Targeted Radionuclide Therapy

A special issue of Pharmaceutics (ISSN 1999-4923). This special issue belongs to the section "Drug Targeting and Design".

Deadline for manuscript submissions: closed (10 November 2021) | Viewed by 61379

Special Issue Editor


E-Mail Website
Guest Editor
Department of Radiology and Nuclear Medicine, Erasmus MC, University Medical Center Rotterdam, Wytemaweg 80, 3015 CN Rotterdam, The Netherlands
Interests: nuclear medicine; theranostics; molecular imaging; bioorthogonal chemistry
Special Issues, Collections and Topics in MDPI journals

Special Issue Information

Dear Colleagues,

Identification of the molecular alterations involved in carcinogenesis has opened up new opportunities for personalized treatment of cancer patients, such as targeted radionuclide therapy. From the early 1990s, targeted radionuclide therapy has received tremendous attention due to the encouraging (pre)clinical results obtained with radiotherapeutics combining a targeting vector (i.e., small molecules, peptides, antibodies) and a cytotoxic radionuclide. A broad variety of radiotherapeutics has been successfully developed to eradicate specifically cancer cells, leading to the recent FDA and EMA registration of a radiolabeled drug for the treatment of neuroendocrine tumors; but many other novel radiopharmaceuticals hold enormous potential for future applications. However, challenges remain and can eventually be overcome by diverse strategies. Hence, the development of new generation of radiopharmaceuticals with improved therapeutic efficacy holds great promise. Production of novel therapeutic radionuclide, targeted alpha therapy (TAT), modification of the pharmacokinetic profile of the radiotherapeutics, bioorthogonal labeling and pretargeting are examples of major research directions, which will be highlighted in this Special Issue of Pharmaceutics on "Targeted Radionuclide Therapy". Original research papers, communications, or review articles are welcome for this Special Issue.

Dr. Yann Seimbille
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. 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
  • Theranostics
  • Cancer
  • Labeling chemistry
  • In vitro and in vivo characterization
  • Targeted alpha therapy
  • Pretargeting
  • Dosimetry
  • Radiobiology

Published Papers (17 papers)

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

Research

Jump to: Review

11 pages, 2661 KiB  
Communication
Short-Interval, Low-Dose Peptide Receptor Radionuclide Therapy in Combination with PD-1 Checkpoint Immunotherapy Induces Remission in Immunocompromised Patients with Metastatic Merkel Cell Carcinoma
by Alexandra Aicher, Anca Sindrilaru, Diana Crisan, Wolfgang Thaiss, Jochen Steinacker, Meinrad Beer, Thomas Wiegel, Karin Scharffetter-Kochanek, Ambros J. Beer and Vikas Prasad
Pharmaceutics 2022, 14(7), 1466; https://doi.org/10.3390/pharmaceutics14071466 - 14 Jul 2022
Cited by 8 | Viewed by 1936
Abstract
Merkel cell carcinoma (MCC) is a neuroendocrine skin cancer of the elderly, with high metastatic potential and poor prognosis. In particular, the primary resistance to immune checkpoint inhibitors (ICI) in metastatic (m)MCC patients represents a challenge not yet met by any efficient treatment [...] Read more.
Merkel cell carcinoma (MCC) is a neuroendocrine skin cancer of the elderly, with high metastatic potential and poor prognosis. In particular, the primary resistance to immune checkpoint inhibitors (ICI) in metastatic (m)MCC patients represents a challenge not yet met by any efficient treatment modality. Herein, we describe a novel therapeutic concept with short-interval, low-dose 177Lutetium (Lu)-high affinity (HA)-DOTATATE [177Lu]Lu-HA-DOTATATE peptide receptor radionuclide therapy (SILD-PRRT) in combination with PD-1 ICI to induce remission in patients with ICI-resistant mMCC. We report on the initial refractory response of two immunocompromised mMCC patients to the PD-L1 inhibitor avelumab. After confirming the expression of somatostatin receptors (SSTR) on tumor cells by [68Ga]Ga-HA-DOTATATE-PET/CT (PET/CT), we employed low-dose PRRT (up to six treatments, mean activity 3.5 GBq per cycle) at 3–6 weeks intervals in combination with the PD-1 inhibitor pembrolizumab to restore responsiveness to ICI. This combination enabled the synergistic application of PD-1 checkpoint immunotherapy with low-dose PRRT at more frequent intervals, and was very well tolerated by both patients. PET/CTs demonstrated remarkable responses at all metastatic sites (lymph nodes, distant skin, and bones), which were maintained for 3.6 and 4.8 months, respectively. Both patients eventually succumbed with progressive disease after 7.7 and 8 months, respectively, from the start of treatment with SILD-PRRT and pembrolizumab. We demonstrate that SILD-PRRT in combination with pembrolizumab is safe and well-tolerated, even in elderly, immunocompromised mMCC patients. The restoration of clinical responses in ICI-refractory patients as proposed here could potentially be used not only for patients with mMCC, but many other cancer types currently treated with PD-1/PD-L1 inhibitors. Full article
(This article belongs to the Special Issue Targeted Radionuclide Therapy)
Show Figures

Figure 1

15 pages, 2482 KiB  
Article
Comparison of Choi, RECIST and Somatostatin Receptor PET/CT Based Criteria for the Evaluation of Response and Response Prediction to PRRT
by Kevin Zwirtz, Juliane Hardt, Güliz Acker, Alexander D. J. Baur, Marianne Pavel, Kai Huang, Winfried Brenner and Vikas Prasad
Pharmaceutics 2022, 14(6), 1278; https://doi.org/10.3390/pharmaceutics14061278 - 16 Jun 2022
Cited by 8 | Viewed by 2135
Abstract
Aim: The most suitable method for assessment of response to peptide receptor radionuclide therapy (PRRT) of neuroendocrine tumors (NET) is still under debate. In this study we aimed to compare size (RECIST 1.1), density (Choi), Standardized Uptake Value (SUV) and a newly defined [...] Read more.
Aim: The most suitable method for assessment of response to peptide receptor radionuclide therapy (PRRT) of neuroendocrine tumors (NET) is still under debate. In this study we aimed to compare size (RECIST 1.1), density (Choi), Standardized Uptake Value (SUV) and a newly defined ZP combined parameter derived from Somatostatin Receptor (SSR) PET/CT for prediction of both response to PRRT and overall survival (OS). Material and Methods: Thirty-four NET patients with progressive disease (F:M 23:11; mean age 61.2 y; SD ± 12) treated with PRRT using either Lu-177 DOTATOC or Lu-177 DOTATATE and imaged with Ga-68 SSR PET/CT approximately 10–12 weeks prior to and after each treatment cycle were retrospectively analyzed. Median duration of follow-up after the first cycle was 63.9 months (range 6.2–86.2). A total of 77 lesions (2–8 per patient) were analyzed. Response assessment was performed according to RECIST 1.1, Choi and modified EORTC (MORE) criteria. In addition, a new parameter named ZP, the product of Hounsfield unit (HU) and SUVmean (Standard Uptake Value) of a tumor lesion, was tested. Further, SUV values (max and mean) of the tumor were normalized to SUV of normal liver parenchyma. Tumor response was defined as CR, PR, or SD. Gold standard for comparison of baseline parameters for prediction of response of individual target lesions to PRRT was change in size of lesions according to RECIST 1.1. For prediction of overall survival, the response after the first and second PRRT were tested. Results: Based on RECIST 1.1, Choi, MORE, and ZP, 85.3%, 64.7%, 61.8%, and 70.6% achieved a response whereas 14.7%, 35.3%, 38.2%, and 29.4% demonstrated PD (progressive disease), respectively. Baseline ZP and ZPnormalized were found to be the only parameters predictive of lesion progression after three PRRT cycles (AUC ZP 0.753; 95% CI 0.6–0.9, p 0.037; AUC ZPnormalized 0.766; 95% CI 0.6–0.9; p 0.029). Based on a cut-off-value of 1201, ZP achieved a sensitivity of 86% and a specificity of 67%, while ZPnormalized reached a sensitivity of 86% and a specificity of 76% at a cut-off-value of 198. Median OS in the total cohort was not reached. In univariate analysis amongst all parameters, only patients having progressive disease according to MORE after the second cycle of PRRT were found to have significantly shorter overall survival (median OS in objective responders not reached, in PD 29.2 months; p 0.015). Patients progressive after two cycles of PRRT according to ZP had shorter OS compared to those responding (median OS for responders not reached, for PD 47.2 months, p 0.066). Conclusions: In this explorative study, we showed that Choi, RECIST 1.1, and SUVmax-based response evaluation varied significantly from each other. Only patients showing progressive disease after two PRRT cycles according to MORE criteria had a worse prognosis while baseline ZP and ZPnormalized performed best in predicting lesion progression after three cycles of PRRT. Full article
(This article belongs to the Special Issue Targeted Radionuclide Therapy)
Show Figures

Figure 1

15 pages, 2617 KiB  
Article
Evaluation of Aminopolycarboxylate Chelators for Whole-Body Clearance of Free 225Ac: A Feasibility Study to Reduce Unexpected Radiation Exposure during Targeted Alpha Therapy
by Mitsuyoshi Yoshimoto, Yukie Yoshii, Hiroki Matsumoto, Mitsuhiro Shinada, Masashi Takahashi, Chika Igarashi, Fukiko Hihara, Tomoko Tachibana, Ayano Doi, Tatsuya Higashi, Hirofumi Fujii and Kohshin Washiyama
Pharmaceutics 2021, 13(10), 1706; https://doi.org/10.3390/pharmaceutics13101706 - 16 Oct 2021
Cited by 3 | Viewed by 2016
Abstract
Actinium-225 (225Ac) is a promising radionuclide used in targeted alpha therapy (TAT). Although 225Ac labeling of bifunctional chelating ligands is effective, previous in vivo studies reported that free 225Ac can be released from the drugs and that such free [...] Read more.
Actinium-225 (225Ac) is a promising radionuclide used in targeted alpha therapy (TAT). Although 225Ac labeling of bifunctional chelating ligands is effective, previous in vivo studies reported that free 225Ac can be released from the drugs and that such free 225Ac is predominantly accumulated in the liver and could cause unexpected toxicity. To accelerate the clinical development of 225Ac TAT with a variety of drugs, preparing methods to deal with any unexpected toxicity would be valuable. The aim of this study was to evaluate the feasibility of various chelators for reducing and excreting free 225Ac and compare their chemical structures. Nine candidate chelators (D-penicillamine, dimercaprol, Ca-DTPA, Ca-EDTA, CyDTA, GEDTA TTHA, Ca-TTHA, and DO3A) were evaluated in vitro and in vivo. The biodistribution and dosimetry of free 225Ac were examined in mice before an in vivo chelating study. The liver exhibited pronounced 225Ac uptake, with an estimated human absorbed dose of 4.76 SvRBE5/MBq. Aminopolycarboxylate chelators with five and six carboxylic groups, Ca-DTPA and Ca-TTHA, significantly reduced 225Ac retention in the liver (22% and 30%, respectively). Significant 225Ac reductions were observed in the heart and remainder of the body with both Ca-DTPA and Ca-TTHA, and in the lung, kidney, and spleen with Ca-TTHA. In vitro interaction analysis supported the in vivo reduction ability of Ca-DTPA and Ca-TTHA. In conclusion, aminopolycarboxylate chelators with five and six carboxylic groups, Ca-DTPA and Ca-TTHA, were effective for whole-body clearance of free 225Ac. This feasibility study provides useful information for reducing undesirable radiation exposure from free 225Ac. Full article
(This article belongs to the Special Issue Targeted Radionuclide Therapy)
Show Figures

Graphical abstract

16 pages, 3008 KiB  
Article
Development of a Compartmental Pharmacokinetic Model for Molecular Radiotherapy with 131I-CLR1404
by Sara Neira, Araceli Gago-Arias, Isabel Gónzalez-Crespo, Jacobo Guiu-Souto and Juan Pardo-Montero
Pharmaceutics 2021, 13(9), 1497; https://doi.org/10.3390/pharmaceutics13091497 - 17 Sep 2021
Viewed by 2364
Abstract
Pharmacokinetic modeling of the radiopharmaceuticals used in molecular radiotherapy is an important step towards accurate radiation dosimetry of such therapies. In this paper, we present a pharmacokinetic model for CLR1404, a phospholipid ether analog that, labeled with 124I/131I, has emerged [...] Read more.
Pharmacokinetic modeling of the radiopharmaceuticals used in molecular radiotherapy is an important step towards accurate radiation dosimetry of such therapies. In this paper, we present a pharmacokinetic model for CLR1404, a phospholipid ether analog that, labeled with 124I/131I, has emerged as a promising theranostic agent. We follow a systematic approach for the model construction based on a decoupling process applied to previously published experimental data, and using the goodness-of-fit, Sobol’s sensitivity analysis, and the Akaike Information Criterion to construct the optimal form of the model, investigate potential simplifications, and study factor prioritization. This methodology was applied to previously published experimental human time-activity curves for 9 organs. The resulting model consists of 17 compartments involved in the CLR1404 metabolism. Activity dynamics in most tissues are well described by a blood contribution plus a two-compartment system, describing fast and slow uptakes. The model can fit both clinical and pre-clinical kinetic data of 124I/131I. In addition, we have investigated how simple fits (exponential and biexponential) differ from the complete model. Such fits, despite providing a less accurate description of time-activity curves, may be a viable alternative when limited data is available in a practical case. Full article
(This article belongs to the Special Issue Targeted Radionuclide Therapy)
Show Figures

Figure 1

21 pages, 5915 KiB  
Article
Targeted Radionuclide Therapy Using Auger Electron Emitters: The Quest for the Right Vector and the Right Radionuclide
by Malick Bio Idrissou, Alexandre Pichard, Bryan Tee, Tibor Kibedi, Sophie Poty and Jean-Pierre Pouget
Pharmaceutics 2021, 13(7), 980; https://doi.org/10.3390/pharmaceutics13070980 - 29 Jun 2021
Cited by 14 | Viewed by 3002
Abstract
Auger electron emitters (AEEs) are attractive tools in targeted radionuclide therapy to specifically irradiate tumour cells while sparing healthy tissues. However, because of their short range, AEEs need to be brought close to sensitive targets, particularly nuclear DNA, and to a lower extent, [...] Read more.
Auger electron emitters (AEEs) are attractive tools in targeted radionuclide therapy to specifically irradiate tumour cells while sparing healthy tissues. However, because of their short range, AEEs need to be brought close to sensitive targets, particularly nuclear DNA, and to a lower extent, cell membrane. Therefore, radioimmunoconjugates (RIC) have been developed for specific tumour cell targeting and transportation to the nucleus. Herein, we assessed, in A-431CEA-luc and SK-OV-31B9 cancer cells that express low and high levels of HER2 receptors, two 111In-RIC consisting of the anti-HER2 antibody trastuzumab conjugated to NLS or TAT peptides for nuclear delivery. We found that NLS and TAT peptides improved the nuclear uptake of 111In-trastuzumab conjugates, but this effect was limited and non-specific. Moreover, it did not result in a drastic decrease of clonogenic survival. Indium-111 also contributed to non-specific cytotoxicity in vitro due to conversion electrons (30% of the cell killing). Comparison with [125I]I-UdR showed that the energy released in the cell nucleus by increasing the RIC’s nuclear uptake or by choosing an AEE that releases more energy per decay should be 5 to 10 times higher to observe a significant therapeutic effect. Therefore, new Auger-based radiopharmaceuticals need to be developed. Full article
(This article belongs to the Special Issue Targeted Radionuclide Therapy)
Show Figures

Figure 1

16 pages, 3697 KiB  
Article
Radioimmunotheranostic Pair Based on the Anti-HER2 Monoclonal Antibody: Influence of Chelating Agents and Radionuclides on Biological Properties
by Ana Cláudia Camargo Miranda, Sofia Nascimento dos Santos, Leonardo Lima Fuscaldi, Luiza Mascarenhas Balieiro, Maria Helena Bellini, Maria Inês Calil Cury Guimarães and Elaine Bortoleti de Araújo
Pharmaceutics 2021, 13(7), 971; https://doi.org/10.3390/pharmaceutics13070971 - 27 Jun 2021
Cited by 3 | Viewed by 2398
Abstract
The oncogene HER2 is an important molecular target in oncology because it is associated with aggressive disease and the worst prognosis. The development of non-invasive imaging techniques and target therapies using monoclonal antibodies is a rapidly developing field. Thus, this work proposes the [...] Read more.
The oncogene HER2 is an important molecular target in oncology because it is associated with aggressive disease and the worst prognosis. The development of non-invasive imaging techniques and target therapies using monoclonal antibodies is a rapidly developing field. Thus, this work proposes the study of the radioimmunotheranostic pair, [111In]In-DTPA-trastuzumab and [177Lu]Lu-DOTA-trastuzumab, evaluating the influence of the chelating agents and radionuclides on the biological properties of the radioimmunoconjugates (RICs). The trastuzumab was immunoconjugated with the chelators DTPA and DOTA and radiolabeled with [111In]InCl3 and [177Lu]LuCl3, respectively. The stability of the RICs was evaluated in serum, and the immunoreactive and internalization fractions were determined in SK-BR-3 breast cancer cells. The in vivo pharmacokinetics and dosimetry quantification and the ex vivo biodistribution were performed in normal and SK-BR-3 tumor-bearing mice. The data showed that there was no influence of the chelating agents and radionuclides on the immunoreactive and internalization fractions of RICs. In contrast, they influenced the stability of RICs in serum, as well as the pharmacokinetics, dosimetry and biodistribution profiles. Therefore, the results showed that the nature of the chelating agent and radionuclide could influence the biological properties of the radioimmunotheranostic pair. Full article
(This article belongs to the Special Issue Targeted Radionuclide Therapy)
Show Figures

Graphical abstract

17 pages, 6786 KiB  
Article
Comparative Preclinical Evaluation of HER2-Targeting ABD-Fused Affibody® Molecules 177Lu-ABY-271 and 177Lu-ABY-027: Impact of DOTA Position on ABD Domain
by Yongsheng Liu, Anzhelika Vorobyeva, Tianqi Xu, Anna Orlova, Annika Loftenius, Theresa Bengtsson, Per Jonasson, Vladimir Tolmachev and Fredrik Y. Frejd
Pharmaceutics 2021, 13(6), 839; https://doi.org/10.3390/pharmaceutics13060839 - 07 Jun 2021
Cited by 6 | Viewed by 3223
Abstract
Radiolabeled Affibody-based targeting agent 177Lu-ABY-027, a fusion of an anti-HER2 Affibody molecule with albumin binding domain (ABD) site-specifically labeled at the C-terminus, has demonstrated a promising biodistribution profile in mice; binding of the construct to albumin prevents glomerular filtration and significantly reduces [...] Read more.
Radiolabeled Affibody-based targeting agent 177Lu-ABY-027, a fusion of an anti-HER2 Affibody molecule with albumin binding domain (ABD) site-specifically labeled at the C-terminus, has demonstrated a promising biodistribution profile in mice; binding of the construct to albumin prevents glomerular filtration and significantly reduces renal uptake. In this study, we tested the hypothesis that site-specific positioning of the chelator at helix 1 of ABD, at a maximum distance from the albumin binding site, would further increase the strength of binding to albumin and decrease the renal uptake. The new construct, ABY-271 with DOTA conjugated at the back of ABD, has been labelled with 177Lu. Targeting properties of 177Lu-ABY-271 and 177Lu-ABY-027 were compared directly. 177Lu-ABY-271 specifically accumulated in SKOV-3 xenografts in mice. The tumor uptake of 177Lu-ABY-271 exceeded uptake in any other organ 24 h and later after injection. However, the renal uptake of 177Lu-ABY-271 was two-fold higher than the uptake of 177Lu-ABY-027. Thus, the placement of chelator on helix 1 of ABD does not provide desirable reduction of renal uptake. To conclude, minimal modification of the design of Affibody molecules has a strong effect on biodistribution, which cannot be predicted a priori. This necessitates extensive structure-properties relationship studies to find an optimal design of Affibody-based targeting agents for therapy. Full article
(This article belongs to the Special Issue Targeted Radionuclide Therapy)
Show Figures

Graphical abstract

14 pages, 2578 KiB  
Article
Efficacy and Safety of [225Ac]Ac-PSMA-617 Augmented [177Lu]Lu-PSMA-617 Radioligand Therapy in Patients with Highly Advanced mCRPC with Poor Prognosis
by Florian Rosar, Jonas Krause, Mark Bartholomä, Stephan Maus, Tobias Stemler, Ina Hierlmeier, Johannes Linxweiler, Samer Ezziddin and Fadi Khreish
Pharmaceutics 2021, 13(5), 722; https://doi.org/10.3390/pharmaceutics13050722 - 14 May 2021
Cited by 23 | Viewed by 2507
Abstract
The use of 225Ac in prostate-specific membrane antigen (PSMA)-targeted radioligand therapy (RLT), either as monotherapy or in combination with 177Lu, is a promising therapy approach in patients with metastatic castration-resistant prostate carcinoma (mCRPC). In this study, we report the efficacy and [...] Read more.
The use of 225Ac in prostate-specific membrane antigen (PSMA)-targeted radioligand therapy (RLT), either as monotherapy or in combination with 177Lu, is a promising therapy approach in patients with metastatic castration-resistant prostate carcinoma (mCRPC). In this study, we report the efficacy and safety of [225Ac]Ac-PSMA-617 augmented [177Lu]Lu-PSMA-617 RLT in 177Lu-naive mCRPC patients (n = 15) with poor prognosis (presence of visceral metastases, high total tumor burden with diffuse bone metastases or a short PSA doubling time of <2 months). Biochemical (by PSA serum value) and molecular imaging response (by [68Ga]Ga-PSMA-11 PET/CT) was assessed after two cycles of [177Lu]Lu-PSMA-617 RLT, with at least one [225Ac]Ac-PSMA-617 augmentation. In addition, PSA-based progression-free survival (PSA-PFS), overall survival (OS) and toxicity (according to CTCAE) were analyzed. We observed a biochemical- and molecular imaging-based partial remission in 53.3% (8/15) and 66.7% (10/15) of patients, respectively. The median PSA-PFS and OS was 9.1 and 14.8 months, respectively. No serious acute adverse events were recorded. Two out of fifteen patients experienced grade 3 anemia. No other grade 3/4 toxicities were observed. RLT-related xerostomia (grade 1/2) was recorded in 2/15 patients. Our data showed a high clinical efficacy with a favorable side effects profile of [225Ac]Ac-PSMA-617 augmented [177Lu]Lu-PSMA-617 RLT in this highly challenging patient cohort. Full article
(This article belongs to the Special Issue Targeted Radionuclide Therapy)
Show Figures

Figure 1

15 pages, 1821 KiB  
Article
Development of [225Ac]Ac-PSMA-I&T for Targeted Alpha Therapy According to GMP Guidelines for Treatment of mCRPC
by Eline L. Hooijman, Yozlem Chalashkan, Sui Wai Ling, Figen F. Kahyargil, Marcel Segbers, Frank Bruchertseifer, Alfred Morgenstern, Yann Seimbille, Stijn L. W. Koolen, Tessa Brabander and Erik de Blois
Pharmaceutics 2021, 13(5), 715; https://doi.org/10.3390/pharmaceutics13050715 - 13 May 2021
Cited by 27 | Viewed by 7676
Abstract
Recently, promising results of the antitumor effects were observed in patients with metastatic castration-resistant prostate cancer treated with 177Lu-labeled PSMA-ligands. Radionuclide therapy efficacy may even be improved by using the alpha emitter Ac-225. Higher efficacy is claimed due to high linear energy [...] Read more.
Recently, promising results of the antitumor effects were observed in patients with metastatic castration-resistant prostate cancer treated with 177Lu-labeled PSMA-ligands. Radionuclide therapy efficacy may even be improved by using the alpha emitter Ac-225. Higher efficacy is claimed due to high linear energy transfer specifically towards PSMA positive cells, causing more double-strand breaks. This study aims to manufacture [225Ac]Ac-PSMA-I&T according to good manufacturing practice guidelines for the translation of [225Ac]Ac-PSMA-I&T into a clinical phase 1 dose escalation study. Quencher addition during labeling was investigated. Quality control of [225Ac]Ac-PSMA-I&T was based on measurement of Fr-221 (218 keV), in equilibrium with Ac-225 in approximately six half-lives of Fr-221 (T½ = 4.8 min). Radio-(i)TLC methods were utilized for identification of the different radiochemical forms, gamma counter for concentration determination, and HPGe-detector for the detection of the radiochemical yield. Radiochemical purity was determined by HPLC. The final patient dose was prepared and diluted with an optimized concentration of quenchers as during labeling, with an activity of 8–12 MBq (±5%), pH > 5.5, 100 ± 20 μg/dose, PSMA-I&T, radiochemical yield >95%, radiochemical purity >90% (up to 3 h), endotoxin levels of <5 EU/mL, osmolarity of 2100 mOsmol, and is produced according to current guidelines. The start of the phase I dose escalation study is planned in the near future. Full article
(This article belongs to the Special Issue Targeted Radionuclide Therapy)
Show Figures

Figure 1

11 pages, 1695 KiB  
Article
Competition (‘Steal’ Phenomenon) between [68Ga]Ga-PSMA-11 Uptake in Prostate Tumor Tissue Versus Healthy Tissue
by Esmée C. A. van der Sar, Bart de Keizer, Marnix G. E. H. Lam and Arthur J. A. T. Braat
Pharmaceutics 2021, 13(5), 699; https://doi.org/10.3390/pharmaceutics13050699 - 11 May 2021
Cited by 2 | Viewed by 1431
Abstract
We aimed to clarify whether a steal ‘phenomenon’ exists by investigating if uptake of ‘prostate specific membrane antigen’ (PSMA) in prostate tumor tissue correlates with the uptake in healthy tissue. Patients with prostate cancer referred for a [68Ga]Ga-PSMA-11 PET/CT were identified [...] Read more.
We aimed to clarify whether a steal ‘phenomenon’ exists by investigating if uptake of ‘prostate specific membrane antigen’ (PSMA) in prostate tumor tissue correlates with the uptake in healthy tissue. Patients with prostate cancer referred for a [68Ga]Ga-PSMA-11 PET/CT were identified retrospectively. Semi-automated quantitative image analysis was performed; fractional healthy tissue [68Ga]Ga-PSMA-11 uptake volume (HT-PSMA (SUV*cm3)) in the lacrimal, submandibular, and parotid glands, and kidneys, and the fractional total lesion [68Ga]Ga-PSMA-11 uptake volume (TL-PSMA (SUV*cm3)) of prostate cancer were used. Ninety-two patients, age 78 ± 8 years, were analyzed. Median TL-PSMA was 703.37 SUV*cm3 (IQR 119.56–2778.20), median HT-PSMA of the lacrimal, submandibular, and parotid glands, and kidneys was: 13.69 (IQR 7.29–19.06), 194.75 (IQR 133.67–276.53), 552.54 (IQR 379.98–737.16), and 8092.75 SUV*cm3 (IQR 5793.02–11,385.86), respectively. A significant (p-value ≤ 0.001) but weak–moderate correlation was found between the TL-PSMA and HT-PSMA of the parotid- and submandibular glands, and kidneys (correlation coefficient of −0.447, −0.345, and −0.394, respectively). No correlation was found between TL-PSMA and HT-PSMA of the lacrimal glands. The existence of a ‘steal’ phenomenon cannot be confirmed in this study. Healthy tissue uptake of [68Ga]Ga-PSMA-11 is only partially influenced by TL-PSMA. Thus, modification of therapeutic PSMA activity should not be adjusted based on TL-PSMA alone. Full article
(This article belongs to the Special Issue Targeted Radionuclide Therapy)
Show Figures

Figure 1

13 pages, 3095 KiB  
Communication
Simultaneous Visualization of 161Tb- and 177Lu-Labeled Somatostatin Analogues Using Dual-Isotope SPECT Imaging
by Francesca Borgna, Patrick Barritt, Pascal V. Grundler, Zeynep Talip, Susan Cohrs, Jan Rijn Zeevaart, Ulli Köster, Roger Schibli, Nicholas P. van der Meulen and Cristina Müller
Pharmaceutics 2021, 13(4), 536; https://doi.org/10.3390/pharmaceutics13040536 - 12 Apr 2021
Cited by 17 | Viewed by 4417
Abstract
The decay of terbium-161 results in the emission of β¯-particles as well as conversion and Auger electrons, which makes terbium-161 interesting for therapeutic purposes. The aim of this study was to use dual-isotope SPECT imaging in order to demonstrate visually that terbium-161 and [...] Read more.
The decay of terbium-161 results in the emission of β¯-particles as well as conversion and Auger electrons, which makes terbium-161 interesting for therapeutic purposes. The aim of this study was to use dual-isotope SPECT imaging in order to demonstrate visually that terbium-161 and lutetium-177 are interchangeable without compromising the pharmacokinetic profile of the radiopharmaceutical. The 161Tb- and 177Lu-labeled somatostatin (SST) analogues DOTATOC (agonist) and DOTA-LM3 (antagonist) were tested in vitro to demonstrate equal properties regarding distribution coefficients and cell uptake into SST receptor-positive AR42J tumor cells. The radiopeptides were further investigated in AR42J tumor-bearing nude mice using the method of dual-isotope (terbium-161/lutetium-177) SPECT/CT imaging to enable the visualization of their distribution profiles in the same animal. Equal pharmacokinetic profiles were demonstrated for either of the two peptides, irrespective of whether it was labeled with terbium-161 or lutetium-177. Moreover, the visualization of the sub-organ distribution confirmed similar behavior of 161Tb- and 177Lu-labeled SST analogues. The data were verified in quantitative biodistribution studies using either type of peptide labeled with terbium-161 or lutetium-177. While the radionuclide did not have an impact on the organ distribution, this study confirmed previous data of a considerably higher tumor uptake of radiolabeled DOTA-LM3 as compared to the radiolabeled DOTATOC. Full article
(This article belongs to the Special Issue Targeted Radionuclide Therapy)
Show Figures

Figure 1

14 pages, 1330 KiB  
Article
One Step Closer to Clinical Translation: Enhanced Tumor Targeting of [99mTc]Tc-DB4 and [111In]In-SG4 in Mice Treated with Entresto
by Panagiotis Kanellopoulos, Aikaterini Kaloudi, Maritina Rouchota, George Loudos, Marion de Jong, Eric P. Krenning, Berthold A. Nock and Theodosia Maina
Pharmaceutics 2020, 12(12), 1145; https://doi.org/10.3390/pharmaceutics12121145 - 26 Nov 2020
Cited by 9 | Viewed by 1892
Abstract
Background: Peptide radioligands may serve as radionuclide carriers to tumor sites overexpressing their cognate receptor for diagnostic or therapeutic purposes. Treatment of mice with the neprilysin (NEP)-inhibitor phosphoramidon was previously shown to improve the metabolic stability and tumor uptake of biodegradable radiopeptides. Aiming [...] Read more.
Background: Peptide radioligands may serve as radionuclide carriers to tumor sites overexpressing their cognate receptor for diagnostic or therapeutic purposes. Treatment of mice with the neprilysin (NEP)-inhibitor phosphoramidon was previously shown to improve the metabolic stability and tumor uptake of biodegradable radiopeptides. Aiming to clinical translation of this methodology, we herein investigated the impact of the approved pill Entresto, releasing the potent NEP-inhibitor LBQ657 in vivo, on the stability and tumor uptake of two radiopeptides. Methods: The metabolic stability of [99mTc]Tc-DB4 (DB4, N4-Pro-Gln-Arg-Tyr-Gly-Asn-Gln-Trp-Ala-Val-Gly-His-Leu-Nle-NH2) and [111In]In-SG4 (SG4, DOTA-DGlu-Ala-Tyr-Gly-Trp-Nle-Asp-Phe-NH2) was tested in LBQ657/Entresto-treated mice vs. untreated controls. The uptake in gastrin-releasing peptide receptor (GRPR)-, or cholecystokinin subtype 2 receptor (CCK2R)-positive tumors respectively, was compared between LBQ657/Entresto-treated mice and untreated controls. Results: LBQ657/Entresto treatment induced marked stabilization of [99mTc] Tc-DB4 and [111In]In-SG4 in peripheral mice blood, resulting in equally enhanced tumor uptake at 4 h post-injection. Accordingly, the [99mTc]Tc-DB4 uptake of 7.13 ± 1.76%IA/g in PC-3 tumors increased to 16.17 ± 0.71/17.50 ± 3.70%IA/g (LBQ657/Entresto) and the [111In]In-SG4 uptake of 3.07 ± 0.87%IA/g in A431-CCK2R(+) tumors to 8.11 ± 1.45/9.61 ± 1.70%IA/g. Findings were visualized by SPECT/CT. Conclusions: This study has shown the efficacy of Entresto to notably improve the profile of [99mTc]Tc-DB4 and [111In]In-SG4 in mice, paving the way for clinical translation of this approach. Full article
(This article belongs to the Special Issue Targeted Radionuclide Therapy)
Show Figures

Figure 1

12 pages, 2747 KiB  
Article
Evaluation of Actinium-225 Labeled Minigastrin Analogue [225Ac]Ac-DOTA-PP-F11N for Targeted Alpha Particle Therapy
by Yun Qin, Stefan Imobersteg, Alain Blanc, Stephan Frank, Roger Schibli, Martin P. Béhé and Michal Grzmil
Pharmaceutics 2020, 12(11), 1088; https://doi.org/10.3390/pharmaceutics12111088 - 12 Nov 2020
Cited by 18 | Viewed by 3138
Abstract
The overexpression of cholecystokinin B receptor (CCKBR) in human cancers led to the development of radiolabeled minigastrin analogues for targeted radionuclide therapy, which aims to deliver cytotoxic radiation specifically to cancer cells. Alpha emitters (e.g., actinium-225) possess high potency in cancer cell-killing and [...] Read more.
The overexpression of cholecystokinin B receptor (CCKBR) in human cancers led to the development of radiolabeled minigastrin analogues for targeted radionuclide therapy, which aims to deliver cytotoxic radiation specifically to cancer cells. Alpha emitters (e.g., actinium-225) possess high potency in cancer cell-killing and hold promise for the treatment of malignant tumors. In these preclinical studies, we developed and evaluated CCKBR-targeted alpha particle therapy. The cellular uptake and cytotoxic effect of actinium-225 labeled and HPLC-purified minigastrin analogue [225Ac]Ac-PP-F11N were characterized in the human squamous cancer A431 cells transfected with CCKBR. Nude mice bearing A431/CCKBR tumors were used for biodistribution and therapy studies followed by histological analysis and SPECT/CT imaging. In vitro, [225Ac]Ac-PP-F11N showed CCKBR-specific and efficient internalization rate and potent cytotoxicity. The biodistribution studies of [225Ac]Ac-PP-F11N revealed CCKBR-specific uptake in tumors, whereas the therapeutic studies demonstrated dose-dependent inhibition of tumor growth and extended mean survival time, without apparent toxicity. The histological analysis of kidney and stomach indicated no severe adverse effects after [225Ac]Ac-PP-F11N administration. The post-therapy SPECT-CT images with [111In]In-PP-F11N confirmed no CCKBR-positive tumor left in the mice with complete remission. In conclusion, our study demonstrates therapeutic efficacy of [225Ac]Ac-PP-F11N without acute radiotoxicity in CCKBR-positive cancer model. Full article
(This article belongs to the Special Issue Targeted Radionuclide Therapy)
Show Figures

Figure 1

Review

Jump to: Research

31 pages, 1085 KiB  
Review
90Y/177Lu-DOTATOC: From Preclinical Studies to Application in Humans
by Licia Uccelli, Alessandra Boschi, Corrado Cittanti, Petra Martini, Stefano Panareo, Eugenia Tonini, Alberto Nieri, Luca Urso, Matteo Caracciolo, Luca Lodi, Aldo Carnevale, Melchiore Giganti and Mirco Bartolomei
Pharmaceutics 2021, 13(9), 1463; https://doi.org/10.3390/pharmaceutics13091463 - 13 Sep 2021
Cited by 19 | Viewed by 3593
Abstract
The PRRT (Peptide Receptor Radionuclide Therapy) is a promising modality treatment for patients with inoperable or metastatic neuroendocrine tumors (NETs). Progression-free survival (PFS) and overall survival (OS) of these patients are favorably comparable with standard therapies. The protagonist in this type of therapy [...] Read more.
The PRRT (Peptide Receptor Radionuclide Therapy) is a promising modality treatment for patients with inoperable or metastatic neuroendocrine tumors (NETs). Progression-free survival (PFS) and overall survival (OS) of these patients are favorably comparable with standard therapies. The protagonist in this type of therapy is a somatostatin-modified peptide fragment ([Tyr3] octreotide), equipped with a specific chelating system (DOTA) capable of creating a stable bond with β-emitting radionuclides, such as yttrium-90 and lutetium-177. In this review, covering twenty five years of literature, we describe the characteristics and performances of the two most used therapeutic radiopharmaceuticals for the NETs radio-treatment: [90Y]Y-DOTATOC and [177Lu]Lu-DOTATOC taking this opportunity to retrace the most significant results that have determined their success, promoting them from preclinical studies to application in humans. Full article
(This article belongs to the Special Issue Targeted Radionuclide Therapy)
Show Figures

Figure 1

50 pages, 2264 KiB  
Review
Overview of the Most Promising Radionuclides for Targeted Alpha Therapy: The “Hopeful Eight”
by Romain Eychenne, Michel Chérel, Férid Haddad, François Guérard and Jean-François Gestin
Pharmaceutics 2021, 13(6), 906; https://doi.org/10.3390/pharmaceutics13060906 - 18 Jun 2021
Cited by 63 | Viewed by 8164
Abstract
Among all existing radionuclides, only a few are of interest for therapeutic applications and more specifically for targeted alpha therapy (TAT). From this selection, actinium-225, astatine-211, bismuth-212, bismuth-213, lead-212, radium-223, terbium-149 and thorium-227 are considered as the most suitable. Despite common general features, [...] Read more.
Among all existing radionuclides, only a few are of interest for therapeutic applications and more specifically for targeted alpha therapy (TAT). From this selection, actinium-225, astatine-211, bismuth-212, bismuth-213, lead-212, radium-223, terbium-149 and thorium-227 are considered as the most suitable. Despite common general features, they all have their own physical characteristics that make them singular and so promising for TAT. These radionuclides were largely studied over the last two decades, leading to a better knowledge of their production process and chemical behavior, allowing for an increasing number of biological evaluations. The aim of this review is to summarize the main properties of these eight chosen radionuclides. An overview from their availability to the resulting clinical studies, by way of chemical design and preclinical studies is discussed. Full article
(This article belongs to the Special Issue Targeted Radionuclide Therapy)
Show Figures

Figure 1

18 pages, 642 KiB  
Review
Combination Therapy, a Promising Approach to Enhance the Efficacy of Radionuclide and Targeted Radionuclide Therapy of Prostate and Breast Cancer
by Tyrillshall S. T. Damiana and Simone U. Dalm
Pharmaceutics 2021, 13(5), 674; https://doi.org/10.3390/pharmaceutics13050674 - 07 May 2021
Cited by 7 | Viewed by 2926
Abstract
In recent years, radionuclide therapy (RT) and targeted radionuclide therapy (TRT) have gained great interest in cancer treatment. This is due to promising results obtained in both preclinical and clinical studies. However, a complete response is achieved in only a small percentage of [...] Read more.
In recent years, radionuclide therapy (RT) and targeted radionuclide therapy (TRT) have gained great interest in cancer treatment. This is due to promising results obtained in both preclinical and clinical studies. However, a complete response is achieved in only a small percentage of patients that receive RT or TRT. As a consequence, there have been several strategies to improve RT and TRT outcomes including the combination of these treatments with other well-established anti-cancer therapies, for example, chemotherapy. Combinations of RT and TRT with other therapies with distinct mechanisms of action represent a promising strategy. As for prostate cancer and breast cancer, the two most prevalent cancer types worldwide, several combination-based therapies have been evaluated. In this review, we will provide an overview of the RT and TRT agents currently used or being investigated in combination with hormone therapy, chemotherapy, immunotherapy, and external beam radiation therapy for the treatment of prostate cancer and breast cancer. Full article
(This article belongs to the Special Issue Targeted Radionuclide Therapy)
Show Figures

Figure 1

25 pages, 3690 KiB  
Review
Bismuth-213 for Targeted Radionuclide Therapy: From Atom to Bedside
by Stephen Ahenkorah, Irwin Cassells, Christophe M. Deroose, Thomas Cardinaels, Andrew R. Burgoyne, Guy Bormans, Maarten Ooms and Frederik Cleeren
Pharmaceutics 2021, 13(5), 599; https://doi.org/10.3390/pharmaceutics13050599 - 21 Apr 2021
Cited by 49 | Viewed by 6274
Abstract
In contrast to external high energy photon or proton therapy, targeted radionuclide therapy (TRNT) is a systemic cancer treatment allowing targeted irradiation of a primary tumor and all its metastases, resulting in less collateral damage to normal tissues. The α-emitting radionuclide bismuth-213 ( [...] Read more.
In contrast to external high energy photon or proton therapy, targeted radionuclide therapy (TRNT) is a systemic cancer treatment allowing targeted irradiation of a primary tumor and all its metastases, resulting in less collateral damage to normal tissues. The α-emitting radionuclide bismuth-213 (213Bi) has interesting properties and can be considered as a magic bullet for TRNT. The benefits and drawbacks of targeted alpha therapy with 213Bi are discussed in this review, covering the entire chain from radionuclide production to bedside. First, the radionuclide properties and production of 225Ac and its daughter 213Bi are discussed, followed by the fundamental chemical properties of bismuth. Next, an overview of available acyclic and macrocyclic bifunctional chelators for bismuth and general considerations for designing a 213Bi-radiopharmaceutical are provided. Finally, we provide an overview of preclinical and clinical studies involving 213Bi-radiopharmaceuticals, as well as the future perspectives of this promising cancer treatment option. Full article
(This article belongs to the Special Issue Targeted Radionuclide Therapy)
Show Figures

Graphical abstract

Back to TopTop