at 70 °C), and in vitro stability was evaluated. In vitro cytotoxicity was assessed in U-87 MG cancer cells, and in vivo biodistribution was performed by intravenous and intratumoral administration of [225Ac]225Ac-Au@TADOTAGA in U-87 MG tumor-bearing mice. A preliminary study to assess therapeutic efficacy of the intratumorally-injected radio-nanomedicine was performed over a period of 22 days, while the necrotic effect on tumors was evaluated by a histopathology study. We have shown that [225Ac]225Ac-Au@TADOTAGA resulted in the retardation of tumor growth after its intratumoral injection in U87MG tumor-bearing mice, even though very low activities were injected per mouse. This gold nanoparticle radiopharmaceutical could be applied as an unconventional brachytherapy in injectable form for local radiation treatment of cancer. Full article
(This article belongs to the Special Issue Targeted Radionuclide Tumor Therapy)
Show Figures

Graphical abstract

13 pages, 1671 KiB  
Article
Combination of Proton Therapy and Radionuclide Therapy in Mice: Preclinical Pilot Study at the Paul Scherrer Institute
by Cristina Müller, Maria De Prado Leal, Marco D. Dominietto, Christoph A. Umbricht, Sairos Safai, Rosalind L. Perrin, Martina Egloff, Peter Bernhardt, Nicholas P. van der Meulen, Damien C. Weber, Roger Schibli and Antony J. Lomax
Pharmaceutics 2019, 11(9), 450; https://doi.org/10.3390/pharmaceutics11090450 - 02 Sep 2019
Cited by 4 | Viewed by 3349
Abstract
Proton therapy (PT) is a treatment with high dose conformality that delivers a highly-focused radiation dose to solid tumors. Targeted radionuclide therapy (TRT), on the other hand, is a systemic radiation therapy, which makes use of intravenously-applied radioconjugates. In this project, it was [...] Read more.
Proton therapy (PT) is a treatment with high dose conformality that delivers a highly-focused radiation dose to solid tumors. Targeted radionuclide therapy (TRT), on the other hand, is a systemic radiation therapy, which makes use of intravenously-applied radioconjugates. In this project, it was aimed to perform an initial dose-searching study for the combination of these treatment modalities in a preclinical setting. Therapy studies were performed with xenograft mouse models of folate receptor (FR)-positive KB and prostate-specific membrane antigen (PSMA)-positive PC-3 PIP tumors, respectively. PT and TRT using 177Lu-folate and 177Lu-PSMA-617, respectively, were applied either as single treatments or in combination. Monitoring of the mice over nine weeks revealed a similar tumor growth delay after PT and TRT, respectively, when equal tumor doses were delivered either by protons or by β¯-particles, respectively. Combining the methodologies to provide half-dose by either therapy approach resulted in equal (PC-3 PIP tumor model) or even slightly better therapy outcomes (KB tumor model). In separate experiments, preclinical positron emission tomography (PET) was performed to investigate tissue activation after proton irradiation of the tumor. The high-precision radiation delivery of PT was confirmed by the resulting PET images that accurately visualized the irradiated tumor tissue. In this study, the combination of PT and TRT resulted in an additive effect or a trend of synergistic effects, depending on the type of tumor xenograft. This study laid the foundation for future research regarding therapy options in the situation of metastasized solid tumors, where surgery or PT alone are not a solution but may profit from combination with systemic radiation therapy. Full article
(This article belongs to the Special Issue Targeted Radionuclide Tumor Therapy)
Show Figures

Graphical abstract

13 pages, 9360 KiB  
Article
Therapeutic Potential of 47Sc in Comparison to 177Lu and 90Y: Preclinical Investigations
by Klaudia Siwowska, Patrycja Guzik, Katharina A. Domnanich, Josep M. Monné Rodríguez, Peter Bernhardt, Bernard Ponsard, Roger Hasler, Francesca Borgna, Roger Schibli, Ulli Köster, Nicholas P. van der Meulen and Cristina Müller
Pharmaceutics 2019, 11(8), 424; https://doi.org/10.3390/pharmaceutics11080424 - 20 Aug 2019
Cited by 25 | Viewed by 3962
Abstract
Targeted radionuclide therapy with 177Lu- and 90Y-labeled radioconjugates is a clinically-established treatment modality for metastasized cancer. 47Sc is a therapeutic radionuclide that decays with a half-life of 3.35 days and emits medium-energy β-particles. In this study, 47Sc [...] Read more.
Targeted radionuclide therapy with 177Lu- and 90Y-labeled radioconjugates is a clinically-established treatment modality for metastasized cancer. 47Sc is a therapeutic radionuclide that decays with a half-life of 3.35 days and emits medium-energy β-particles. In this study, 47Sc was investigated, in combination with a DOTA-folate conjugate, and compared to the therapeutic properties of 177Lu-folate and 90Y-folate, respectively. In vitro, 47Sc-folate demonstrated effective reduction of folate receptor-positive ovarian tumor cell viability similar to 177Lu-folate, but 90Y-folate was more potent at equal activities due to the higher energy of emitted β-particles. Comparable tumor growth inhibition was observed in mice that obtained the same estimated absorbed tumor dose (~21 Gy) when treated with 47Sc-folate (12.5 MBq), 177Lu-folate (10 MBq), and 90Y-folate (5 MBq), respectively. The treatment resulted in increased median survival of 39, 43, and 41 days, respectively, as compared to 26 days in untreated controls. There were no statistically significant differences among the therapeutic effects observed in treated groups. Histological assessment revealed no severe side effects two weeks after application of the radiofolates, even at double the activity used for therapy. Based on the decay properties and our results, 47Sc is likely to be comparable to 177Lu when employed for targeted radionuclide therapy. It may, therefore, have potential for clinical translation and be of particular interest in tandem with 44Sc or 43Sc as a diagnostic match, enabling the realization of radiotheragnostics in future. Full article
(This article belongs to the Special Issue Targeted Radionuclide Tumor Therapy)
Show Figures

Graphical abstract

20 pages, 4390 KiB  
Article
Evaluation of Tumor-Targeting Properties of an Antagonistic Bombesin Analogue RM26 Conjugated with a Non-Residualizing Radioiodine Label Comparison with a Radiometal-Labelled Counterpart
by Maryam Oroujeni, Ayman Abouzayed, Fanny Lundmark, Bogdan Mitran, Anna Orlova, Vladimir Tolmachev and Ulrika Rosenström
Pharmaceutics 2019, 11(8), 380; https://doi.org/10.3390/pharmaceutics11080380 - 02 Aug 2019
Cited by 5 | Viewed by 4041
Abstract
Radiolabelled antagonistic bombesin analogues are successfully used for targeting of gastrin-releasing peptide receptors (GRPR) that are overexpressed in prostate cancer. Internalization of antagonistic bombesin analogues is slow. We hypothesized that the use of a non-residualizing radioiodine label might not affect the tumour uptake [...] Read more.
Radiolabelled antagonistic bombesin analogues are successfully used for targeting of gastrin-releasing peptide receptors (GRPR) that are overexpressed in prostate cancer. Internalization of antagonistic bombesin analogues is slow. We hypothesized that the use of a non-residualizing radioiodine label might not affect the tumour uptake but would reduce the retention in normal organs, where radiopharmaceutical would be internalized. To test this hypothesis, tyrosine was conjugated via diethylene glycol linker to N-terminus of an antagonistic bombesin analogue RM26 to form Tyr-PEG2-RM26. [111In]In-DOTA-PEG2-RM26 was used as a control with a residualizing label. Tyr-PEG2-RM26 was labelled with 125I with 95% radiochemical purity and retained binding specificity to GRPR. The IC50 values for Tyr-PEG2-RM26 and DOTA-PEG2-RM26 were 1.7 ± 0.3 nM and 3.3 ± 0.5 nM, respectively. The cellular processing of [125I]I-Tyr-PEG2-RM26 by PC-3 cells showed unusually fast internalization. Biodistribution showed that uptake in pancreas and tumour was GRPR-specific for both radioconjugates. Blood clearance of [125I]I-Tyr-PEG2-RM26 was appreciably slower and activity accumulation in all organs was significantly higher than for [111In]In-DOTA-PEG2-RM26. Tumor uptake of [111In]In-DOTA-PEG2-RM26 was significantly higher than for [125I]I-Tyr-PEG2-RM26, resulting in higher tumour-to-organ ratio for [111In]In-DOTA-PEG2-RM26 at studied time points. Incorporation of amino acids with hydrophilic side-chains next to tyrosine might overcome the problems associated with the use of tyrosine as a prosthetic group for radioiodination. Full article
(This article belongs to the Special Issue Targeted Radionuclide Tumor Therapy)
Show Figures

Graphical abstract

18 pages, 4962 KiB  
Article
Synthesis and Preclinical Evaluation of Radio-Iodinated GRPR/PSMA Bispecific Heterodimers for the Theranostics Application in Prostate Cancer
by Ayman Abouzayed, Cheng-Bin Yim, Bogdan Mitran, Sara S. Rinne, Vladimir Tolmachev, Mats Larhed, Ulrika Rosenström and Anna Orlova
Pharmaceutics 2019, 11(7), 358; https://doi.org/10.3390/pharmaceutics11070358 - 23 Jul 2019
Cited by 17 | Viewed by 4835
Abstract
Gastrin-releasing peptide receptor (GRPR) and prostate-specific membrane antigen (PSMA) are overexpressed in most prostate cancers. GRPR expression is higher in early stages while PSMA expression increases with progression. The possibility of targeting both markers with a single theranostics radiotracer could improve patient management. [...] Read more.
Gastrin-releasing peptide receptor (GRPR) and prostate-specific membrane antigen (PSMA) are overexpressed in most prostate cancers. GRPR expression is higher in early stages while PSMA expression increases with progression. The possibility of targeting both markers with a single theranostics radiotracer could improve patient management. Three GRPR/PSMA-targeting bispecific heterodimers (urea derivative PSMA-617 and bombesin-based antagonist RM26 linked via X-triazolyl-Tyr-PEG2, X = PEG2 (BO530), (CH2)8 (BO535), none (BO536)) were synthesized by solid-phase peptide synthesis. Peptides were radio-iodinated and evaluated in vitro for binding specificity, cellular retention, and affinity. In vivo specificity for all heterodimers was studied in PC-3 (GRPR-positive) and LNCaP (PSMA-positive) xenografts. [125I]I-BO530 was evaluated in PC-3pip (GRPR/PSMA-positive) xenografts. Micro single-photon emission computed tomography/computed tomography (microSPECT/CT) scans were acquired. The heterodimers were radiolabeled with high radiochemical yields, bound specifically to both targets, and demonstrated high degree of activity retention in PC-3pip cells. Only [125I]I-BO530 demonstrated in vivo specificity to both targets. A biodistribution study of [125I]I-BO530 in PC-3pip xenografted mice showed high tumor activity uptake (30%–35%ID/g at 3 h post injection (pi)). Activity uptake in tumors was stable and exceeded all other organs 24 h pi. Activity uptake decreased only two-fold 72 h pi. The GRPR/PSMA-targeting heterodimer [125I]I-BO530 is a promising agent for theranostics application in prostate cancer. Full article
(This article belongs to the Special Issue Targeted Radionuclide Tumor Therapy)
Show Figures

Graphical abstract

11 pages, 1788 KiB  
Article
Comparative Radioimmunotherapy of Experimental Melanoma with Novel Humanized Antibody to Melanin Labeled with 213Bismuth and 177Lutetium
by Kevin J. H. Allen, Rubin Jiao, Mackenzie E. Malo, Connor Frank, Darrell R. Fisher, David Rickles and Ekaterina Dadachova
Pharmaceutics 2019, 11(7), 348; https://doi.org/10.3390/pharmaceutics11070348 - 18 Jul 2019
Cited by 16 | Viewed by 3687
Abstract
Melanoma is a cancer with increasing incidence and there is a need for alternatives to immunotherapy within effective approaches to treatment of metastatic melanoma. We performed comparative radioimmunotherapy (RIT) of experimental B16-F10 melanoma with novel humanized IgG to melanin h8C3 labeled with a [...] Read more.
Melanoma is a cancer with increasing incidence and there is a need for alternatives to immunotherapy within effective approaches to treatment of metastatic melanoma. We performed comparative radioimmunotherapy (RIT) of experimental B16-F10 melanoma with novel humanized IgG to melanin h8C3 labeled with a beta emitter, 177Lu, and an alpha-emitter, 213Bi, as well as biodistribution, microSPECT/CT imaging, and mouse and human dosimetry calculations. microSPECT/CT imaging showed that a humanized antibody that targets “free” melanin in the tumor microenvironment had high tumor uptake in B16F10 murine melanoma in C57Bl/6 mice, with little to no uptake in naturally melanized tissues. Extrapolation of the mouse dosimetry data to an adult human demonstrated that doses delivered to major organs and the whole body by 177Lu-h8C3 would be approximately two times higher than those delivered by 213Bi-h8C3, while the doses to the tumor would be almost similar. RIT results indicated that 213Bi-h8C3 was more effective in slowing down the tumor growth than 177Lu-h8C3, while both radiolabeled antibodies did not produce significant hematologic or systemic side effects. We concluded that h8C3 antibody labeled with 213Bi is a promising reagent for translation into a clinical trial in patients with metastatic melanoma. Full article
(This article belongs to the Special Issue Targeted Radionuclide Tumor Therapy)
Show Figures

Graphical abstract

14 pages, 3390 KiB  
Article
Elucidating the Influence of Tumor Presence on the Polymersome Circulation Time in Mice
by Robin M. de Kruijff, René Raavé, Annemarie Kip, Janneke Molkenboer-Kuenen, Stefan J. Roobol, Jeroen Essers, Sandra Heskamp and Antonia G. Denkova
Pharmaceutics 2019, 11(5), 241; https://doi.org/10.3390/pharmaceutics11050241 - 20 May 2019
Cited by 18 | Viewed by 3349
Abstract
The use of nanoparticles as tumor-targeting agents is steadily increasing, and the influence of nanoparticle characteristics such as size and stealthiness have been established for a large number of nanocarrier systems. However, not much is known about the impact of tumor presence on [...] Read more.
The use of nanoparticles as tumor-targeting agents is steadily increasing, and the influence of nanoparticle characteristics such as size and stealthiness have been established for a large number of nanocarrier systems. However, not much is known about the impact of tumor presence on nanocarrier circulation times. This paper reports on the influence of tumor presence on the in vivo circulation time and biodistribution of polybutadiene-polyethylene oxide (PBd-PEO) polymersomes. For this purpose, polymersomes were loaded with the gamma-emitter 111In and administered intravenously, followed by timed ex vivo biodistribution. A large reduction in circulation time was observed for tumor-bearing mice, with a circulation half-life of merely 5 min (R2 = 0.98) vs 117 min (R2 = 0.95) in healthy mice. To determine whether the rapid polymersome clearance observed in tumor-bearing mice was mediated by macrophages, chlodronate liposomes were administered to both healthy and tumor-bearing mice prior to the intravenous injection of radiolabeled polymersomes to deplete their macrophages. Pretreatment with chlodronate liposomes depleted macrophages in the spleen and liver and restored the circulation time of the polymersomes with no significant difference in circulation time between healthy mice and tumor-bearing mice pretreated with clodronate liposomes (15.2 ± 1.2% ID/g and 13.6 ± 2.7% ID/g, respectively, at 4 h p.i. with p = 0.3). This indicates that activation of macrophages due to tumor presence indeed affected polymersome clearance rate. Thus, next to particle design, the presence of a tumor can also greatly impact circulation times and should be taken into account when designing studies to evaluate the distribution of polymersomes. Full article
(This article belongs to the Special Issue Targeted Radionuclide Tumor Therapy)
Show Figures

Graphical abstract

Review

Jump to: Research

21 pages, 1274 KiB  
Review
The Future of PSMA-Targeted Radionuclide Therapy: An Overview of Recent Preclinical Research
by Eline A.M. Ruigrok, Wytske M. van Weerden, Julie Nonnekens and Marion de Jong
Pharmaceutics 2019, 11(11), 560; https://doi.org/10.3390/pharmaceutics11110560 - 29 Oct 2019
Cited by 52 | Viewed by 8424
Abstract
Prostate specific membrane antigen (PSMA) has become a major focus point in the research and development of prostate cancer (PCa) imaging and therapeutic strategies using radiolabeled tracers. PSMA has shown to be an excellent target for PCa theranostics because of its high expression [...] Read more.
Prostate specific membrane antigen (PSMA) has become a major focus point in the research and development of prostate cancer (PCa) imaging and therapeutic strategies using radiolabeled tracers. PSMA has shown to be an excellent target for PCa theranostics because of its high expression on the membrane of PCa cells and the increase in expression during disease progression. Therefore, numerous PSMA-targeting tracers have been developed and (pre)clinically studied with promising results. However, many of these PSMA-targeting tracers show uptake in healthy organs such as the salivary glands, causing radiotoxicity. Furthermore, not all patients respond to PSMA-targeted radionuclide therapy (TRT). This created the necessity of additional preclinical research studies in which existing tracers are reevaluated and new tracers are developed in order to improve PSMA-TRT by protecting the (PSMA-expressing) healthy organs and improving tumor uptake. In this review we will give an overview of the recent preclinical research projects regarding PCa-TRT using PSMA-specific radiotracers, which will give an indication of where the PSMA-TRT research movement is going and what we can expect in future clinical trials. Full article
(This article belongs to the Special Issue Targeted Radionuclide Tumor Therapy)
Show Figures

Graphical abstract

28 pages, 2343 KiB  
Review
The Significance of NK1 Receptor Ligands and Their Application in Targeted Radionuclide Tumour Therapy
by Agnieszka Majkowska-Pilip, Paweł Krzysztof Halik and Ewa Gniazdowska
Pharmaceutics 2019, 11(9), 443; https://doi.org/10.3390/pharmaceutics11090443 - 01 Sep 2019
Cited by 34 | Viewed by 5952
Abstract
To date, our understanding of the Substance P (SP) and neurokinin 1 receptor (NK1R) system shows intricate relations between human physiology and disease occurrence or progression. Within the oncological field, overexpression of NK1R and this SP/NK1R system have been implicated in cancer cell [...] Read more.
To date, our understanding of the Substance P (SP) and neurokinin 1 receptor (NK1R) system shows intricate relations between human physiology and disease occurrence or progression. Within the oncological field, overexpression of NK1R and this SP/NK1R system have been implicated in cancer cell progression and poor overall prognosis. This review focuses on providing an update on the current state of knowledge around the wide spectrum of NK1R ligands and applications of radioligands as radiopharmaceuticals. In this review, data concerning both the chemical and biological aspects of peptide and nonpeptide ligands as agonists or antagonists in classical and nuclear medicine, are presented and discussed. However, the research presented here is primarily focused on NK1R nonpeptide antagonistic ligands and the potential application of SP/NK1R system in targeted radionuclide tumour therapy. Full article
(This article belongs to the Special Issue Targeted Radionuclide Tumor Therapy)
Show Figures

Graphical abstract

25 pages, 2619 KiB  
Review
Therapeutic Applications of Pretargeting
by Marjolein Verhoeven, Yann Seimbille and Simone U. Dalm
Pharmaceutics 2019, 11(9), 434; https://doi.org/10.3390/pharmaceutics11090434 - 01 Sep 2019
Cited by 39 | Viewed by 4972
Abstract
Targeted therapies, such as radioimmunotherapy (RIT), present a promising treatment option for the eradication of tumor lesions. RIT has shown promising results especially for hematologic malignancies, but the therapeutic efficacy is limited by unfavorable tumor-to-background ratios resulting in high radiotoxicity. Pretargeting strategies can [...] Read more.
Targeted therapies, such as radioimmunotherapy (RIT), present a promising treatment option for the eradication of tumor lesions. RIT has shown promising results especially for hematologic malignancies, but the therapeutic efficacy is limited by unfavorable tumor-to-background ratios resulting in high radiotoxicity. Pretargeting strategies can play an important role in addressing the high toxicity profile of RIT. Key to pretargeting is the concept of decoupling the targeting vehicle from the cytotoxic agent and administrating them separately. Studies have shown that this approach has the ability to enhance the therapeutic index as it can reduce side effects caused by off-target irradiation and thereby increase curative effects due to higher tolerated doses. Pretargeted RIT (PRIT) has been explored for imaging and treatment of different cancer types over the years. This review will give an overview of the various targeted therapies in which pretargeting has been applied, discussing PRIT with alpha- and beta-emitters and as part of combination therapy, plus its use in drug delivery systems. Full article
(This article belongs to the Special Issue Targeted Radionuclide Tumor Therapy)
Show Figures

Graphical abstract

23 pages, 1372 KiB  
Review
Beyond the Barrier: Targeted Radionuclide Therapy in Brain Tumors and Metastases
by Janik Puttemans, Tony Lahoutte, Matthias D’Huyvetter and Nick Devoogdt
Pharmaceutics 2019, 11(8), 376; https://doi.org/10.3390/pharmaceutics11080376 - 01 Aug 2019
Cited by 17 | Viewed by 3930
Abstract
Brain tumors are notoriously difficult to treat. The blood-brain barrier provides a sanctuary site where residual and metastatic cancer cells can evade most therapeutic modalities. The delicate nature of the brain further complicates the decision of eliminating as much tumorous tissue as possible [...] Read more.
Brain tumors are notoriously difficult to treat. The blood-brain barrier provides a sanctuary site where residual and metastatic cancer cells can evade most therapeutic modalities. The delicate nature of the brain further complicates the decision of eliminating as much tumorous tissue as possible while protecting healthy tissue. Despite recent advances in immunotherapy, radiotherapy and systemic treatments, prognosis of newly diagnosed patients remains dismal, and recurrence is still a universal problem. Several strategies are now under preclinical and clinical investigation to optimize delivery and maximize the cytotoxic potential of pharmaceuticals with regards to brain tumors. This review provides an overview of targeted radionuclide therapy approaches for the treatment of primary brain tumors and brain metastases, with an emphasis on biological targeting moieties that specifically target key biomarkers involved in cancer development. Full article
(This article belongs to the Special Issue Targeted Radionuclide Tumor Therapy)
Show Figures

Figure 1

Back to TopTop