Next Article in Journal
Prognostic Significance of Gene Expression and DNA Methylation Markers in Circulating Tumor Cells and Paired Plasma Derived Exosomes in Metastatic Castration Resistant Prostate Cancer
Next Article in Special Issue
The Value of Real-World Data in Understanding Prostate Cancer Risk and Improving Clinical Care: Examples from Swedish Registries
Previous Article in Journal
Infant Acute Myeloid Leukemia: A Unique Clinical and Biological Entity
Previous Article in Special Issue
Identification of Germline Genetic Variants that Increase Prostate Cancer Risk and Influence Development of Aggressive Disease
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Preclinical and Clinical Status of PSMA-Targeted Alpha Therapy for Metastatic Castration-Resistant Prostate Cancer

by
Asta Juzeniene
1,*,
Vilde Yuli Stenberg
1,2,3,
Øyvind Sverre Bruland
3,4 and
Roy Hartvig Larsen
2
1
Department of Radiation Biology, Institute for Cancer Research, Norwegian Radium Hospital, Oslo University Hospital, Montebello, 0379 Oslo, Norway
2
Nucligen, Ullernchausséen 64, 0379 Oslo, Norway
3
Institute for Clinical Medicine, University of Oslo, Box 1171 Blindern, 0318 Oslo, Norway
4
Department of Oncology, Norwegian Radium Hospital, Oslo University Hospital, 0379 Oslo, Norway
*
Author to whom correspondence should be addressed.
Submission received: 14 January 2021 / Revised: 7 February 2021 / Accepted: 8 February 2021 / Published: 13 February 2021
(This article belongs to the Collection Prostate Cancer—from Molecular Mechanisms to Clinical Care)

Abstract

:

Simple Summary

Currently, there is no treatment that can cure patients with late stage metastatic prostate cancer. Prostate-specific membrane antigen is a type of protein overexpressed on the membrane surface of most prostate cancer cells. The preclinical and clinical experiences in the rapidly evolving field of targeted alpha-particle radiation therapy for metastatic prostate cancer overexpressing prostate-specific membrane antigen are reviewed. Targeted alpha therapy employs radionuclides emitting highly energetic alpha-particles (cytotoxic payload) chelated to small molecules or monoclonal antibodies designed to target prostate-specific membrane antigen. In this review, we summarize the availability of therapeutic alpha-emitting radionuclides (terbium-149, astatine-211, bismuth-212 (lead-212), bismuth-213, radium-223, actinium-225, thorium-227), and the development of small molecules and antibodies targeting prostate-specific membrane antigen. The limitations in studies using animal models of human prostate cancer to accurately predict efficacy and toxicity in patients are addressed. We have attempted to also critically discuss hurdles related to logistical and supply aspects between different alpha-emitting prostate-specific membrane antigen-targeting radiopharmaceuticals. Lastly, we discuss the potentials, limitations, and future perspectives of prostate-specific membrane antigen-targeted alpha therapy.

Abstract

Bone, lymph node, and visceral metastases are frequent in castrate-resistant prostate cancer patients. Since such patients have only a few months’ survival benefit from standard therapies, there is an urgent need for new personalized therapies. The prostate-specific membrane antigen (PSMA) is overexpressed in prostate cancer and is a molecular target for imaging diagnostics and targeted radionuclide therapy (theragnostics). PSMA-targeted α therapies (PSMA-TAT) may deliver potent and local radiation more selectively to cancer cells than PSMA-targeted β therapies. In this review, we summarize both the recent preclinical and clinical advances made in the development of PSMA-TAT, as well as the availability of therapeutic α-emitting radionuclides, the development of small molecules and antibodies targeting PSMA. Lastly, we discuss the potentials, limitations, and future perspectives of PSMA-TAT.

1. Introduction

Prostate cancer is the second most common cancer in men worldwide, with an estimated 1.3 million new cases and 359,000 deaths in 2018 [1]. The tumors of 10–20% of prostate cancer patients become refractory to androgen deprivation therapy and progress as metastatic castration-resistant prostate cancer (mCRPC) [2,3]. Bone metastases dominate, but lymph node and visceral metastases are also frequent in mCRPC patients [4,5,6]. Treatment options for mCRPC have expanded rapidly in the last 20 years [7,8,9,10,11]. The US Food and Drug Administration (FDA) and the European Medicines Agency (EMA) have introduced docetaxel (chemotherapy, approved in 2004/2007), cabazitaxel (chemotherapy, approved in 2010/2011), sipuleucel-T (autologous immunotherapy, approved in 2010/2013), abiraterone acetate (hormone therapy, approved in 2011/2011), enzalutamide (hormone therapy, approved in 2010/2013), Xofigo (radium-223, targeted alpha therapy, approved in 2013/2013), olaparib (PARP inhibitor therapy, approved in 2020/-), and rucaparib (PARP inhibitor therapy, approved in 2020/-) for the treatment of mCRPC [7,8,9,10,11]. However, mCRPC still remains incurable [11,12,13]. This may partly be explained by the inter-patient and intra-patient heterogeneity of the disease [14,15,16]. There is an urgent need for new personalized, highly effective targeted therapies for these patients. Several cell-surface proteins, such as glycoproteins, have been investigated as targets for the treatment of mCRPC [17,18,19]. The prostate-specific membrane antigen (PSMA), also known as glutamate carboxypeptidase II (GCPII) or folate hydrolase 1 (FOLH1), is one of the cell-surface proteins overexpressed in prostate cancer [20,21,22,23]. PSMA expression correlates with disease progression and Gleason score [20,21,22,24,25,26,27]. PSMA has a large extracellular domain, which can be recognized by antibodies, their fragments, small molecules, nanobodies, and aptamers [28,29]. Additionally, PSMA internalizes the bound targeting molecules and any payload attached to them, making it an excellent molecular target for both diagnostic imaging and targeted therapy, applying a theragnostic approach [24,30,31,32]. Many small molecules and antibodies targeting PSMA have been developed, labeled with β emitters (177Lu, 161Tb, 131I, 90Y, 67Cu, 47Sc), and studied in preclinical and clinical studies [33,34,35,36]. The small molecule radioligands 177Lu-PSMA-617 and 177Lu-PSMA-I&T are already being used in salvage/compassionate therapy in end-stage mCRPC patients [33,37,38]. A recent meta-analysis found a median progression-free survival of 11 months, overall survival of 13.7 months, and low toxicity profile [38]. Treatment of mCRPC patients with 177Lu-PSMA ligands give better therapeutic outcomes and cause fewer adverse effects than third-line treatment with enzalutamide and cabazitaxel [39]. Both radioligands are now being tested in phase 3 trials, VISION (177Lu-PSMA-617, NCT03511664) and SPLASH (177Lu-PSMA-I&T, NCT04647526). However, only 45% of mCRPC patients have a biochemical response (prostate-specific antigen (PSA) decline ≥ 50%) to 177Lu-PSMA-617, and around 30% of patients do not respond at all or are not suitable for this therapy due to diffuse red marrow infiltration [40,41,42]. Additionally, the presence of visceral metastases is associated with poor response and survival outcomes in mCRPC patients treated with 177Lu-PSMA [43]. Lu-177 emits β particles with a maximum energy of 497 keV and an average energy of 133 keV with a maximum soft-tissue penetration of 1.5 mm [44]. Such β particles will give a high absorbed dose to targeted macroscopic tumors while depositing a low absorbed dose to small cell clusters or single metastatic cancer cells because the range of the electrons is too long [45,46]. Preclinical and clinical studies suggest that radionuclide therapy with high linear energy transfer (LET) and short-range α emitters may have advantages over low LET β emitters [47]. PSMA-targeted α therapy (PSMA-TAT) may deliver potent and local radiation more selectively to cancer cells than PSMA-targeted β therapies. In this review, we summarize the recent advances made in preclinical and clinical development of PSMA-TAT, the availability of therapeutic α-emitting radionuclides, the development of small molecules and antibodies targeting PSMA, and discuss the potentials, limitations, and future perspectives of PSMA-TAT.

2. Prostate-Specific Membrane Antigen (PSMA)

PSMA is a transmembrane glycoprotein of about 100 kDa with folate hydrolase, carboxypeptidase, and internalization activities [29]. It contains a transmembrane region of 24 amino acids, an N-terminal cytoplasmatic sequence of 19 amino acids, and a large extracellular domain of 707 amino acids (Figure 1). The extracellular domain of PSMA is highly glycosylated, and glycosylation is required for enzymatic activity [48,49]. Expression of PSMA is low in normal prostate tissue, kidneys, duodenum, salivary and lacrimal glands, brain, and intestines [50] but is increased in prostate cancer [21,23,32]. PSMA expression is correlated with prostate growth and significantly associated with the degree of differentiation and progression of mCRPC [29]. The precise mechanisms of the physiological function of PSMA in prostate cancer and its regulation are unknown. It has been suggested that elevated levels of PSMA enzymatic activity in prostate cancer cells increases cell folate uptake and proliferation and contributes to prostate carcinogenesis and progression [51].

3. Targeted Radionuclide Therapy of Prostate Cancer

Targeted radionuclide therapy (TRT) is a rapidly growing treatment modality for mCRPC [33,55,56]. TRT involves the use of radioisotopes or radiolabeled compounds that either naturally accumulate in or are designed to target and deliver a cytotoxic amount of radiation to prostate cancer cells sparing the surrounding normal tissues [55,56]. The systemic administration of tumor-targeted radiopharmaceuticals allows the simultaneous treatment of wide-spread bone and extraskeletal metastases [56,57], whereas a multiple and large-field irradiation using external beam radiotherapy is too toxic [58]. Currently, the β-emitting radionuclide 177Lu is the most clinically used for TRT [37,38]. However, α particles present significantly higher energies than β particles, which combined with very short path lengths (<0.1 mm) result in high LET and a greater probability of generating DNA double-strand breaks upon interaction with cell nuclei (Table 1). This occurs almost independently of tissue oxygenation, dose rate, and cellular resistance to photon irradiation and chemotherapy [59]. Beta particle irradiation produces mainly single-strand breaks, exhibiting lower cytotoxic potency than α particles (Table 1). Consequently, the use of α emitters allows the specific targeting and killing of individual malignant cells while minimizing the toxicity to surrounding healthy tissue. Therefore, α particles are highly cytotoxic and promising candidates for TRT.

3.1. Radionuclides Used for PSMA-TAT

Ra-223 (Xofigo) is the first-in-class and only α-emitting radiopharmaceutical currently approved by the FDA and the EMA for the treatment of mCRPC patients with symptomatic bone metastases and no known extraskeletal metastatic disease [63,64,65]. Ra-223 has a natural affinity for areas of high bone turnover because it is a bone-seeking calcium mimetic [66]. Targeted 223Ra therapy for mCRPC with its overall survival benefit has revolutionized the field of TRT [67,68]. Regional lymph node and visceral metastases, most frequently in lung and liver, are seen in approximately 8–15% of newly diagnosed patients but increase over time, currently affecting close to half of men during the course of their disease and represent the lethal phase of the disease [69,70]. Extraskeletal metastatic disease is now of growing concern in patients with mCRPC failing chemotherapy and/or the new oral anti-hormonal therapies targeting the androgen receptor axis (abiraterone and enzalutamide) evident by recent developments in diagnostic nuclear medicine; i.e., PSMA-positron emission tomography (PET) imaging [69,70]. An ideal TAT for mCRPC must exhibit a dual mode of action to combat the entire spectrum of metastases, both micrometastases and overt metastatic disease in lymph nodes, the skeleton, and visceral organs. The majority of bone, visceral, and lymph node metastases highly express PSMA [21,22]. Several α-emitting radionuclides, 149Tb, 211At, 212Pb/212Bi, 223Ra, 225Ac, and 227Th, and PSMA targeting agents are now being actively evaluated for PSMA-TAT (Table 2, Table 3, Table 4 and Table 5). The availability, physical and chemical properties of radionuclides play an important role in identifying α-emitting radionuclides suitable for therapy. The half-life of the radionuclide should neither be too long to avoid extended radiation effects after radiopharmaceutical administration nor too short to have enough time for logistical aspects, such as production and transportation.
The radiopharmaceuticals must be sufficiently stable compounds both in vitro and in vivo. The nuclear recoil energy of about 100–200 keV from α-decay is sufficient to break chemical bonds between the targeting moiety and the radionuclide, which can lead to circulating carrier-free daughter radionuclides in blood [114,118,119,120,121]. The decay of the parent radionuclides 212Pb/212Bi, 225Ac, 223Ra, and 227Th form multiple α-emitting daughter radionuclides (Figure 2). The released daughter radionuclide can be retained inside the tumor and enhance the cytotoxic effect, but if the released daughter radionuclide has a long enough half-life, it can redistribute within the body and damage healthy tissues [118,120,122].

3.2. PSMA Targeting Agents in TRT

The extracellular domain makes up to 95% of PSMA and provides an accessible target for small molecules, antibodies, and their fragments [29,80]. A number of PSMA-targeting small molecules (also denoted as ligands or inhibitors) and antibodies have been developed for molecular imaging by single-photon emission computed tomography (SPECT) and PET and/or TRT [29,30,123,124]. Small molecules offer potential advantages over antibodies, such as small size, easy synthesis and modification, high specificity and affinity, good permeability with rapid accumulation in tumor, and rapid clearance from most normal tissues (Table 3).
Many different PSMA ligands have been developed [124,125,126,127], and several of them have been investigated for TAT in preclinical and clinical studies (Table 4 and Table 5). Small molecule PSMA ligands consist of three components: a binding entity (motif), a linker (spacer), and a radiolabel-bearing moiety (a bifunctional chelator for radiolabeling with metal radionuclides or a prosthetic group for astatination) [41,125,126,128]. The linker region is used to adjust the molecular size and polarity to impact the in vivo distribution properties [125,126,127,129,130,131,132,133]. The first small molecule inhibitors targeting PSMA, based on the glutamate-urea-lysine entity (MIP-1072 and MIP-1095), were introduced into the clinic in 2013 for prostate cancer imaging with 123I [134]. These inhibitors showed a rapid advantageous localization in tumor lesions, including soft tissue and bone metastases. In 2014, German research groups from Heidelberg and Munich developed PSMA-11 (also called PSMA-HBED-CC) and PSMA I&T ligands, respectively [135,136]. In 2015, the Heidelberg group developed PSMA-617 [130]. In 2020, the FDA approved 68Ga-PSMA-11 as the first drug for PET imaging of PSMA positive lesions in men with prostate cancer [137]. Unfortunately, the chelator HBED-CC (N,N’-bis [2-hydroxy-5-(carboxyethyl)benzyl] ethylenediamine-N,N’-diacetic acid) used in PSMA-11 is not suitable for the stable complexation with therapeutic radionuclides [138]. To date, PSMA-617 and PSMA I&T are the most commonly used theragnostic PSMA radioligands because they can be radiolabeled with diagnostic, e.g., 68Ga and 44Sc for PET and 111In for SPECT, as well as therapeutic radiometals [138,139,140,141,142,143,144]. In an attempt to prolong circulation in the blood and therewith, to increase the dose delivered to tumors and the tumor-to-kidney ratio, PSMA ligands have been structurally modified by adding albumin-binding moieties [123,132,145,146,147,148,149,150]. Preclinical and clinical studies are required to demonstrate whether PSMA inhibitors with enhanced albumin binding (HTK01169, PSMA-ALB-56, RPS-074, EB-PSMA-617, CTT1403) can increase the efficacy of PSMA-TRT [103,146,148,150,151]. A recently published review provides a comprehensive overview of the current status of selected PSMA inhibitors that have been developed from 1996–2020, emphasizing recent synthetic advances and chemical strategies while highlighting the therapeutic potential and drawbacks of each inhibitor [152].
Several antibodies and their fragments are also being evaluated for TRT in preclinical and clinical studies [97,104,153,154]. The antibodies J591, 107-1A4, PSMA-TTC, and their conjugates recognize extracellular epitopes of PSMA (Figure 1) and are the most investigated for TAT [81,96,97,104,154]. J591, 107-1A4, and PSMA-TTC were described and characterized in 1997 by Liu et al. [155], in 1998 by Brown et al. [156], and in 2020 by Hammer et al. [104], respectively. The half-life of the radionuclide must be matched with the antibody residence time in the tumor to deliver the maximum irradiation dose [157]. Antibodies have a plasma half-life of about 2–7 days, and several days are needed to reach maximum tumor uptake (Table 3). Radionuclides with too short or too long half-lives may lead to suboptimal efficacy or toxicity.

3.3. Radium-223 for PSMA-TAT

Among all α-emitting radionuclides, 223Ra has the most suitable half-life (t1/2 = 11.4 days), decay properties, and safety based on extensive clinical use as Xofigo for TAT. Ra-223 decays via a chain of five short-lived daughter radionuclides (t1/2 from 0.5 s up to 36.1 min) to stable 207Pb, emitting four α particles and two β particles (Figure 2). No redistribution of radioactive daughters has yet been reported [66,158]. Consequently, high decay energy (27–28 MeV) is released over a short range [66,158,159]. Ra-223 can be produced in large quantities from 227Ac (t1/2 = 21.7 years) generator [66]. The half-life of 223Ra provides sufficient time for its preparation, distribution, and administration to patients. Unfortunately, due to the bone-seeking characteristics of 223Ra, its clinical use is limited to prostate cancer patients with osteoblastic metastases [66]. Chelate complex formation is essential to treat extraskeletal cancer metastases. A number of bifunctional chelators, which carry a functional unit for the immobilization of the radiometal and a functional group for the covalent attachment to a biological target molecule, have been tested to chelate 223Ra [160,161]. Unfortunately, 223Ra, like other alkaline earth metals, forms very weak complexes [160,161]. To overcome this limitation, several nanomaterials, hydroxyapatites, polyoxopalladates, nanozeolites, barium sulfate, and titanium dioxide nanoparticles have been tested for stable incorporation of radium and linking to the targeting vector [84,162,163,164,165,166]. So far, only one paper has been published investigating the encapsulation of 223Ra into functionalized nanozeolites for PSMA-TAT [84]. NaA zeolite is one of the synthetic, non-toxic microporous crystalline aluminosilicate zeolites that can accommodate a wide variety of various molecules, including radionuclides [165,167]. Czerwińska et al. [84] modified NaA nanozeolite with silane-PEG (polyethylene glycol) and functionalized it with anti-PSMA D2B antibody as a carrier of 223Ra and its daughter radionuclides for PSMA-TAT. The obtained 120 nm 223RaA–silane-PEG-PSMA D2B bioconjugate was highly stable (<2% of 223Ra and <6% of the daughter radionuclides were released up to 12 days) [84]. The radioimmunoconjugate bound specifically and internalized into PSMA-expressing C4-2 cells, but not into PSMA-negative DU-145 cells. Treatment of C4-2 cells with 20 kBq/mL of the radioimmunoconjugate resulted in an 80% reduction in metabolic activity [84]. Further preclinical studies in vivo are needed to validate the therapeutic efficacy and toxicity of 223RaA-silane-PEG-D2B. It is too early to know if this approach will be successful.

3.4. Bismuth-213 for PSMA-TAT

Bi-213 is a mixed α and β emitter with a half-life of 45.6 min (Figure 2). Preclinical studies have demonstrated that TAT with 213Bi labeled antibody J591, small molecule inhibitor PSMA-I&T or nanobody JVZ-008 showed efficient and rapid tumor targeting, induced apoptosis in PSMA-overexpressing cell lines, significantly delayed spheroid growth of xenograft tumor growth in nude mice [81,96,97,98]. Small molecule PSMA-I&T induced more double-strand breaks than nanobody JVZ-008 [98]. A dosimetry estimate comparing 213Bi-PSMA-617 and 225Ac-PSMA-617 has demonstrated that the short-lived 213Bi is an inferior choice for TAT with PSMA-617 [168]. To our knowledge, there is only one single case report to date on the clinical application of 213Bi-PSMA-617 [105]. The patient was treated with two cycles of 213Bi-PSMA-617 with a cumulative activity of 592 MBq. A biochemical response (decrease in PSA level from 237 μg/L to 43 μg/L) was observed [105]. The short half-life of 213Bi makes this radionuclide less suitable for routine therapeutic applications due to logistical challenges. Hence, the subject is not discussed further.

3.5. Astatine-211 for PSMA-TAT

At-211 has a half-life of 7.2 hours (Figure 2) and is a promising radiohalogen for PSMA-TAT [86,87,88]. It decays by electron capture (58.3%) to 211Po (t1/2 = 0.52 s), which decays to stable 207Pb by emitting an α particle (7.45 MeV, 100%). Additionally, its daughter 211Po emits K X-rays in the range of 77 to 92 keV that allows the quantification of 211At radioactivity and scintigraphic imaging of 211At in vivo [169]. In 2009, modified anti-PSMA antibody 107-1A4 was labeled with 211At [86]. SCID mice bearing intra-tibial human prostate cancer C4-2B tumors were treated with 370 kBq 211At-107-1A4 [86]. The treatment decreased PSA levels in mice without any toxicity. The same year urea-based PSMA inhibitor labeled with 211At, 2-[3-[5-(4-[211At]astato-benzoylamino)-1-carboxy-pentyl]-ureido]-pentanedioic acid (ABCPUP) was synthesized [82]. In vitro studies demonstrated that 211At-ABCUP had significantly higher uptake in PC3 PIP (PSMA-positive) compared to PC3 (PSMA-negative) human prostate cancer cells. In 2016, Kiess et al. [88] synthesized a urea-based small-molecule targeting PSMA, (2S)-2-(3-(1-carboxy-5-(4-211At-astatobenzamido) pentyl)ureido)-pentanedioic acid (211At-PSMA 6), that significantly improved survival in mice bearing PC3 PIP micrometastases. However, the high uptake of 211At-PSMA 6 in renal proximal tubules resulted in late nephrotoxicity (≤12 months) [88]. The maximum tolerated single dose of 211At-PSMA 6 in immunocompetent CD1 mice was 37 kBq, and the lethal dose to 10% of mice was 111 kBq [88]. Such toxicity limits the clinical use of 211At-PSMA 6. In 2017, Kelly et al. [89] designed six novel urea-based ligands for dual-targeting PSMA and human serum albumin. Compounds with higher affinity for human serum albumin showed prolonged blood retention resulting in reduced kidney uptake. Their most potent compounds, RPS-027 and MIP-1095, were labeled with 131I as a surrogate for 211At, and their biodistribution was tested in mice bearing LNCaP xenograft tumors [89]. Similar tumor uptake was observed for both products, but 131I-RPS-027 had a five-fold reduction in kidney uptake compared to MIP-1095. Recently, to reduce kidney uptake, protein-Glu-urea-Lys conjugates and potential metabolites were synthesized and radio iodinated (as the surrogate for 211At) and administered to athymic mice bearing C4-2B tumor xenografts [90]. Conjugation of PEGylated PSMA derivatives to proteins reduce kidney uptake, but long polyethylene glycol (PEG) linkers have reduced uptake in tumor [90]. There are not many studies testing 211At for PSMA-TAT since the availability of 211At is limited. At-211 is produced mostly in cyclotrons by the bombardment of natural 209Bi with α-beam at energies below 28.4 MeV [169,170]. Unfortunately, there are only about 30 cyclotrons in the world that have the beam characteristics required for 211At production [169].

3.6. Actinium-225 for PSMA-TAT

Ac-225 (t1/2 = 10 days) decays via a chain of five daughter radionuclides (t1/2 from 3.7 µs up to 45.6 min) to stable 209Bi, emitting four α particles and two β particles (Figure 2). Earlier, the potential use of 225Ac for TAT was limited because of difficulties in the selection of chelating agents able to form strong bonds, leading to renal toxicity induced by the longer-lived decay product 213Bi [55,121]. Nevertheless, some stable complexes have been synthesized, and several more recent clinical trials have demonstrated the potential of 225Ac-PSMA-617 and 225Ac-PSMA-I&T for the treatment of mCRPC (Table 5). The first-in-human PSMA-TAT study was published in 2016 by Kratochwil et al. [42] from Heidelberg. Two mCRPC patients with challenging clinical situations and extensive pretreatment were treated with 100 kBq/kg of 225Ac-PSMA-617 at bi-monthly intervals as salvage therapy after the presence of a PSMA-positive tumor phenotype had been validated by 68Ga-PSMA-11 PET/CT [42]. The first patient was not suitable for 177Lu-PSMA-617 (diffuse red marrow infiltration), and the second one was resistant to 177Lu-PSMA-617 [42]. Both patients showed a complete response on the PET/CT scan, and PSA declined below the measurable level [42]. Salivary gland toxicity leading to dry mouth syndrome or xerostomia was reported in both patients [42]. The second study with 14 mCRPC patients found that a treatment activity of 100 kBq/kg of body weight of 225Ac-PSMA-617 per cycle every 8 weeks was the most optimal when considering both efficacy (biochemical response) and tolerability [106]. Severe xerostomia was the dose-limiting toxicity [106]. This standardized treatment protocol for 225Ac-PSMA-617 is routinely applied for salvage therapy of end-stage mCRPC patients in many studies [106,107,113]. The efficacy of 225Ac-PSMA-617 TAT was evaluated in 40 mCRPC patients [107]. This study demonstrated a PSA decline of more than 50% in 63% of patients, with a median duration of tumor control of 9 months [107]. The median overall survival was more than 12 months [107]. The majority of mCRPC patients at the Heidelberg clinic were heavily pretreated before TAT with chemotherapy, radiotherapy, and androgen deprivation therapy (abiraterone (85%) and enzalutamide (60%)) [107]. In South Africa, chemotherapy-naïve mCRPC patients treated with 225Ac-PSMA-617 TAT had reduced toxicity to salivary glands, a 90% PSA decline in 88% of the patients, and 50% achieved undetectable serum PSA and remained in remission 12 months after therapy [109]. The first clinical data using 225Ac-PSMA-I&T showed highly comparable biochemical responses as after 225Ac-PSMA-617 TAT [116,117].
Since PSMA-617 crosses the blood–brain barrier and accumulates in cerebral metastases [171], a significant regression of cerebral metastases was demonstrated using 225Ac-PSMA-617 [108]. Prostate cancer patients with brain metastases have limited treatment options and poor survival, and TAT with 225Ac-PSMA-617 may have substantial therapeutic potential for these patients.
In the clinical setting, several studies reported toxicity related to TAT with 225Ac-PSMA-617/PSMA-I&T (Table 5). Xerostomia is a common side effect that causes 10–25% of patients to stop TAT with 225Ac-PSMA [42,107,111,112,117,172]. Xerostomia should, therefore, be prevented. Modification of the administered activity of 225Ac-PSMA-617 and the number of cycles of TAT may decrease the side effects while still achieving response [111,173]. Sialendoscopy with dilatation, saline irrigation, and steroid injection (prednisolone) have been investigated in patients with some but limited success [174]. A case report in one patient describes the potential beneficial effects of intraparenchymal injections of botulinum toxin before 225Ac-PSMA-617 TAT [175]. External cooling of the salivary gland using ice packs from 30 min pre-infusion through 2 h post-infusion of radiopharmaceuticals was expected to reduce PSMA radioligand uptake due to vasoconstriction [172]. However, the relative contributions of salivary gland cooling and the reduced 225Ac-PSMA-617 activity in minimizing xerostomia severity remain unclear. Therefore, effective methods to reduce salivary toxicity are needed.
Due to the physiological expression of PSMA in kidneys and predominantly renal excretion of 225Ac-PSMA-617, there is concern about possible radiation toxicity to the kidneys that may cause acute and long-term effects [138,176,177]. It has been reported that the kidney function deteriorated in a patient with one functional kidney after 225Ac-PSMA-617 [109] and that chronic kidney disease was found in two patients with mCRPC after 225Ac-PSMA-617 therapy [114]. Until now, retention times of PSMA ligands either in kidneys or in tumor cells have not yet been evaluated systematically [168]. If PSMA on the surface of cancer cells is not sufficiently internalized after binding of the ligand, TAT with 225Ac with multiple unstable daughters might be suboptimal and toxic [168]. It has also been speculated that the radioactive daughters of 225Ac, but not 225Ac-PSMA-617, can accumulate in the tubular cells and irradiate the kidneys, leading to renal injury [114]. In such cases, the therapeutic potential of 225Ac-PSMA will be substantially decreased, and toxicity increased. Further studies are necessary to evaluate the stability, retention times, and intracellular localization in cancer cells and kidneys of the 225Ac-PSMA complexes intended for TAT.
The first report on the use of 225Ac-PSMA-617 in vivo was only published in 2019 [99]. NSG mice bearing subcutaneous C4-2 tumors were treated with 20, 40, and 100 kBq/mouse of 225Ac-PSMA-617 [99]. Significant tumor growth inhibition was observed in all treatment groups compared to the control. However, mice treated with 100 kBq had some weight loss, while the mice treated with lower activities experienced only transient weight loss. In 2020, the same group reported a useful mouse model of human metastatic prostate cancer by injecting C4-2 cells expressing firefly luciferase into the left ventricle of NSG mice, which was then used to evaluate the effectiveness of 225Ac-PSMA-617 at various disease stages [100]. Early treatment, one-week post-inoculation of C4-2 cells, with 40 kBq/mouse of 225Ac-PSMA-617 prevented liver metastases and led to significant survival benefit [100]. In 2019, Kelly et al. [103] studied the albumin-binding and PSMA-targeting ligand RPS-074 labeled with 225Ac in BALB/c mice bearing LNCaP xenograft tumors. Significant tumor growth inhibition was observed in mice treated with 74 and 148 kBq of 225Ac-RPS-074. A single injection of 148 kBq induced a complete response in 6 of 7 tumors, with no apparent toxic effects. In 2020, Current et al. [101] documented that 225Ac-PSMA-617 efficacy is dependent on cellular PSMA levels and intra-tumoral PSMA heterogeneity.
Another approach to reducing toxicity in salivary glands and kidneys is to use antibodies (e.g., J591) instead of small molecule PSMA for TAT [83,154]. J591 binds to a different site of PSMA compared to PSMA ligands and has a much lower distribution in salivary glands and kidneys [154]. Ongoing clinical trials with 227Ac-J591 will provide the missing information on stability, efficacy, and toxicity (Table 6).
The mainly retrospective studies have reported promising response rates, progression-free survival, and overall survival (Table 5). The limited availability of 225Ac is the main challenge for its clinical use. Many research centers investigate the possibility of producing 225Ac in commercial quantities. In the future, scaled-up production of 225Ac could be achieved by the use of a high current cyclotron or electron linear accelerator (linac) [178,179].

3.7. Thorium-227 for PSMA-TAT

Th-227 has a physical half-life of 18.7 days and decays through radioactive 223Ra and the other short-lived radionuclides in its decay chain to stable 207Pb by emitting five α particles (Figure 2). The long half-life of 227Th allows transportation and preparation of the radiopharmaceutical. Due to its chemical properties, 227Th can be linked to a variety of antibodies and proteins [72,180]. Th-227 complexed with octadentate 3,2-hydroxypyridinone (3,2-HOPO) chelators that are conjugated to antibodies or other targeting moieties results in highly stable targeted 227Th conjugates (TTCs) [104]. TTCs, therefore, represent a new promising class of TAT [104]. A novel, fully human antibody-based TAT, PSMA-TTC (BAY 2315497), has been recently developed by Bayer and tested in mice bearing prostate cancer tumors [104]. The antitumor efficacy of PSMA-TTC was observed in different prostate cancer models. However, survival and long-term toxicity have not been reported. These preclinical data encouraged the further investigation of BAY 2315497 in an ongoing phase I trial in mCRPC (Table 6).

3.8. Terbium-149 for PSMA-TAT

A very interesting alternative to the presented α emitters is the 149Tb, currently studied in preclinical radioimmunotherapy [85]. Tb-149 has a half-life of 4.1 hours and is considered a promising theragnostic radionuclide. It decays by α emission (3.97 MeV, 16.7%), as well as electron capture (76.2%), positron emission (7.1%), gamma rays, and X-rays. Therefore, it can be used for TAT and PET imaging [181]. Longer half-life of 149Tb compared to 213Bi (t1/2 = 46 min) and 212Bi (t1/2 = 1.0 h), absence of α-emitting daughters, and chelation with S-2-(4-isothiocyanatobenzyl)-1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid (DOTA) are clear advantages of 149Tb [181]. Antitumor activity of 149Tb-PSMA-617 has been demonstrated in mice bearing PC3 PIP tumors [85]. Tb-149 is a rare-earth element, and its production and chemical separation are associated with serious difficulties, which partially explains why 149Tb is not in clinical use yet [182]. Additionally, the presence of long-lived daughters appearing during the decay of 149Tb (Figure 2) complicates dosimetry and might increase the risk of undesirable physiological effects. The long-lived daughter radionuclides, 149Eu (t1/2 = 93 d) and 145Sm (t1/2 = 340 d), are potential bone seekers [183]. Due to the recoil energy of the alpha decay of 149Tb, there is a high risk of release of these radionuclides into the bloodstream leading to potential accumulation in bone and, thus, to long-term irradiation of the bone marrow [183].

3.9. Lead-212 for PSMA-TAT

Another suitable radionuclide for PSMA-TAT is β emitter 212Pb (t1/2 = 10.6 h) that acts as an in vivo generator of α particles via its short-lived progenies 212Bi (t1/2 = 60.6 min) and 212Po (t1/2 = 0.3 µs) (Figure 2). The decay of one 212Pb atom releases on average one α particle and a mean α energy of 7.9 MeV. The use of 212Pb circumvents the impractical short half-life of 212Bi and delivers around ten times the dose per administered activity unit compared to 212Bi alone, reducing the amount of activity required [184,185]. Pb-212 can be obtained at an industrial scale from 224Ra-based generators using 228Th (t1/2 = 1.9 years) as a long-term generator. It has suitable properties in terms of chelation chemistry and forms stable complexes both with the versatile chelator DOTA and the lead-specific chelator S-2-(4-isothiocyanatobenzyl)-1,4,7,10-tetraaza-1,4,7,10-tetra(2-carbamoylmethyl)-cyclododecane (TCMC) [186]. Comprehensive reviews describing the use of 212Pb for TAT of cancer have recently been published [170,187,188].
Several research groups have developed PSMA-targeting ligands specifically designed for 212Pb chelation that show promising tumor-targeting in preclinical models [91,92,94,95]. The urea-based PSMA ligands NG001, labeled with 212Pb, and CA012 and L2, labeled with the surrogate 203Pb, show high tumor uptake (8–25%ID/g) 1–2 h post-injection in mice bearing PSMA-positive tumors, which is comparable with clinically used 177Lu-PSMA-617 [91,92,94,130,148]. In NSG mice bearing PC3 PIP tumors, 1.5 and 3.7 MBq of 212Pb-L2 significantly inhibited tumor growth with therapeutic indices (TI) of 1.9 and 3.0, respectively, with time to reach a 10-fold tumor increase used as an endpoint [92]. In a micrometastatic tumor model, 3.7 MBq of 212Pb-L2 demonstrated an increased survival benefit compared to 37 MBq of 177Lu-PSMA-617 (TI of 1.2 vs. 1.0, respectively). However, a long-term toxicity study of 212Pb-L2 in healthy, immunocompetent mice identified the kidney as the dose-limiting organ, and the maximum tolerated dose (MTD) of the radioligand was determined to be 1.5 MBq. In athymic nude mice bearing C4-2 xenografts, an injected activity of only 0.32 MBq of 212Pb-NG001 demonstrated increased survival compared to the control group with a therapeutic index of >2.0 (median survival of 15 days vs. >30 days) [93]. The results warrant further preclinical studies to evaluate the long-term toxicity of 212Pb-NG001.
With shorter-lived radionuclides, such as 212Pb, the high initial kidney uptake could present a potential toxicity problem because of the higher dose rate of these radionuclides [189]. However, the mentioned Pb-labeled ligand NG001 exhibit lower kidney uptake than PSMA-617 [94]. Promising therapeutic results labeled with the short-lived 213Bi (t1/2 = 45.6 min) with an even higher dose rate than 212Pb were reported [105]. Another challenge with 212Pb is the retention of daughter radionuclides in the chelator after decay. Up to 36% of 212Bi could dissociate from DOTA and TCMC chelators of antibody complexes because of high recoil energies of the α-emitting daughters [120,184,190,191]. However, no translocation of the 212Bi daughter was detected in non-targeted organs during a 24 hour study period of 212Pb-NG001, likely prevented by the rapid tumor targeting and cellular internalization, as well as the rapid normal tissue clearance of the radioligand [94].

3.10. Dual Alpha (224Ra&212Pb) for TAT

An ideal PSMA-TAT for mCRPC must combat the entire spectrum of metastases present in the patients. A dual-alpha approach that uses the high LET ionizing energy of 224Ra and daughter radionuclides to target various mCRPC lesions have been presented [95]. Here, a 224Ra solution in transient equilibrium with daughter radionuclides was used for in situ labeling of a PSMA-targeting ligand, i.e., 212Pb is complexed by the ligand in the presence of 224Ra [95,184,185]. The resulting solution has dual-targeting properties; natural bone-seeking 224Ra will target osteoblastic metastatic lesions, and the 212Pb-labeled PSMA ligand will target extraskeletal metastases by selective binding to the surface of PSMA-expressing cells [95]. Thus, the dual-alpha approach is providing a bone-seeker and a PSMA-seeker in one radiopharmaceutical solution. The accumulation of 224Ra in bone and 212Pb-labeled PSMA ligand in tumor sites was verified in C4-2 tumor-bearing mice and warrants further investigation in vivo [95].

3.11. Combination Treatments with PSMA-TAT

Around 20% of patients treated with 225Ac-PSMA-617 have a poor response (Table 5) or early resistance against 225Ac-PSMA-617 [107], despite sufficient expression of PSMA and uptake of 225Ac-PSMA-617 in their tumors [192]. Several combination treatments with PSMA-TAT have been proposed [173,192,193]. Their goal is to increase PSMA-TAT efficacy by using therapies with different action mechanisms together with TAT, keeping toxic effects to a minimum. The tandem PSMA-RLT approach has been introduced to increase efficacy and reduce toxicity [173,194]. A single course of tandem therapy with low-activity 225Ac-PSMA-617 and full-activity 177Lu-PSMA-617 has enhanced response to PSMA-RLT and minimized xerostomia severity in men with late-stage/end-stage mCRPC [173,194]. There is a subgroup of patients (~17%) with poor response to 225Ac-PSMA-617 who harbor mutations in DNA damage–repair and checkpoint genes [192]. Combining PSMA-TAT and DNA damage–repair–targeting agents, such as poly(ADP-ribose)-polymerase inhibitors, have been suggested for these patients [192]. Lastly, it has been hypothesized that PSMA-TAT may increase tumor immunogenicity, and the use of immune checkpoint inhibitors may improve efficacy [102]. Synergistic antitumor efficacy between 225Ac-PSMA-617 and PD-1 blockage has recently been observed in C57BL/6-mice bearing syngeneic RM1-PGLS tumors [102].

4. Limitations of Preclinical Studies Related to Clinical PSMA-TAT

A major concern of PSMA-targeting radioligands is the radiotoxicity in PSMA-expressing organs. Only the most optimal and safest PSMA-targeted radioligands from preclinical studies are tested in humans. However, radiotoxicity in rodents is not the most accurate predictor of toxicity in humans because the human, rat, and mouse PSMA have different patterns of anatomical expression in normal tissues [195,196,197]. For example, PSMA levels in human submandibular gland are approximately four-fold higher compared with mouse and fifty-four-fold higher compared with rat submandibular gland [197]. Rupp et al. demonstrated that the accumulation of PSMA ligands in salivary glands in humans is high due to PSMA-unrelated uptake mechanisms [198], while PSMA radioligands (177Lu, 212Pb) show negligible uptake in mouse salivary glands [94,95,132]. The details of salivary glands’ uptake mechanisms are currently unclear and must be further investigated using different clinical models. Furthermore, PSMA levels in human kidneys are approximately two-fold lower compared with mouse kidneys [138]. Many patients treated with PSMA-TAT report severe xerostomia, and only a few of them, nephrotoxicity (Table 5). The direct comparison of PSMA-TAT data from various groups is not easy since different mouse strains, cell lines, and different specific activities of radiopharmaceuticals have been used in the various studies. PSMA-TAT efficacy increases with increasing PSMA levels [101]. For example, transduced PC3 PIP cells present much higher PSMA expression levels than LNCaP or C4-2 cells (Table 7), and the direct comparisons of survival, tumor uptake, and tumor to kidney ratio of different radiopharmaceuticals must be done carefully.
To date, PSMA-617 and PSMA-I&T are the most often clinically applied small molecule ligands for TRT and diagnostics [39,202]. Both ligands contain a peptidomimetic glutamate–urea–lysine binding motif, but chelators are different [138,203]. PSMA-617 contains a DOTA chelator while PSMA I&T contains a DOTAGA (1,4,7,10-tetraazacyclododecane-1-(glutamic acid)-4,7,10-triacetic acid) chelator, [138,203]. Biodistribution studies of 177Lu-PSMA-617 and 177Lu-PSMA-I&T in mice bearing PC3 PIP or PC295 PDX prostate tumors demonstrated comparable tumor uptake at all time points, while 177Lu-PSMA-I&T had much higher (10–40 folds) renal uptake, resulting in an unfavorable tumor-to-kidney ratio [204,205]. The data from these preclinical studies are in partial discrepancy with clinical studies: biodistribution and absorbed doses in tumor or kidneys were comparable for PSMA-617 and PSMA-I&T [206]. Therefore, caution must be taken, so that novel PSMA ligands with clinical potential are not discarded based on preclinical data.

5. Future of PSMA-TAT

The success of PSMA-TAT depends on the availability of α emitters and chelators, enhanced tumor uptake of linkers and targeting moieties, and reduced toxicity and progeny redistribution.
Further preclinical proof of concepts with PSMA-TAT in relevant murine models of mCRPC regarding the impact of dose rate and relative biological effectiveness is sorely needed. This also relates to direct comparisons of 225Ac, 227Th, and 212Pb PSMA-TAT with 177Lu-PSMA TRT.
The detailed mechanisms of action of PSMA-TAT are far from fully understood [72]. Elucidating in vitro and in vivo molecular and cellular mechanisms related to targeted and bystander effects of PSMA-TAT are important. It has been suggested that TAT bears the potential to induce immunogenic cell death through the release of damage-associated molecular patterns from dying tumor cells that result in the activation of tumor-specific immune responses [207]. However, the role of PSMA-TAT in modulating the immunogenicity of prostate cancer cells remains unknown.
The dosimetry for the labeled α emitter and its progeny is challenging and still in the early stage because of the heterogeneous antigen expression among cancer cells and the nature of short-range, high-LET alpha radiation (Table 1 and Table 2). This needs to be further investigated using modeling methods. Autoradiographs can help to obtain high-resolution images, but they can only be performed ex vivo. In such cases, estimates of radiation doses could be based on experimental data and modeling. Recommended therapeutic activities (e.g., 4–8 MBq 225Ac-PSMA-617) limit the clinical applicability of SPECT [208]. Theranostic imaging protocols using 68Ga-PSMA ligands does not provide any information about the translocation of daughter radionuclides, and the impact of recoiling daughter molecules on dosimetry of α-emitting radiopharmaceuticals on healthy tissues are unknown.

6. Conclusions

Preclinical studies and preliminary efficacy and safety data on PSMA-TAT in mCRPC patients are very encouraging. Because of production cost, logistical, and supply problems with several of the more promising α-emitters, the future of clinical PSMA-TAT should focus on radionuclides that are suitable for large scale production and supply, such as 212Pb, which can be produced at the industrial scale with existing methods. Dual targeting with 212Pb/224Ra is particularly promising since most late-stage prostate cancers have skeletal involvement, and 212Pb-PSMA ligand and 224Ra represent two different targeting approaches in one radiopharmaceutical solution. The future of clinical PSMA-TAT may also include novel PSMA ligands and combined approaches.

Author Contributions

Writing—original draft preparation, A.J.; writing—review and editing, A.J., V.Y.S., Ø.S.B. and R.H.L. All authors have read and agreed to the published version of the manuscript.

Funding

This study was funded by the Norwegian Research Council (Grant No: 290639, Oslo, Norway) and South-Eastern Norway Regional Health Authority (Grant No: 2020028, Helse Sør-Øst, Oslo, Norway).

Conflicts of Interest

R.H.L., Ø.S.B. and V.Y.S. hold ownership interest in Nucligen. A.J. declares no conflict of interest.

References

  1. Bray, F.; Ferlay, J.; Soerjomataram, I.; Siegel, R.L.; Torre, L.A.; Jemal, A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2018, 68, 394–424. [Google Scholar] [CrossRef] [Green Version]
  2. Ingrosso, G.; Detti, B.; Scartoni, D.; Lancia, A.; Giacomelli, I.; Baki, M.; Carta, G.; Livi, L.; Santoni, R. Current therapeutic options in metastatic castration-resistant prostate cancer. Semin. Oncol. 2018, 45, 303–315. [Google Scholar] [CrossRef]
  3. Labriola, M.K.; Atiq, S.; Hirshman, N.; Bitting, R.L. Management of men with metastatic castration-resistant prostate cancer following potent androgen receptor inhibition: A review of novel investigational therapies. Prostate Cancer Prostat. Dis. 2020. [Google Scholar] [CrossRef]
  4. Den, R.B.; George, D.; Pieczonka, C.; McNamara, M. Ra-223 Treatment for Bone Metastases in Castrate-Resistant Prostate Cancer: Practical Management Issues for Patient Selection. Am. J. Clin. Oncol. 2019, 42, 399–406. [Google Scholar] [CrossRef] [PubMed]
  5. Pezaro, C.; Omlin, A.; Lorente, D.; Rodrigues, D.N.; Ferraldeschi, R.; Bianchini, D.; Mukherji, D.; Riisnaes, R.; Altavilla, A.; Crespo, M.; et al. Visceral disease in castration-resistant prostate cancer. Eur. Urol. 2014, 65, 270–273. [Google Scholar] [CrossRef] [Green Version]
  6. Parker, C.; Nilsson, S.; Heinrich, D.; Helle, S.I.; O’Sullivan, J.M.; Fossa, S.D.; Chodacki, A.; Wiechno, P.; Logue, J.; Seke, M.; et al. Alpha emitter radium-223 and survival in metastatic prostate cancer. N. Engl. J. Med. 2013, 369, 213–223. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Mukherji, D.; El Dika, I.; Temraz, S.; Haidar, M.; Shamseddine, A. Evolving treatment approaches for the management of metastatic castration-resistant prostate cancer—Role of radium-223. Ther. Clin. Risk Manag. 2014, 10, 373–380. [Google Scholar] [CrossRef] [Green Version]
  8. Moussa, M.; Papatsoris, A.; Abou Chakra, M.; Sryropoulou, D.; Dellis, A. Pharmacotherapeutic strategies for castrate-resistant prostate cancer. Expert Opin. Pharmacother. 2020, 1–18. [Google Scholar] [CrossRef]
  9. Norum, J.; Nieder, C. Treatments for Metastatic Prostate Cancer (mPC): A Review of Costing Evidence. Pharmacoeconomics 2017, 35, 1223–1236. [Google Scholar] [CrossRef]
  10. Brönimann, S.; Lemberger, U.; Bruchbacher, A.; Shariat, S.F.; Hassler, M.R. Poly(ADP-ribose) polymerase inhibitors in prostate and urothelial cancer. Curr. Opin. Urol. 2020, 30, 519–526. [Google Scholar] [CrossRef]
  11. Pollard, M.E.; Moskowitz, A.J.; Diefenbach, M.A.; Hall, S.J. Cost-effectiveness analysis of treatments for metastatic castration resistant prostate cancer. Asian J. Urol. 2017, 4, 37–43. [Google Scholar] [CrossRef]
  12. Agrawal, S. The role of 225Ac-PSMA-617 in chemotherapy-naive patients with advanced prostate cancer: Is it the new beginning. Indian J. Urol. IJU J. Urol. Soc. India 2020, 36, 69–70. [Google Scholar] [CrossRef] [PubMed]
  13. de Bono, J.; Mateo, J.; Fizazi, K.; Saad, F.; Shore, N.; Sandhu, S.; Chi, K.N.; Sartor, O.; Agarwal, N.; Olmos, D.; et al. Olaparib for Metastatic Castration-Resistant Prostate Cancer. N. Engl. J. Med. 2020, 382, 2091–2102. [Google Scholar] [CrossRef] [PubMed]
  14. Reyes, E.E.; VanderWeele, D.J.; Isikbay, M.; Duggan, R.; Campanile, A.; Stadler, W.M.; Vander Griend, D.J.; Szmulewitz, R.Z. Quantitative characterization of androgen receptor protein expression and cellular localization in circulating tumor cells from patients with metastatic castration-resistant prostate cancer. J. Transl. Med. 2014, 12, 1–15. [Google Scholar] [CrossRef] [Green Version]
  15. Arasaratnam, M.; Crumbaker, M.; Bhatnagar, A.; McKay, M.J.; Molloy, M.P.; Gurney, H. Inter- and intra-patient variability in pharmacokinetics of abiraterone acetate in metastatic prostate cancer. Cancer Chemother. Pharmacol. 2019, 84, 139–146. [Google Scholar] [CrossRef] [PubMed]
  16. Kyriakopoulos, C.E.; Heath, E.I.; Ferrari, A.; Sperger, J.M.; Singh, A.; Perlman, S.B.; Roth, A.R.; Perk, T.G.; Modelska, K.; Porcari, A.; et al. Exploring Spatial-Temporal Changes in (18)F-Sodium Fluoride PET/CT and Circulating Tumor Cells in Metastatic Castration-Resistant Prostate Cancer Treated With Enzalutamide. J. Clin. Oncol. 2020, 38, 3662–3671. [Google Scholar] [CrossRef] [PubMed]
  17. Ross, J.S.; Gray, K.E.; Webb, I.J.; Gray, G.S.; Rolfe, M.; Schenkein, D.P.; Nanus, D.M.; Millowsky, M.I.; Bander, N.H. Antibody-based therapeutics: Focus on prostate cancer. Cancer Metastasis Rev. 2005, 24, 521–537. [Google Scholar] [CrossRef]
  18. Barve, A.; Jin, W.; Cheng, K. Prostate cancer relevant antigens and enzymes for targeted drug delivery. J. Control. Release Off. J. Control. Release Soc. 2014, 187, 118–132. [Google Scholar] [CrossRef] [Green Version]
  19. Evans, J.C.; Malhotra, M.; Cryan, J.F.; O’Driscoll, C.M. The therapeutic and diagnostic potential of the prostate specific membrane antigen/glutamate carboxypeptidase II (PSMA/GCPII) in cancer and neurological disease. Br. J. Pharmacol. 2016, 173, 3041–3079. [Google Scholar] [CrossRef] [Green Version]
  20. Ross, J.S.; Sheehan, C.E.; Fisher, H.A.; Kaufman, R.P., Jr.; Kaur, P.; Gray, K.; Webb, I.; Gray, G.S.; Mosher, R.; Kallakury, B.V. Correlation of primary tumor prostate-specific membrane antigen expression with disease recurrence in prostate cancer. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2003, 9, 6357–6362. [Google Scholar]
  21. Hupe, M.C.; Philippi, C.; Roth, D.; Kumpers, C.; Ribbat-Idel, J.; Becker, F.; Joerg, V.; Duensing, S.; Lubczyk, V.H.; Kirfel, J.; et al. Expression of Prostate-Specific Membrane Antigen (PSMA) on Biopsies Is an Independent Risk Stratifier of Prostate Cancer Patients at Time of Initial Diagnosis. Front. Oncol. 2018, 8, 623. [Google Scholar] [CrossRef]
  22. Paschalis, A.; Sheehan, B.; Riisnaes, R.; Rodrigues, D.N.; Gurel, B.; Bertan, C.; Ferreira, A.; Lambros, M.B.K.; Seed, G.; Yuan, W.; et al. Prostate-specific Membrane Antigen Heterogeneity and DNA Repair Defects in Prostate Cancer. Eur. Urol. 2019, 76, 469–478. [Google Scholar] [CrossRef] [Green Version]
  23. Kasperzyk, J.L.; Finn, S.P.; Flavin, R.; Fiorentino, M.; Lis, R.; Hendrickson, W.K.; Clinton, S.K.; Sesso, H.D.; Giovannucci, E.L.; Stampfer, M.J.; et al. Prostate-specific membrane antigen protein expression in tumor tissue and risk of lethal prostate cancer. Cancer Epidemiol. Prev. Biomark. 2013, 22, 2354–2363. [Google Scholar] [CrossRef] [Green Version]
  24. Ghosh, A.; Heston, W.D. Tumor target prostate specific membrane antigen (PSMA) and its regulation in prostate cancer. J. Cell. Biochem. 2004, 91, 528–539. [Google Scholar] [CrossRef]
  25. Troyer, J.K.; Beckett, M.L.; Wright, G.L., Jr. Detection and characterization of the prostate-specific membrane antigen (PSMA) in tissue extracts and body fluids. Int. J. Cancer 1995, 62, 552–558. [Google Scholar]
  26. Silver, D.A.; Pellicer, I.; Fair, W.R.; Heston, W.D.; Cordon-Cardo, C. Prostate-specific membrane antigen expression in normal and malignant human tissues. Clin. Cancer Res. 1997, 3, 81–85. [Google Scholar]
  27. Kinoshita, Y.; Kuratsukuri, K.; Landas, S.; Imaida, K.; Rovito, P.M., Jr.; Wang, C.Y.; Haas, G.P. Expression of prostate-specific membrane antigen in normal and malignant human tissues. World J. Surg. 2006, 30, 628–636. [Google Scholar] [CrossRef]
  28. Jin, W.; Barve, A.; Cheng, K. PSMA-specific ligands in prostate cancer diagnosis and therapy. EMJ Urol. 2016, 4, 62–69. [Google Scholar]
  29. Cimadamore, A.; Cheng, M.; Santoni, M.; Lopez-Beltran, A.; Battelli, N.; Massari, F.; Galosi, A.B.; Scarpelli, M.; Montironi, R. New Prostate Cancer Targets for Diagnosis, Imaging, and Therapy: Focus on Prostate-Specific Membrane Antigen. Front. Oncol. 2018, 8, 653. [Google Scholar] [CrossRef] [Green Version]
  30. Vahidfar, N.; Fallahpoor, M.; Farzanehfar, S.; Divband, G.; Ahmadzadehfar, H. Historical review of pharmacological development and dosimetry of PSMA-based theranostics for prostate cancer. J. Radioanal. Nucl. Chem. 2019, 322, 237–248. [Google Scholar] [CrossRef]
  31. Jones, W.; Griffiths, K.; Barata, P.C.; Paller, C.J. PSMA Theranostics: Review of the Current Status of PSMA-Targeted Imaging and Radioligand Therapy. Cancers 2020, 12, 1367. [Google Scholar] [CrossRef]
  32. Bravaccini, S.; Puccetti, M.; Bocchini, M.; Ravaioli, S.; Celli, M.; Scarpi, E.; De Giorgi, U.; Tumedei, M.M.; Raulli, G.; Cardinale, L.; et al. PSMA expression: A potential ally for the pathologist in prostate cancer diagnosis. Sci. Rep. 2018, 8, 1–8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Ruigrok, E.A.M.; van Weerden, W.M.; Nonnekens, J.; de Jong, M. The Future of PSMA-Targeted Radionuclide Therapy: An Overview of Recent Preclinical Research. Pharmaceutics 2019, 11, 560. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Rathke, H.; Flechsig, P.; Mier, W.; Bronzel, M.; Mavriopoulou, E.; Hohenfellner, M.; Giesel, F.L.; Haberkorn, U.; Kratochwil, C. Dosimetry Estimate and Initial Clinical Experience with (90)Y-PSMA-617. J. Nucl. Med. 2019, 60, 806–811. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Kelly, J.M.; Ponnala, S.; Amor-Coarasa, A.; Zia, N.A.; Nikolopoulou, A.; Williams, C., Jr.; Schlyer, D.J.; DiMagno, S.G.; Donnelly, P.S.; Babich, J.W. Preclinical Evaluation of a High-Affinity Sarcophagine-Containing PSMA Ligand for (64)Cu/(67)Cu-Based Theranostics in Prostate Cancer. Mol. Pharm. 2020, 17, 1954–1962. [Google Scholar] [CrossRef]
  36. Carlos Dos Santos, J.; Beijer, B.; Bauder-Wüst, U.; Schäfer, M.; Leotta, K.; Eder, M.; Benešová, M.; Kleist, C.; Giesel, F.; Kratochwil, C.; et al. Development of Novel PSMA Ligands for Imaging and Therapy with Copper Isotopes. J. Nucl. Med. 2020, 61, 70–79. [Google Scholar] [CrossRef]
  37. Yadav, M.P.; Ballal, S.; Bal, C.; Sahoo, R.K.; Damle, N.A.; Tripathi, M.; Seth, A. Efficacy and Safety of 177Lu-PSMA-617 Radioligand Therapy in Metastatic Castration-Resistant Prostate Cancer Patients. Clin. Nucl. Med. 2020, 45, 19–31. [Google Scholar] [CrossRef] [PubMed]
  38. Yadav, M.P.; Ballal, S.; Sahoo, R.K.; Dwivedi, S.N.; Bal, C. Radioligand Therapy With (177)Lu-PSMA for Metastatic Castration-Resistant Prostate Cancer: A Systematic Review and Meta-Analysis. Am. J. Roentgenol. 2019, 213, 275–285. [Google Scholar] [CrossRef]
  39. von Eyben, F.E.; Roviello, G.; Kiljunen, T.; Uprimny, C.; Virgolini, I.; Kairemo, K.; Joensuu, T. Third-line treatment and (177)Lu-PSMA radioligand therapy of metastatic castration-resistant prostate cancer: A systematic review. Eur. J. Nucl. Med. Mol. Imaging 2018, 45, 496–508. [Google Scholar] [CrossRef] [Green Version]
  40. Rahbar, K.; Ahmadzadehfar, H.; Kratochwil, C.; Haberkorn, U.; Schafers, M.; Essler, M.; Baum, R.P.; Kulkarni, H.R.; Schmidt, M.; Drzezga, A.; et al. German Multicenter Study Investigating 177Lu-PSMA-617 Radioligand Therapy in Advanced Prostate Cancer Patients. J. Nucl. Med. 2017, 58, 85–90. [Google Scholar] [CrossRef] [Green Version]
  41. Chakravarty, R.; Siamof, C.M.; Dash, A.; Cai, W. Targeted alpha-therapy of prostate cancer using radiolabeled PSMA inhibitors: A game changer in nuclear medicine. Am. J. Nucl. Med. Mol. Imaging 2018, 8, 247–267. [Google Scholar]
  42. Kratochwil, C.; Bruchertseifer, F.; Giesel, F.L.; Weis, M.; Verburg, F.A.; Mottaghy, F.; Kopka, K.; Apostolidis, C.; Haberkorn, U.; Morgenstern, A. 225Ac-PSMA-617 for PSMA-Targeted alpha-Radiation Therapy of Metastatic Castration-Resistant Prostate Cancer. J. Nucl. Med. 2016, 57, 1941–1944. [Google Scholar] [CrossRef] [Green Version]
  43. Satapathy, S.; Mittal, B.R.; Sood, A. Visceral Metastases as Predictors of Response and Survival Outcomes in Patients of Castration-Resistant Prostate Cancer Treated With 177Lu-Labeled Prostate-Specific Membrane Antigen Radioligand Therapy: A Systematic Review and Meta-analysis. Clin. Nucl. Med. 2020, 45, 935–942. [Google Scholar] [CrossRef]
  44. Emmett, L.; Willowson, K.; Violet, J.; Shin, J.; Blanksby, A.; Lee, J. Lutetium (177) PSMA radionuclide therapy for men with prostate cancer: A review of the current literature and discussion of practical aspects of therapy. J. Med. Radiat. Sci. 2017, 64, 52–60. [Google Scholar] [CrossRef]
  45. Bernhardt, P.; Forssell-Aronsson, E.; Jacobsson, L.; Skarnemark, G. Low-energy electron emitters for targeted radiotherapy of small tumours. Acta Oncol. 2001, 40, 602–608. [Google Scholar]
  46. Hindié, E.; Zanotti-Fregonara, P.; Quinto, M.A.; Morgat, C.; Champion, C. Dose Deposits from 90Y, 177Lu, 111In, and 161Tb in Micrometastases of Various Sizes: Implications for Radiopharmaceutical Therapy. J. Nucl. Med. 2016, 57, 759–764. [Google Scholar] [CrossRef] [Green Version]
  47. Behr, T.M.; Béhé, M.; Stabin, M.G.; Wehrmann, E.; Apostolidis, C.; Molinet, R.; Strutz, F.; Fayyazi, A.; Wieland, E.; Gratz, S.; et al. High-linear energy transfer (LET) alpha versus low-LET beta emitters in radioimmunotherapy of solid tumors: Therapeutic efficacy and dose-limiting toxicity of 213Bi- versus 90Y-labeled CO17-1A Fab’ fragments in a human colonic cancer model. Cancer Res. 1999, 59, 2635–2643. [Google Scholar]
  48. Barinka, C.; Sácha, P.; Sklenár, J.; Man, P.; Bezouska, K.; Slusher, B.S.; Konvalinka, J. Identification of the N-glycosylation sites on glutamate carboxypeptidase II necessary for proteolytic activity. Protein Sci. Publ. Protein Soc. 2004, 13, 1627–1635. [Google Scholar] [CrossRef] [Green Version]
  49. Davis, M.I.; Bennett, M.J.; Thomas, L.M.; Bjorkman, P.J. Crystal structure of prostate-specific membrane antigen, a tumor marker and peptidase. Proc. Natl. Acad. Sci. USA 2005, 102, 5981–5986. [Google Scholar] [CrossRef] [Green Version]
  50. Sokoloff, R.L.; Norton, K.C.; Gasior, C.L.; Marker, K.M.; Grauer, L.S. A dual-monoclonal sandwich assay for prostate-specific membrane antigen: Levels in tissues, seminal fluid and urine. Prostate 2000, 43, 150–157. [Google Scholar] [CrossRef]
  51. Yao, V.; Berkman, C.E.; Choi, J.K.; O’Keefe, D.S.; Bacich, D.J. Expression of prostate-specific membrane antigen (PSMA), increases cell folate uptake and proliferation and suggests a novel role for PSMA in the uptake of the non-polyglutamated folate, folic acid. Prostate 2010, 70, 305–316. [Google Scholar] [CrossRef]
  52. Maurer, T.; Eiber, M.; Schwaiger, M.; Gschwend, J.E. Current use of PSMA-PET in prostate cancer management. Nat. Rev. Urol. 2016, 13, 226–235. [Google Scholar] [CrossRef]
  53. Wibmer, A.G.; Burger, I.A.; Sala, E.; Hricak, H.; Weber, W.A.; Vargas, H.A. Molecular Imaging of Prostate Cancer. Radiographics 2016, 36, 142–159. [Google Scholar] [CrossRef] [Green Version]
  54. Rajasekaran, A.K.; Anilkumar, G.; Christiansen, J.J. Is prostate-specific membrane antigen a multifunctional protein? Am. J. Physiol. Cell Physiol. 2005, 288, C975–C981. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Czerwińska, M.; Bilewicz, A.; Kruszewski, M.; Wegierek-Ciuk, A.; Lankoff, A. Targeted Radionuclide Therapy of Prostate Cancer-From Basic Research to Clinical Perspectives. Molecules 2020, 25, 1743. [Google Scholar] [CrossRef] [Green Version]
  56. Rahbar, K.; Afshar-Oromieh, A.; Jadvar, H.; Ahmadzadehfar, H. PSMA Theranostics: Current Status and Future Directions. Mol. Imaging 2018, 17, 1536012118776068. [Google Scholar] [CrossRef] [Green Version]
  57. Jadvar, H. Targeted Radionuclide Therapy: An Evolution Toward Precision Cancer Treatment. Am. J. Roentgenol. 2017, 209, 277–288. [Google Scholar] [CrossRef] [Green Version]
  58. Quast, U. Whole body radiotherapy: A TBI-guideline. J. Med. Phys. 2006, 31, 5–12. [Google Scholar] [CrossRef]
  59. Seidl, C. Radioimmunotherapy with α-particle-emitting radionuclides. Immunotherapy 2014, 6, 431–458. [Google Scholar] [CrossRef]
  60. Parker, C.; Lewington, V.; Shore, N.; Kratochwil, C.; Levy, M.; Linden, O.; Noordzij, W.; Park, J.; Saad, F. Targeted Alpha Therapy, an Emerging Class of Cancer Agents: A Review. JAMA Oncol. 2018, 4, 1765–1772. [Google Scholar] [PubMed]
  61. Navalkissoor, S.; Grossman, A. Targeted Alpha Particle Therapy for Neuroendocrine Tumours: The Next Generation of Peptide Receptor Radionuclide Therapy. Neuroendocrinology 2019, 108, 256–264. [Google Scholar] [CrossRef] [PubMed]
  62. Tomita, M.; Maeda, M. Mechanisms and biological importance of photon-induced bystander responses: Do they have an impact on low-dose radiation responses. J. Radiat. Res. 2015, 56, 205–219. [Google Scholar] [CrossRef] [Green Version]
  63. Parker, C.; Heidenreich, A.; Nilsson, S.; Shore, N. Current approaches to incorporation of radium-223 in clinical practice. Prostate Cancer Prostat. Dis. 2018, 21, 37–47. [Google Scholar] [CrossRef]
  64. Dandapani, S.V.; Wong, J.; Twardowski, P. Review of Radium-223 and Metastatic Castration-Sensitive Prostate Cancer. Cancer Biother. Radiopharm. 2020, 35, 490–496. [Google Scholar] [CrossRef] [PubMed]
  65. Etchebehere, E.; Brito, A.E.; Rezaee, A.; Langsteger, W.; Beheshti, M. Therapy assessment of bone metastatic disease in the era of (223)radium. Eur. J. Nucl. Med. Mol. Imaging 2017, 44, 84–96. [Google Scholar] [CrossRef] [PubMed]
  66. Bruland, Ø.S.; Nilsson, S.; Fisher, D.R.; Larsen, R.H. High-linear energy transfer irradiation targeted to skeletal metastases by the alpha-emitter 223Ra: Adjuvant or alternative to conventional modalities? Clin. Cancer Res. 2006, 12, 6250s–6257s. [Google Scholar] [CrossRef] [Green Version]
  67. Hoskin, P.; Sartor, O.; O’Sullivan, J.M.; Johannessen, D.C.; Helle, S.I.; Logue, J.; Bottomley, D.; Nilsson, S.; Vogelzang, N.J.; Fang, F.; et al. Efficacy and safety of radium-223 dichloride in patients with castration-resistant prostate cancer and symptomatic bone metastases, with or without previous docetaxel use: A prespecified subgroup analysis from the randomised, double-blind, phase 3 ALSYMPCA trial. Lancet Oncol. 2014, 15, 1397–1406. [Google Scholar]
  68. De Vincentis, G.; Gerritsen, W.; Gschwend, J.E.; Hacker, M.; Lewington, V.; O’Sullivan, J.M.; Oya, M.; Pacilio, M.; Parker, C.; Shore, N.; et al. Advances in targeted alpha therapy for prostate cancer. Ann. Oncol. Off. J. Eur. Soc. Med. Oncol. 2019, 30, 1728–1739. [Google Scholar] [CrossRef]
  69. Facchini, G.; Cavaliere, C.; D’Aniello, C.; Iovane, G.; Rossetti, S. Abiraterone acetate treatment in patients with castration-resistant prostate cancer with visceral metastases: A real-world experience. Anti-Cancer Drugs 2019, 30, 179–185. [Google Scholar] [CrossRef]
  70. Rahbar, K.; Boegemann, M.; Yordanova, A.; Eveslage, M.; Schafers, M.; Essler, M.; Ahmadzadehfar, H. PSMA targeted radioligandtherapy in metastatic castration resistant prostate cancer after chemotherapy, abiraterone and/or enzalutamide. A retrospective analysis of overall survival. Eur. J. Nucl. Med. Mol. Imaging 2018, 45, 12–19. [Google Scholar] [CrossRef]
  71. Ferrier, M.G.; Radchenko, V. An Appendix of Radionuclides Used in Targeted Alpha Therapy. J. Med. Imaging Radiat. Sci. 2019, 50, S58–S65. [Google Scholar] [CrossRef] [Green Version]
  72. Liberal, F.D.C.G.; O’Sullivan, J.M.; McMahon, S.J.; Prise, K.M. Targeted Alpha Therapy: Current Clinical Applications. Cancer Biother. Radiopharm. 2020, 35, 404–417. [Google Scholar] [CrossRef]
  73. Garashchenko, B.L.; Korsakova, V.A.; Yakovlev, R.Y. Radiopharmaceuticals Based on Alpha Emitters: Preparation, Properties, and Application. Phys. Nucl. 2018, 81, 1515–1525. [Google Scholar] [CrossRef]
  74. Poty, S.; Francesconi, L.C.; McDevitt, M.R.; Morris, M.J.; Lewis, J.S. Alpha-Emitters for Radiotherapy: From Basic Radiochemistry to Clinical Studies-Part 1. J. Nucl. Med. 2018, 59, 878–884. [Google Scholar] [CrossRef] [Green Version]
  75. National Nuclear Data Center, Brookhaven National Laboratory, Upton, NY. Available online: https://www.nndc.bnl.gov/ (accessed on 4 September 2020).
  76. Covell, D.G.; Barbet, J.; Holton, O.D.; Black, C.D.; Parker, R.J.; Weinstein, J.N. Pharmacokinetics of monoclonal immunoglobulin G1, F(ab’)2, and Fab’ in mice. Cancer Res. 1986, 46, 3969–3978. [Google Scholar]
  77. Tabrizi, M.A.; Tseng, C.M.; Roskos, L.K. Elimination mechanisms of therapeutic monoclonal antibodies. Drug Discov. Today 2006, 11, 81–88. [Google Scholar] [CrossRef]
  78. Ovacik, M.; Lin, K. Tutorial on Monoclonal Antibody Pharmacokinetics and Its Considerations in Early Development. Clin. Transl. Sci. 2018, 11, 540–552. [Google Scholar] [CrossRef]
  79. Miyahira, A.K.; Pienta, K.J.; Morris, M.J.; Bander, N.H.; Baum, R.P.; Fendler, W.P.; Goeckeler, W.; Gorin, M.A.; Hennekes, H.; Pomper, M.G.; et al. Meeting report from the Prostate Cancer Foundation PSMA-directed radionuclide scientific working group. Prostate 2018, 78, 775–789. [Google Scholar] [CrossRef]
  80. Holmes, E.H. PSMA specific antibodies and their diagnostic and therapeutic use. Expert Opin. Investig. Drugs 2001, 10, 511–519. [Google Scholar] [CrossRef]
  81. Ballangrud, A.M.; Yang, W.H.; Charlton, D.E.; McDevitt, M.R.; Hamacher, K.A.; Panageas, K.S.; Ma, D.; Bander, N.H.; Scheinberg, D.A.; Sgouros, G. Response of LNCaP spheroids after treatment with an alpha-particle emitter (213Bi)-labeled anti-prostate-specific membrane antigen antibody (J591). Cancer Res. 2001, 61, 2008–2014. [Google Scholar]
  82. Vaidyanathan, G.; Mease, R.; Affleck, D.; Chen, Y.; Welsh, P.; Hens, M.; Pomper, M.; Zalutsky, M. An astatine-211 labeled PSMA inhibitor for targeted alpha-particle radiotherapy of prostate carcinoma. J. Nucl. Med. 2009, 50, 40. [Google Scholar]
  83. Bandekar, A.; Zhu, C.; Jindal, R.; Bruchertseifer, F.; Morgenstern, A.; Sofou, S. Anti-prostate-specific membrane antigen liposomes loaded with 225Ac for potential targeted antivascular α-particle therapy of cancer. J. Nucl. Med. 2014, 55, 107–114. [Google Scholar] [CrossRef] [Green Version]
  84. Czerwińska, M.; Fracasso, G.; Pruszyński, M.; Bilewicz, A.; Kruszewski, M.; Majkowska-Pilip, A.; Lankoff, A. Design and Evaluation of (223)Ra-Labeled and Anti-PSMA Targeted NaA Nanozeolites for Prostate Cancer Therapy-Part I. Materials 2020, 13, 3875. [Google Scholar] [CrossRef]
  85. Umbricht, C.A.; Koster, U.; Bernhardt, P.; Gracheva, N.; Johnston, K.; Schibli, R.; van der Meulen, N.P.; Muller, C. Alpha-PET for Prostate Cancer: Preclinical investigation using (149)Tb-PSMA-617. Sci. Rep. 2019, 9, 1–10. [Google Scholar] [CrossRef] [Green Version]
  86. Wilbur, D.S.; Hamlin, D.; Nguyen, H.; Nakamae, H.; Chyan, M.K.; Vessella, R.L.; Sandmaier, B. Preliminary studies using At-211-labeled anti-PSMA MAb for treatment of metastatic prostate cancer in a mouse model. J. Nucl. Med. 2009, 50, 39. [Google Scholar]
  87. Wilbur, D.S.; Chyan, M.K.; Hamlin, D.K.; Nguyen, H.; Vessella, R.L. Reagents for astatination of biomolecules. 5. Evaluation of hydrazone linkers in (211)At- and (125)I-labeled closo-decaborate(2-) conjugates of Fab’ as a means of decreasing kidney retention. Bioconjug. Chem. 2011, 22, 1089–1102. [Google Scholar] [CrossRef] [Green Version]
  88. Kiess, A.P.; Minn, I.; Vaidyanathan, G.; Hobbs, R.F.; Josefsson, A.; Shen, C.; Brummet, M.; Chen, Y.; Choi, J.; Koumarianou, E.; et al. (2S)-2-(3-(1-Carboxy-5-(4-211At-Astatobenzamido)Pentyl)Ureido)-Pentanedioic Acid for PSMA-Targeted alpha-Particle Radiopharmaceutical Therapy. J. Nucl. Med. 2016, 57, 1569–1575. [Google Scholar] [CrossRef] [Green Version]
  89. Kelly, J.M.; Amor-Coarasa, A.; Nikolopoulou, A.; Wüstemann, T.; Barelli, P.; Kim, D.; Williams, C., Jr.; Zheng, X.; Bi, C.; Hu, B.; et al. Dual-Target Binding Ligands with Modulated Pharmacokinetics for Endoradiotherapy of Prostate Cancer. J. Nucl. Med. 2017, 58, 1442–1449. [Google Scholar] [CrossRef] [Green Version]
  90. Li, Y.; Chyan, M.K.; Hamlin, D.K.; Nguyen, H.; Vessella, R.; Wilbur, D.S. Evaluation of radioiodinated protein conjugates and their potential metabolites containing lysine-urea-glutamate (LuG), PEG and closo-decaborate(2-) as models for targeting astatine-211 to metastatic prostate cancer. Nucl. Med. Biol. 2020, 92, 217–227. [Google Scholar] [CrossRef]
  91. Dos Santos, J.C.; Schafer, M.; Bauder-Wust, U.; Lehnert, W.; Leotta, K.; Morgenstern, A.; Kopka, K.; Haberkorn, U.; Mier, W.; Kratochwil, C. Development and dosimetry of (203)Pb/(212)Pb-labelled PSMA ligands: Bringing “the lead“ into PSMA-targeted alpha therapy? Eur. J. Nucl. Med. Mol. Imaging 2019, 46, 1081–1091. [Google Scholar] [CrossRef] [Green Version]
  92. Banerjee, S.R.; Minn, I.L.; Kumar, V.; Josefsson, A.; Lisok, A.; Brummet, M.; Chen, J.; Kiess, A.; Baidoo, K.; Brayton, C.; et al. Preclinical evaluation of 203/212Pb-labeled low-molecular-weight compounds for targeted radiopharmaceutical therapy of prostate cancer. J. Nucl. Med. 2020, 61, 80–88. [Google Scholar] [CrossRef]
  93. Larsen, R.H. Lead and Thorium Compounds. U.S. Patent 10377778B2, 13 August 2019. [Google Scholar]
  94. Stenberg, V.Y.; Juzeniene, A.; Chen, Q.; Yang, X.; Bruland, O.S.; Larsen, R.H. Preparation of the alpha-emitting PSMA targeted radioligand [(212) Pb]Pb-NG001 for prostate cancer. J. Label. Compd. Radiopharm. 2020, 63, 129–143. [Google Scholar] [CrossRef] [Green Version]
  95. Stenberg, V.Y.; Juzeniene, A.; Bruland, Ø.S.; Larsen, R.H. In situ Generated 212Pb-PSMA Ligand in a 224Ra-Solution for Dual Targeting of Prostate Cancer Sclerotic Stroma and PSMA-positive Cells. Curr. Radiopharm. 2020, 13, 130–141. [Google Scholar] [CrossRef]
  96. McDevitt, M.R.; Barendswaard, E.; Ma, D.; Lai, L.; Curcio, M.J.; Sgouros, G.; Ballangrud, A.M.; Yang, W.H.; Finn, R.D.; Pellegrini, V.; et al. An alpha-particle emitting antibody ([213Bi]J591) for radioimmunotherapy of prostate cancer. Cancer Res. 2000, 60, 6095–6100. [Google Scholar]
  97. Li, Y.; Tian, Z.; Rizvi, S.M.; Bander, N.H.; Allen, B.J. In vitro and preclinical targeted alpha therapy of human prostate cancer with Bi-213 labeled J591 antibody against the prostate specific membrane antigen. Prostate Cancer Prostat. Dis. 2002, 5, 36–46. [Google Scholar] [CrossRef]
  98. Nonnekens, J.; Chatalic, K.L.; Molkenboer-Kuenen, J.D.; Beerens, C.E.; Bruchertseifer, F.; Morgenstern, A.; Veldhoven-Zweistra, J.; Schottelius, M.; Wester, H.J.; van Gent, D.C.; et al. (213)Bi-Labeled Prostate-Specific Membrane Antigen-Targeting Agents Induce DNA Double-Strand Breaks in Prostate Cancer Xenografts. Cancer Biother. Radiopharm. 2017, 32, 67–73. [Google Scholar] [CrossRef]
  99. Meyer, C.; Stuparu, A.; Wei, L.; Capri, J.; Le, T.; Radu, C.; Czernin, J.; Dahlbom, M.; Slavik, R. Therapeutic Efficacy and Dosimetry of Targeted Alpha Therapy using 225Ac-PSMA-617 in a Murine Model of Prostate Cancer. J. Med. Imaging Radiat. Sci. 2019, 50, S93–S94. [Google Scholar] [CrossRef]
  100. Stuparu, A.D.; Meyer, C.A.L.; Evans-Axelsson, S.L.; Lückerath, K.; Wei, L.H.; Kim, W.; Poddar, S.; Mona, C.E.; Dahlbom, M.; Girgis, M.D.; et al. Targeted alpha therapy in a systemic mouse model of prostate cancer—A feasibility study. Theranostics 2020, 10, 2612–2620. [Google Scholar] [CrossRef]
  101. Current, K.; Meyer, C.; Magyar, C.E.; Mona, C.E.; Almajano, J.; Slavik, R.; Stuparu, A.D.; Cheng, C.; Dawson, D.W.; Radu, C.G.; et al. Investigating PSMA-targeted radioligand therapy efficacy as a function of cellular PSMA levels and intra-tumoral PSMA heterogeneity. Clin. Cancer Res. 2020, 26, 2946–2955. [Google Scholar] [CrossRef]
  102. Czernin, J.; Current, K.; Mona, C.E.; Nyiranshuti, L.; Hikmat, F.; Radu, C.G.; Lueckerath, K. Immune-Checkpoint Blockade Enhances (225)Ac-PSMA617 Efficacy in a Mouse Model of Prostate Cancer. J. Nucl. Med. 2020, 62, 228–231. [Google Scholar] [CrossRef]
  103. Kelly, J.M.; Amor-Coarasa, A.; Ponnala, S.; Nikolopoulou, A.; Williams, C., Jr.; Thiele, N.A.; Schlyer, D.; Wilson, J.J.; DiMagno, S.G.; Babich, J.W. A Single Dose of (225)Ac-RPS-074 Induces a Complete Tumor Response in an LNCaP Xenograft Model. J. Nucl. Med. 2019, 60, 649–655. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Hammer, S.; Hagemann, U.B.; Zitzmann-Kolbe, S.; Larsen, A.; Ellingsen, C.; Geraudie, S.; Grant, D.; Indrevoll, B.; Smeets, R.; von Ahsen, O.; et al. Preclinical Efficacy of a PSMA-Targeted Thorium-227 Conjugate (PSMA-TTC), a Targeted Alpha Therapy for Prostate Cancer. Clin. Cancer Res. 2020, 26, 1985–1996. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Sathekge, M.; Knoesen, O.; Meckel, M.; Modiselle, M.; Vorster, M.; Marx, S. (213)Bi-PSMA-617 targeted alpha-radionuclide therapy in metastatic castration-resistant prostate cancer. Eur. J. Nucl. Med. Mol. Imaging 2017, 44, 1099–1100. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Kratochwil, C.; Bruchertseifer, F.; Rathke, H.; Bronzel, M.; Apostolidis, C.; Weichert, W.; Haberkorn, U.; Giesel, F.L.; Morgenstern, A. Targeted alpha-Therapy of Metastatic Castration-Resistant Prostate Cancer with (225)Ac-PSMA-617: Dosimetry Estimate and Empiric Dose Finding. J. Nucl. Med. 2017, 58, 1624–1631. [Google Scholar] [CrossRef] [Green Version]
  107. Kratochwil, C.; Bruchertseifer, F.; Rathke, H.; Hohenfellner, M.; Giesel, F.L.; Haberkorn, U.; Morgenstern, A. Targeted alpha-Therapy of Metastatic Castration-Resistant Prostate Cancer with (225)Ac-PSMA-617: Swimmer-Plot Analysis Suggests Efficacy Regarding Duration of Tumor Control. J. Nucl. Med. 2018, 59, 795–802. [Google Scholar] [CrossRef] [Green Version]
  108. Sathekge, M.M.; Bruchertseifer, F.; Lawal, I.O.; Vorster, M.; Knoesen, O.; Lengana, T.; Boshomane, T.G.; Mokoala, K.K.; Morgenstern, A. Treatment of brain metastases of castration-resistant prostate cancer with (225)Ac-PSMA-617. Eur. J. Nucl. Med. Mol. Imaging 2019, 46, 1756–1757. [Google Scholar] [CrossRef] [PubMed]
  109. Sathekge, M.; Bruchertseifer, F.; Knoesen, O.; Reyneke, F.; Lawal, I.; Lengana, T.; Davis, C.; Mahapane, J.; Corbett, C.; Vorster, M.; et al. (225)Ac-PSMA-617 in chemotherapy-naive patients with advanced prostate cancer: A pilot study. Eur. J. Nucl. Med. Mol. Imaging 2019, 46, 129–138. [Google Scholar] [CrossRef] [Green Version]
  110. de Medeiros, R.B.; Grigolon, M.V.; Araújo, T.P.; Srougi, M. Metastatic castration-resistant prostate cancer (mCRPC) treated with 225Ac-PSMA-617. Case report. Braz. J. Oncol. 2019, 15, 1–9. [Google Scholar] [CrossRef]
  111. Sathekge, M.; Bruchertseifer, F.; Vorster, M.; Lawal, I.O.; Knoesen, O.; Mahapane, J.; Davis, C.; Reyneke, F.; Maes, A.; Kratochwil, C.; et al. Predictors of Overall and Disease-Free Survival in Metastatic Castration-Resistant Prostate Cancer Patients Receiving (225)Ac-PSMA-617 Radioligand Therapy. J. Nucl. Med. 2020, 61, 62–69. [Google Scholar] [CrossRef]
  112. Feuerecker, B.; Tauber, R.; Knorr, K.; Heck, M.; Beheshti, A.; Seidl, C.; Bruchertseifer, F.; Pickhard, A.; Gafita, A.; Kratochwil, C.; et al. Activity and Adverse Events of Actinium-225-PSMA-617 in Advanced Metastatic Castration-resistant Prostate Cancer After Failure of Lutetium-177-PSMA. Eur. Urol. 2020. [Google Scholar] [CrossRef]
  113. Yadav, M.P.; Ballal, S.; Sahoo, R.K.; Tripathi, M.; Seth, A.; Bal, C. Efficacy and safety of (225)Ac-PSMA-617 targeted alpha therapy in metastatic castration-resistant Prostate Cancer patients. Theranostics 2020, 10, 9364–9377. [Google Scholar] [CrossRef]
  114. Pelletier, K.; Côté, G.; Fallah-Rad, N.; John, R.; Kitchlu, A. Chronic kidney disease after 225Ac-PSMA617 therapy in patients with metastatic prostate cancer. Kidney Int. Rep. 2021, in press. [Google Scholar]
  115. van der Doelen, M.J.; Mehra, N.; van Oort, I.M.; Looijen-Salamon, M.G.; Janssen, M.J.R.; Custers, J.A.E.; Slootbeek, P.H.J.; Kroeze, L.I.; Bruchertseifer, F.; Morgenstern, A.; et al. Clinical outcomes and molecular profiling of advanced metastatic castration-resistant prostate cancer patients treated with (225)Ac-PSMA-617 targeted alpha-radiation therapy. Urol. Oncol. 2020. [Google Scholar] [CrossRef]
  116. Ilhan, H.; Gosewisch, A.; Böning, G.; Völter, F.; Zacherl, M.; Unterrainer, M.; Bartenstein, P.; Todica, A.; Gildehaus, F.J. Response to (225)Ac-PSMA-I&T after failure of long-term (177)Lu-PSMA RLT in mCRPC. Eur. J. Nucl. Med. Mol. Imaging 2020, 1–2. [Google Scholar] [CrossRef]
  117. Zacherl, M.J.; Gildehaus, F.J.; Mittlmeier, L.; Boening, G.; Gosewisch, A.; Wenter, V.; Schmidt-Hegemann, N.S.; Belka, C.; Kretschmer, A.; Casuscelli, J.; et al. First clinical results for PSMA targeted alpha therapy using (225)Ac-PSMA-I&T in advanced mCRPC patients. J. Nucl. Med. 2020, jnumed-120. [Google Scholar] [CrossRef]
  118. Ackerman, N.L.; de la Fuente Rosales, L.; Falzone, N.; Vallis, K.A.; Bernal, M.A. Targeted alpha therapy with (212)Pb or (225)Ac: Change in RBE from daughter migration. Phys. Med. 2018, 51, 91–98. [Google Scholar] [CrossRef]
  119. Jaggi, J.S.; Seshan, S.V.; McDevitt, M.R.; LaPerle, K.; Sgouros, G.; Scheinberg, D.A. Renal tubulointerstitial changes after internal irradiation with alpha-particle-emitting actinium daughters. J. Am. Soc. Nephrol. 2005, 16, 2677–2689. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  120. de Kruijff, R.M.; Wolterbeek, H.T.; Denkova, A.G. A Critical Review of Alpha Radionuclide Therapy-How to Deal with Recoiling Daughters? Pharmaceuticals 2015, 8, 321–336. [Google Scholar] [CrossRef]
  121. Kozempel, J.; Mokhodoeva, O.; Vlk, M. Progress in Targeted Alpha-Particle Therapy. What We Learned about Recoils Release from In Vivo Generators. Molecules 2018, 23, 581. [Google Scholar] [CrossRef] [Green Version]
  122. Castillo Seoane, D.; de Saint-Hubert, M.; Crabbe, M.; Struelens, L.; Koole, M. Targeted alpha therapy: A critical review of translational dosimetry research with emphasis on actinium-225. Q. J. Nucl. Med. Mol. Imaging Off. Publ. Ital. Assoc. Nucl. Med. Int. Assoc. Radiopharmacol. 2020, 64, 265–277. [Google Scholar] [CrossRef] [PubMed]
  123. Wester, H.J.; Schottelius, M. PSMA-Targeted Radiopharmaceuticals for Imaging and Therapy. Semin. Nucl. Med. 2019, 49, 302–312. [Google Scholar] [CrossRef]
  124. Pastorino, S.; Riondato, M.; Uccelli, L.; Giovacchini, G.; Giovannini, E.; Duce, V.; Ciarmiello, A. Toward the Discovery and Development of PSMA Targeted Inhibitors for Nuclear Medicine Applications. Curr. Radiopharm. 2020, 13, 63–79. [Google Scholar] [CrossRef] [PubMed]
  125. Lutje, S.; Heskamp, S.; Cornelissen, A.S.; Poeppel, T.D.; van den Broek, S.A.; Rosenbaum-Krumme, S.; Bockisch, A.; Gotthardt, M.; Rijpkema, M.; Boerman, O.C. PSMA Ligands for Radionuclide Imaging and Therapy of Prostate Cancer: Clinical Status. Theranostics 2015, 5, 1388–1401. [Google Scholar] [CrossRef] [Green Version]
  126. Lutje, S.; Slavik, R.; Fendler, W.; Herrmann, K.; Eiber, M. PSMA ligands in prostate cancer—Probe optimization and theranostic applications. Methods 2017, 130, 42–50. [Google Scholar] [CrossRef]
  127. Wustemann, T.; Bauder-Wust, U.; Schafer, M.; Eder, M.; Benesova, M.; Leotta, K.; Kratochwil, C.; Haberkorn, U.; Kopka, K.; Mier, W. Design of Internalizing PSMA-specific Glu-ureido-based Radiotherapeuticals. Theranostics 2016, 6, 1085–1095. [Google Scholar] [CrossRef] [Green Version]
  128. Eiber, M.; Fendler, W.P.; Rowe, S.P.; Calais, J.; Hofman, M.S.; Maurer, T.; Schwarzenboeck, S.M.; Kratowchil, C.; Herrmann, K.; Giesel, F.L. Prostate-Specific Membrane Antigen Ligands for Imaging and Therapy. J. Nucl. Med. 2017, 58, 67s–76s. [Google Scholar] [CrossRef] [Green Version]
  129. Tykvart, J.; Schimer, J.; Bařinková, J.; Pachl, P.; Poštová-Slavětínská, L.; Majer, P.; Konvalinka, J.; Šácha, P. Rational design of urea-based glutamate carboxypeptidase II (GCPII) inhibitors as versatile tools for specific drug targeting and delivery. Bioorg. Med. Chem. 2014, 22, 4099–4108. [Google Scholar] [CrossRef] [PubMed]
  130. Benesova, M.; Schafer, M.; Bauder-Wust, U.; Afshar-Oromieh, A.; Kratochwil, C.; Mier, W.; Haberkorn, U.; Kopka, K.; Eder, M. Preclinical Evaluation of a Tailor-Made DOTA-Conjugated PSMA Inhibitor with Optimized Linker Moiety for Imaging and Endoradiotherapy of Prostate Cancer. J. Nucl. Med. 2015, 56, 914–920. [Google Scholar] [CrossRef] [Green Version]
  131. Benesova, M.; Bauder-Wust, U.; Schafer, M.; Klika, K.D.; Mier, W.; Haberkorn, U.; Kopka, K.; Eder, M. Linker Modification Strategies To Control the Prostate-Specific Membrane Antigen (PSMA)-Targeting and Pharmacokinetic Properties of DOTA-Conjugated PSMA Inhibitors. J. Med. Chem. 2016, 59, 1761–1775. [Google Scholar] [CrossRef] [PubMed]
  132. Benesova, M.; Umbricht, C.A.; Schibli, R.; Muller, C. Albumin-Binding PSMA Ligands: Optimization of the Tissue Distribution Profile. Mol. Pharm. 2018, 15, 934–946. [Google Scholar] [CrossRef]
  133. Schmidt, A.; Wirtz, M.; Farber, S.F.; Osl, T.; Beck, R.; Schottelius, M.; Schwaiger, M.; Wester, H.J. Effect of Carbohydration on the Theranostic Tracer PSMA I&T. ACS Omega 2018, 3, 8278–8287. [Google Scholar]
  134. Barrett, J.A.; Coleman, R.E.; Goldsmith, S.J.; Vallabhajosula, S.; Petry, N.A.; Cho, S.; Armor, T.; Stubbs, J.B.; Maresca, K.P.; Stabin, M.G.; et al. First-in-man evaluation of 2 high-affinity PSMA-avid small molecules for imaging prostate cancer. J. Nucl. Med. 2013, 54, 380–387. [Google Scholar] [CrossRef] [Green Version]
  135. Weineisen, M.; Schottelius, M.; Simecek, J.; Eiber, M.; Schwaiger, M.; Wester, H. Development and first in human evaluation of PSMA I&T—A ligand for diagnostic imaging and endoradiotherapy of prostate cancer. J. Nucl. Med. 2014, 55, 1083. [Google Scholar]
  136. Eder, M.; Neels, O.; Müller, M.; Bauder-Wüst, U.; Remde, Y.; Schäfer, M.; Hennrich, U.; Eisenhut, M.; Afshar-Oromieh, A.; Haberkorn, U.; et al. Novel Preclinical and Radiopharmaceutical Aspects of [68Ga]Ga-PSMA-HBED-CC: A New PET Tracer for Imaging of Prostate Cancer. Pharmaceuticals 2014, 7, 779–796. [Google Scholar] [CrossRef]
  137. Carlucci, G.; Ippisch, R.; Slavik, R.; Mishoe, A.; Blecha, J.; Zhu, S. (68)Ga-PSMA-11 NDA Approval: A Novel and Successful Academic Partnership. J. Nucl. Med. 2021, 62, 149–155. [Google Scholar] [CrossRef] [PubMed]
  138. Chatalic, K.L.; Heskamp, S.; Konijnenberg, M.; Molkenboer-Kuenen, J.D.; Franssen, G.M.; Clahsen-van Groningen, M.C.; Schottelius, M.; Wester, H.J.; van Weerden, W.M.; Boerman, O.C.; et al. Towards Personalized Treatment of Prostate Cancer: PSMA I&T, a Promising Prostate-Specific Membrane Antigen-Targeted Theranostic Agent. Theranostics 2016, 6, 849–861. [Google Scholar]
  139. Jilg, C.A.; Reichel, K.; Stoykow, C.; Rischke, H.C.; Bartholomä, M.; Drendel, V.; von Büren, M.; Schultze-Seemann, W.; Meyer, P.T.; Mix, M. Results from extended lymphadenectomies with [(111)In]PSMA-617 for intraoperative detection of PSMA-PET/CT-positive nodal metastatic prostate cancer. EJNMMI Res. 2020, 10, 1–13. [Google Scholar] [CrossRef] [Green Version]
  140. Rauscher, I.; Maurer, T.; Souvatzoglou, M.; Beer, A.J.; Vag, T.; Wirtz, M.; Weirich, G.; Wester, H.J.; Gschwend, J.E.; Schwaiger, M.; et al. Intrapatient Comparison of 111In-PSMA I&T SPECT/CT and Hybrid 68Ga-HBED-CC PSMA PET in Patients With Early Recurrent Prostate Cancer. Clin. Nucl. Med. 2016, 41, e397–e402. [Google Scholar]
  141. Eppard, E.; de la Fuente, A.; Benesova, M.; Khawar, A.; Bundschuh, R.A.; Gartner, F.C.; Kreppel, B.; Kopka, K.; Essler, M.; Rosch, F. Clinical Translation and First In-Human Use of [(44)Sc]Sc-PSMA-617 for PET Imaging of Metastasized Castrate-Resistant Prostate Cancer. Theranostics 2017, 7, 4359–4369. [Google Scholar] [CrossRef]
  142. Umbricht, C.A.; Benesova, M.; Schmid, R.M.; Turler, A.; Schibli, R.; van der Meulen, N.P.; Muller, C. (44)Sc-PSMA-617 for radiotheragnostics in tandem with (177)Lu-PSMA-617-preclinical investigations in comparison with (68)Ga-PSMA-11 and (68)Ga-PSMA-617. EJNMMI Res. 2017, 7, 1–10. [Google Scholar] [CrossRef] [Green Version]
  143. Khawar, A.; Eppard, E.; Sinnes, J.P.; Roesch, F.; Ahmadzadehfar, H.; Kürpig, S.; Meisenheimer, M.; Gaertner, F.C.; Essler, M.; Bundschuh, R.A. [44Sc]Sc-PSMA-617 Biodistribution and Dosimetry in Patients With Metastatic Castration-Resistant Prostate Carcinoma. Clin. Nucl. Med. 2018, 43, 323–330. [Google Scholar] [CrossRef]
  144. Kopka, K.; Benešová, M.; Bařinka, C.; Haberkorn, U.; Babich, J. Glu-Ureido-Based Inhibitors of Prostate-Specific Membrane Antigen: Lessons Learned During the Development of a Novel Class of Low-Molecular-Weight Theranostic Radiotracers. J. Nucl. Med. 2017, 58, 17s–26s. [Google Scholar] [CrossRef] [Green Version]
  145. Kelly, J.M.; Amor-Coarasa, A.; Ponnala, S.; Nikolopoulou, A.; Williams, C., Jr.; DiMagno, S.G.; Babich, J.W. Albumin-Binding PSMA Ligands: Implications for Expanding the Therapeutic Window. J. Nucl. Med. 2019, 60, 656–663. [Google Scholar] [CrossRef] [Green Version]
  146. Ling, X.; Latoche, J.D.; Choy, C.J.; Kurland, B.F.; Laymon, C.M.; Wu, Y.; Salamacha, N.; Shen, D.; Geruntho, J.J.; Rigatti, L.H.; et al. Preclinical Dosimetry, Imaging, and Targeted Radionuclide Therapy Studies of Lu-177-Labeled Albumin-Binding, PSMA-Targeted CTT1403. Mol. Imaging Biol. 2019, 22, 274–284. [Google Scholar] [CrossRef]
  147. Wang, Z.; Tian, R.; Niu, G.; Ma, Y.; Lang, L.; Szajek, L.P.; Kiesewetter, D.O.; Jacobson, O.; Chen, X. Single Low-Dose Injection of Evans Blue Modified PSMA-617 Radioligand Therapy Eliminates Prostate-Specific Membrane Antigen Positive Tumors. Bioconjug. Chem. 2018, 29, 3213–3221. [Google Scholar] [CrossRef] [PubMed]
  148. Kuo, H.T.; Merkens, H.; Zhang, Z.; Uribe, C.F.; Lau, J.; Zhang, C.; Colpo, N.; Lin, K.S.; Benard, F. Enhancing Treatment Efficacy of (177)Lu-PSMA-617 with the Conjugation of an Albumin-Binding Motif: Preclinical Dosimetry and Endoradiotherapy Studies. Mol. Pharm. 2018, 15, 5183–5191. [Google Scholar] [CrossRef]
  149. Umbricht, C.A.; Benesova, M.; Hasler, R.; Schibli, R.; van der Meulen, N.P.; Muller, C. Design and Preclinical Evaluation of an Albumin-Binding PSMA Ligand for (64)Cu-Based PET Imaging. Mol. Pharm. 2018, 15, 5556–5564. [Google Scholar] [CrossRef]
  150. Umbricht, C.A.; Benesova, M.; Schibli, R.; Muller, C. Preclinical Development of Novel PSMA-Targeting Radioligands: Modulation of Albumin-Binding Properties To Improve Prostate Cancer Therapy. Mol. Pharm. 2018, 15, 2297–2306. [Google Scholar] [CrossRef] [PubMed]
  151. Zang, J.; Fan, X.; Wang, H.; Liu, Q.; Wang, J.; Li, H.; Li, F.; Jacobson, O.; Niu, G.; Zhu, Z.; et al. First-in-human study of (177)Lu-EB-PSMA-617 in patients with metastatic castration-resistant prostate cancer. Eur. J. Nucl. Med. Mol. Imaging 2019, 46, 148–158. [Google Scholar] [CrossRef] [PubMed]
  152. Siow, A.; Kowalczyk, R.; Brimble, M.A.; Harris, P.W.R. Evolution of Peptide-Based Prostate-Specific Membrane Antigen (PSMA) Inhibitors: An Approach to Novel Prostate Cancer Therapeutics. Curr. Med. Chem. 2020. [Google Scholar] [CrossRef]
  153. Nováková, Z.; Foss, C.A.; Copeland, B.T.; Morath, V.; Baranová, P.; Havlínová, B.; Skerra, A.; Pomper, M.G.; Barinka, C. Novel Monoclonal Antibodies Recognizing Human Prostate-Specific Membrane Antigen (PSMA) as Research and Theranostic Tools. Prostate 2017, 77, 749–764. [Google Scholar] [CrossRef]
  154. Tagawa, S.T.; Vallabhajosula, S.; Jhanwar, Y.; Ballman, K.V.; Hackett, A.; Emmerich, L.; Babich, J.; Sartor, O.; Harshman, L.C.; Beltran, H.; et al. Phase I dose-escalation study of 225Ac-J591 for progressive metastatic castration resistant prostate cancer (mCRPC). J. Clin. Oncol. 2017, 36, TPS399. [Google Scholar] [CrossRef]
  155. Liu, H.; Moy, P.; Kim, S.; Xia, Y.; Rajasekaran, A.; Navarro, V.; Knudsen, B.; Bander, N.H. Monoclonal antibodies to the extracellular domain of prostate-specific membrane antigen also react with tumor vascular endothelium. Cancer Res. 1997, 57, 3629–3634. [Google Scholar] [PubMed]
  156. Brown, L.G.; Wegner, S.K.; Wang, H.; Buhler, K.R.; Arfman, E.W.; Lange, P.H.; Vessella, R.L. A novel monoclonal antibody 107-1A4 with high prostate specificity: Generation, characterization of antigen expression, and targeting of human prostate cancer xenografts. Prostate Cancer Prostat. Dis. 1998, 1, 208–215. [Google Scholar] [CrossRef] [Green Version]
  157. Kraeber-Bodéré, F.; Rousseau, C.; Bodet-Milin, C.; Mathieu, C.; Guérard, F.; Frampas, E.; Carlier, T.; Chouin, N.; Haddad, F.; Chatal, J.F.; et al. Tumor immunotargeting using innovative radionuclides. Int. J. Mol. Sci. 2015, 16, 3932–3954. [Google Scholar] [CrossRef]
  158. Sartor, O.; Sharma, D. Radium and other alpha emitters in prostate cancer. Transl. Androl. Urol. 2018, 7, 436–444. [Google Scholar] [CrossRef]
  159. Dauer, L.T.; Williamson, M.J.; Humm, J.; O’Donoghue, J.; Ghani, R.; Awadallah, R.; Carrasquillo, J.; Pandit-Taskar, N.; Aksnes, A.K.; Biggin, C.; et al. Radiation safety considerations for the use of 223RaCl₂ DE in men with castration-resistant prostate cancer. Health Phys. 2014, 106, 494–504. [Google Scholar] [CrossRef] [Green Version]
  160. Henriksen, G.; Hoff, P.; Larsen, R.H. Evaluation of potential chelating agents for radium. Appl. Radiat. Isot. 2002, 56, 667–671. [Google Scholar] [CrossRef]
  161. Gott, M.; Yang, P.; Kortz, U.; Stephan, H.; Pietzsch, H.J.; Mamat, C. A (224)Ra-labeled polyoxopalladate as a putative radiopharmaceutical. Chem. Commun. 2019, 55, 7631–7634. [Google Scholar] [CrossRef]
  162. Mokhodoeva, O.; Vlk, M.; Málková, E.; Kukleva, E.; Mičolová, P.; Štamberg, K.; Šlouf, M.; Dzhenloda, R.; Kozempel, J. Study of 223Ra uptake mechanism by Fe3O4 nanoparticles: Towards new prospective theranostic SPIONs. J. Nanopart. Res. 2016, 10, 1–12. [Google Scholar] [CrossRef]
  163. Suchánková, P.; Kukleva, E.; Štamberg, K.; Nykl, P.; Vlk, M.; Kozempel, J. Study of 223Ra uptake mechanism on hydroxyapatite and titanium dioxide nanoparticles as a function of pH. RSC Adv. 2020, 10, 3659–3666. [Google Scholar] [CrossRef] [Green Version]
  164. Reissig, F.; Zarschler, K.; Hübner, R.; Pietzsch, H.J.; Kopka, K.; Mamat, C. Sub-10 nm Radiolabeled Barium Sulfate Nanoparticles as Carriers for Theranostic Applications and Targeted Alpha Therapy. ChemistryOpen 2020, 9, 797–805. [Google Scholar] [CrossRef]
  165. Piotrowska, A.; Leszczuk, E.; Bruchertseifer, F.; Morgenstern, A.; Bilewicz, A. Functionalized NaA nanozeolites labeled with (224,225)Ra for targeted alpha therapy. J. Nanopart. Res. 2013, 15, 1–11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  166. Suchánková, P.; Kukleva, E.; Štamberg, K.; Nykl, P.; Sakmár, M.; Vlk, M.; Kozempel, J. Determination, Modeling and Evaluation of Kinetics of (223)Ra Sorption on Hydroxyapatite and Titanium Dioxide Nanoparticles. Materials 2020, 13, 1915. [Google Scholar] [CrossRef] [Green Version]
  167. Khodadadi Yazdi, M.; Zarrintaj, P.; Hosseiniamoli, H.; Mashhadzadeh, A.H.; Saeb, M.R.; Ramsey, J.D.; Ganjali, M.R.; Mozafari, M. Zeolites for theranostic applications. J. Mater. Chem. B 2020, 8, 5992–6012. [Google Scholar] [CrossRef]
  168. Kratochwil, C.; Schmidt, K.; Afshar-Oromieh, A.; Bruchertseifer, F.; Rathke, H.; Morgenstern, A.; Haberkorn, U.; Giesel, F.L. Targeted alpha therapy of mCRPC: Dosimetry estimate of (213)Bismuth-PSMA-617. Eur. J. Nucl. Med. Mol. Imaging 2018, 45, 31–37. [Google Scholar] [CrossRef] [Green Version]
  169. Lindegren, S.; Albertsson, P.; Bäck, T.; Jensen, H.; Palm, S.; Aneheim, E. Realizing Clinical Trials with Astatine-211: The Chemistry Infrastructure. Cancer Biother. Radiopharm. 2020, 35, 425–436. [Google Scholar] [CrossRef] [Green Version]
  170. Makvandi, M.; Dupis, E.; Engle, J.W.; Nortier, F.M.; Fassbender, M.E.; Simon, S.; Birnbaum, E.R.; Atcher, R.W.; John, K.D.; Rixe, O.; et al. Alpha-Emitters and Targeted Alpha Therapy in Oncology: From Basic Science to Clinical Investigations. Target. Oncol. 2018, 13, 189–203. [Google Scholar] [CrossRef]
  171. Puttemans, J.; Lahoutte, T.; D’Huyvetter, M.; Devoogdt, N. Beyond the Barrier: Targeted Radionuclide Therapy in Brain Tumors and Metastases. Pharmaceutics 2019, 11, 376. [Google Scholar] [CrossRef] [Green Version]
  172. Langbein, T.; Chausse, G.; Baum, R.P. Salivary Gland Toxicity of PSMA Radioligand Therapy: Relevance and Preventive Strategies. J. Nucl. Med. 2018, 59, 1172–1173. [Google Scholar] [CrossRef] [Green Version]
  173. Khreish, F.; Ebert, N.; Ries, M.; Maus, S.; Rosar, F.; Bohnenberger, H.; Stemler, T.; Saar, M.; Bartholomä, M.; Ezziddin, S. (225)Ac-PSMA-617/(177)Lu-PSMA-617 tandem therapy of metastatic castration-resistant prostate cancer: Pilot experience. Eur. J. Nucl. Med. Mol. Imaging 2020, 47, 721–728. [Google Scholar] [CrossRef]
  174. Rathke, H.; Kratochwil, C.; Hohenberger, R.; Giesel, F.L.; Bruchertseifer, F.; Flechsig, P.; Morgenstern, A.; Hein, M.; Plinkert, P.; Haberkorn, U.; et al. Initial clinical experience performing sialendoscopy for salivary gland protection in patients undergoing (225)Ac-PSMA-617 RLT. Eur. J. Nucl. Med. Mol. Imaging 2019, 46, 139–147. [Google Scholar] [CrossRef] [PubMed]
  175. Baum, R.P.; Langbein, T.; Singh, A.; Shahinfar, M.; Schuchardt, C.; Volk, G.F.; Kulkarni, H. Injection of Botulinum Toxin for Preventing Salivary Gland Toxicity after PSMA Radioligand Therapy: An Empirical Proof of a Promising Concept. Nucl. Med. Mol. Imaging 2018, 52, 80–81. [Google Scholar] [CrossRef] [PubMed]
  176. Gallyamov, M.; Meyrick, D.; Barley, J.; Lenzo, N. Renal outcomes of radioligand therapy: Experience of 177lutetium—prostate-specific membrane antigen ligand therapy in metastatic castrate-resistant prostate cancer. Clin. Kidney J. 2019, 13, 1049–1055. [Google Scholar] [CrossRef] [Green Version]
  177. Ristau, B.T.; O’Keefe, D.S.; Bacich, D.J. The prostate-specific membrane antigen: Lessons and current clinical implications from 20 years of research. Urol. Oncol. 2014, 32, 272–279. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  178. Melville, G.; Allen, B.J. Cyclotron and linac production of Ac-225. Appl. Radiat. Isot. 2009, 67, 549–555. [Google Scholar] [CrossRef]
  179. Robertson, A.K.H.; Ramogida, C.F.; Schaffer, P.; Radchenko, V. Development of (225)Ac Radiopharmaceuticals: TRIUMF Perspectives and Experiences. Curr. Radiopharm. 2018, 11, 156–172. [Google Scholar] [CrossRef]
  180. Frantellizzi, V.; Cosma, L.; Brunotti, G.; Pani, A.; Spanu, A.; Nuvoli, S.; De Cristofaro, F.; Civitelli, L.; De Vincentis, G. Targeted Alpha Therapy with Thorium-227. Cancer Biother. Radiopharm. 2020, 35, 437–445. [Google Scholar] [CrossRef]
  181. Müller, C.; Vermeulen, C.; Köster, U.; Johnston, K.; Türler, A.; Schibli, R.; van der Meulen, N.P. Alpha-PET with terbium-149: Evidence and perspectives for radiotheragnostics. EJNMMI Radiopharm. Chem. 2017, 1, 1–5. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  182. Müller, C.; Domnanich, K.A.; Umbricht, C.A.; van der Meulen, N.P. Scandium and terbium radionuclides for radiotheranostics: Current state of development towards clinical application. Br. J. Radiol. 2018, 91, 20180074. [Google Scholar] [CrossRef]
  183. Beyer, G.J.; Miederer, M.; Vranjes-Durić, S.; Comor, J.J.; Künzi, G.; Hartley, O.; Senekowitsch-Schmidtke, R.; Soloviev, D.; Buchegger, F. Targeted alpha therapy in vivo: Direct evidence for single cancer cell kill using 149Tb-rituximab. Eur. J. Nucl. Med. Mol. Imaging 2004, 31, 547–554. [Google Scholar] [CrossRef]
  184. Westrom, S.; Generalov, R.; Bonsdorff, T.B.; Larsen, R.H. Preparation of (212)Pb-labeled monoclonal antibody using a novel (224)Ra-based generator solution. Nucl. Med. Biol. 2017, 51, 1–9. [Google Scholar] [CrossRef]
  185. Larsen, R.H. Radiopharmaceutical Solutions with Advantageous Properties. U.S. Patent 9433690B1, 6 September 2016. [Google Scholar]
  186. Chappell, L.L.; Dadachova, E.; Milenic, D.E.; Garmestani, K.; Wu, C.; Brechbiel, M.W. Synthesis, characterization, and evaluation of a novel bifunctional chelating agent for the lead isotopes 203Pb and 212Pb. Nucl. Med. Biol. 2000, 27, 93–100. [Google Scholar] [CrossRef]
  187. Yong, K.; Brechbiel, M. Application of (212)Pb for Targeted alpha-particle Therapy (TAT): Pre-clinical and Mechanistic Understanding through to Clinical Translation. AIMS Med. Sci. 2015, 2, 228–245. [Google Scholar] [CrossRef] [PubMed]
  188. Nelson, B.J.B.; Andersson, J.D.; Wuest, F. Targeted Alpha Therapy: Progress in Radionuclide Production, Radiochemistry, and Applications. Pharmaceutics 2020, 13, 49. [Google Scholar] [CrossRef]
  189. Gholami, Y.H.; Willowson, K.P.; Forwood, N.J.; Harvie, R.; Hardcastle, N.; Bromley, R.; Ryu, H.; Yuen, S.; Howell, V.M.; Kuncic, Z.; et al. Comparison of radiobiological parameters for (90)Y radionuclide therapy (RNT) and external beam radiotherapy (EBRT) in vitro. EJNMMI Phys. 2018, 5, 1–19. [Google Scholar] [CrossRef]
  190. Mirzadeh, S.; Kumar, K.; Gansow, O.A. The chemical fate of 212Bi-DOTA formed by b-decay of 212Pb(DOTA)2. Radiochim. Acta 1993, 60, 1–10. [Google Scholar]
  191. Maaland, A.F.; Saidi, A.; Torgue, J.; Heyerdahl, H.; Stallons, T.A.R.; Kolstad, A.; Dahle, J. Targeted alpha therapy for chronic lymphocytic leukaemia and non-Hodgkin’s lymphoma with the anti-CD37 radioimmunoconjugate 212Pb-NNV003. PLoS ONE 2020, 15, e0230526. [Google Scholar] [CrossRef] [Green Version]
  192. Kratochwil, C.; Giesel, F.L.; Heussel, C.P.; Kazdal, D.; Endris, V.; Nientiedt, C.; Bruchertseifer, F.; Kippenberger, M.; Rathke, H.; Leichsenring, J.; et al. Patients Resistant Against PSMA-Targeting α-Radiation Therapy Often Harbor Mutations in DNA Damage-Repair-Associated Genes. J. Nucl. Med. 2020, 61, 683–688. [Google Scholar] [CrossRef] [PubMed]
  193. Chan, T.G.; O’Neill, E.; Habjan, C.; Cornelissen, B. Combination Strategies to Improve Targeted Radionuclide Therapy. J. Nucl. Med. 2020, 61, 1544–1552. [Google Scholar] [CrossRef]
  194. Kulkarni, H.; Zhang, J.; Langbein, T.; Schuchardt, C.; Singh, A.; Mueller, D.; Baum, R. Radioligand therapy using combination of Ac-225 and Lu-177 labelled PSMA ligands for progressive end-stage metastatic prostate cancer: Effective trade-off between response and toxicity. J. Nucl. Med. 2019, 60, 464. [Google Scholar]
  195. Roy, J.; Warner, B.; Basuli, F.; Williams, M.; Wong, K.J.; Ton, A.; Chiorini, J.; Choyke, P.; Lin, F.; Jagoda, E. Identifying an appropriate animal model to examine preservation of salivary function with PSMA targeted radiotherapies. J. Nucl. Med. 2018, 59, 1255. [Google Scholar]
  196. Simons, B.W.; Turtle, N.F.; Ulmert, D.H.; Abou, D.S.; Thorek, D.L.J. PSMA expression in the Hi-Myc model; extended utility of a representative model of prostate adenocarcinoma for biological insight and as a drug discovery tool. Prostate 2019, 79, 678–685. [Google Scholar] [CrossRef] [Green Version]
  197. Roy, J.; Warner, B.M.; Basuli, F.; Zhang, X.; Wong, K.; Pranzatelli, T.; Ton, A.T.; Chiorini, J.A.; Choyke, P.L.; Lin, F.I.; et al. Comparison of Prostate-Specific Membrane Antigen Expression Levels in Human Salivary Glands to Non-Human Primates and Rodents. Cancer Biother. Radiopharm. 2020, 35, 284–291. [Google Scholar] [CrossRef] [PubMed]
  198. Rupp, N.J.; Umbricht, C.A.; Pizzuto, D.A.; Lenggenhager, D.; Töpfer, A.; Müller, J.; Muehlematter, U.J.; Ferraro, D.A.; Messerli, M.; Morand, G.B.; et al. First Clinicopathologic Evidence of a Non-PSMA-Related Uptake Mechanism for (68)Ga-PSMA-11 in Salivary Glands. J. Nucl. Med. 2019, 60, 1270–1276. [Google Scholar] [CrossRef] [Green Version]
  199. Wang, X.; Ma, D.; Olson, W.C.; Heston, W.D. In vitro and in vivo responses of advanced prostate tumors to PSMA ADC, an auristatin-conjugated antibody to prostate-specific membrane antigen. Mol. Cancer Ther. 2011, 10, 1728–1739. [Google Scholar] [CrossRef] [Green Version]
  200. Cho, S.; Zammarchi, F.; Williams, D.G.; Havenith, C.E.G.; Monks, N.R.; Tyrer, P.; D’Hooge, F.; Fleming, R.; Vashisht, K.; Dimasi, N.; et al. Antitumor Activity of MEDI3726 (ADCT-401), a Pyrrolobenzodiazepine Antibody-Drug Conjugate Targeting PSMA, in Preclinical Models of Prostate Cancer. Mol. Cancer Ther. 2018, 17, 2176–2186. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  201. Fendler, W.P.; Stuparu, A.D.; Evans-Axelsson, S.; Luckerath, K.; Wei, L.; Kim, W.; Poddar, S.; Said, J.; Radu, C.G.; Eiber, M.; et al. Establishing (177)Lu-PSMA-617 Radioligand Therapy in a Syngeneic Model of Murine Prostate Cancer. J. Nucl. Med. 2017, 58, 1786–1792. [Google Scholar] [CrossRef] [Green Version]
  202. Lin, M.; Ta, R.T.; Kairemo, K.; Le, D.B.; Ravizzini, G.C. Prostate-Specific Membrane Antigen-Targeted Radiopharmaceuticals in Diagnosis and Therapy of Prostate Cancer: Current Status and Future Perspectives. Cancer Biother. Radiopharm. 2020. [Google Scholar] [CrossRef]
  203. Weineisen, M.; Schottelius, M.; Simecek, J.; Baum, R.P.; Yildiz, A.; Beykan, S.; Kulkarni, H.R.; Lassmann, M.; Klette, I.; Eiber, M.; et al. 68Ga- and 177Lu-Labeled PSMA I&T: Optimization of a PSMA-Targeted Theranostic Concept and First Proof-of-Concept Human Studies. J. Nucl. Med. 2015, 56, 1169–1176. [Google Scholar] [PubMed] [Green Version]
  204. Banerjee, S.R.; Kumar, V.; Lisok, A.; Chen, J.; Minn, I.; Brummet, M.; Boinapally, S.; Cole, M.; Ngen, E.; Wharram, B.; et al. (177)Lu-labeled low-molecular-weight agents for PSMA-targeted radiopharmaceutical therapy. Eur. J. Nucl. Med. Mol. Imaging 2019, 46, 2545–2557. [Google Scholar] [CrossRef] [PubMed]
  205. Ruigrok, E.A.M.; van Vliet, N.; Dalm, S.U.; de Blois, E.; van Gent, D.C.; Haeck, J.; de Ridder, C.; Stuurman, D.; Konijnenberg, M.W.; van Weerden, W.M.; et al. Extensive preclinical evaluation of lutetium-177-labeled PSMA-specific tracers for prostate cancer radionuclide therapy. Eur. J. Nucl. Med. Mol. Imaging 2020, 1–12. [Google Scholar] [CrossRef] [PubMed]
  206. Kulkarni, H.R.; Singh, A.; Schuchardt, C.; Niepsch, K.; Sayeg, M.; Leshch, Y.; Wester, H.J.; Baum, R.P. PSMA-Based Radioligand Therapy for Metastatic Castration-Resistant Prostate Cancer: The Bad Berka Experience Since 2013. J. Nucl. Med. 2016, 57, 97s–104s. [Google Scholar] [CrossRef] [Green Version]
  207. Hagemann, U.B.; Wickstroem, K.; Hammer, S.; Bjerke, R.M.; Zitzmann-Kolbe, S.; Ryan, O.B.; Karlsson, J.; Scholz, A.; Hennekes, H.; Mumberg, D.; et al. Advances in Precision Oncology: Targeted Thorium-227 Conjugates As a New Modality in Targeted Alpha Therapy. Cancer Biother. Radiopharm. 2020, 35, 497–510. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  208. Gosewisch, A.; Schleske, M.; Gildehaus, F.J.; Berg, I.; Kaiser, L.; Brosch, J.; Bartenstein, P.; Todica, A.; Ilhan, H.; Böning, G. Image-based dosimetry for (225)Ac-PSMA-I&T therapy using quantitative SPECT. Eur. J. Nucl. Med. Mol. Imaging 2020, 1–2. [Google Scholar] [CrossRef]
Figure 1. The simplified structure of prostate-specific membrane antigen (PSMA), its binding sites for PSMA ligands and antibodies (this figure is based on references [52,53,54]).
Figure 1. The simplified structure of prostate-specific membrane antigen (PSMA), its binding sites for PSMA ligands and antibodies (this figure is based on references [52,53,54]).
Cancers 13 00779 g001
Figure 2. Simplified decay chains of 149Tb, 211At, 225Ac, 227Th, 223Ra, and 224Ra radionuclides. α–alpha particle, β–beta particle, β–electron, β+–positron, ε–electron capture. Information extracted from the National Nuclear Data Center database, Brookhaven National Laboratory [75].
Figure 2. Simplified decay chains of 149Tb, 211At, 225Ac, 227Th, 223Ra, and 224Ra radionuclides. α–alpha particle, β–beta particle, β–electron, β+–positron, ε–electron capture. Information extracted from the National Nuclear Data Center database, Brookhaven National Laboratory [75].
Cancers 13 00779 g002
Table 1. Comparison of conventional external radiation radiotherapy and targeted radionuclide therapy [60,61,62]. AMU, atomic mass units LET, linear energy transfer; DBS, double-strand breaks; SSB, single-strand breaks.
Table 1. Comparison of conventional external radiation radiotherapy and targeted radionuclide therapy [60,61,62]. AMU, atomic mass units LET, linear energy transfer; DBS, double-strand breaks; SSB, single-strand breaks.
PropertiesTargeted Radionuclide TherapyExternal Radiation Therapy
α Emittersβ Emittersχ- and γ-rays
TypeHelium nucleusElectronPhoton
Mass (AMU)4 (heavy)0.0005 (light)0 (massless)
Speed of light6% (slow)98% (fast)100% (fast)
Initial energy4–9 MeV0.05–2.3 MeV0.035–3 MeV
Range in tissue0.04–0.1 mm0.05–12 mmcentimetres
LET50–230 keV/µm0.1–1.0 keV/µm0.2 keV/µm
DNA damageIrrepairable (DSB)Repairable (SSB)Repairable (SSB)
Ionizing abilityVery highMediumLow
Number of DNA hits to kill cells1–5100–2000>>1000
Irradiation fieldWhole-bodyWhole-bodyRegion
Dose distributionVery heterogeneousHeterogeneousHomogenous
Dose rate<1 Gy/h<1 Gy/h60–120 Gy/h, in 2 Gy fractions
Effect of oxygen on cell killingWeakModerateStrong
Bystander effectYesYesYes
Crossfire radiationYesYesYes
Table 2. Overview of α-emitting radionuclides used in PSMA-TAT [71,72,73,74,75].
Table 2. Overview of α-emitting radionuclides used in PSMA-TAT [71,72,73,74,75].
Radio-
Nuclide
Half-
Life
Main Production MethodRadio-
Nuclide Availability
Emitted ParticlesTotal α Energy Emitted per Decay (MeV)Range in Tissue (µm)LET (keV/µm)Emissions for Imaging
149Tb4.1 hAcceleratorModerate1α, 1ε/2ε, 1β+/2β+0.7 *25140165 and 730 keV
211At7.2 hCyclotronModerate1α, 1ε6.955–8071–23077–92 keV
212Pb/
212Bi
10.6 h224Ra generatorVery high1α, 2β7.940–10061–230238 keV
213Bi45.6 min225Ac generatorModerate2α, 2β8.540–10065–230440 and 1566 keV
223Ra11.4 days227Th generatorHigh4α, 2β26.846–7071–23084, 95, 144, 154 and 270 keV
225Ac9.9 days229Th generator; cyclotron, accelerator, etc. in the futureModerate4α, 2β27.947–8561–230218 and 440 keV
227Th18.7 days227Ac generatorHigh5α, 2β32.850–7071–23084, 95, 236 and 270 keV
* Average value per decay: 149Tb has alpha energy of 3.97 MeV but with only 16.7% abundance.
Table 3. Comparisons of prostate-specific membrane antigen (PSMA)-targeting small molecules, antibodies, and their fragments [55,76,77,78,79,80].
Table 3. Comparisons of prostate-specific membrane antigen (PSMA)-targeting small molecules, antibodies, and their fragments [55,76,77,78,79,80].
CharacteristicsSmall MoleculesAntibody FragmentsAntibodies
Molecular weight<1.5 kDa15–110 kDa150 kDa
StructurePeptidomimetic chainOne to four polypeptide chainsComplex; four polypeptide chains
ManufactureEasyDifficultDifficult
StabilityStableInstableInstable
In vivo half-lifeFew hours0.5–30 h2–7 days
PharmacokineticsRapid clearance from blood, rapid tissue penetrationRapid to intermediate clearance from blood, rapid to intermediate tissue penetrationCirculates long in blood, slow tissue penetration, longer tumor retention
ExcretionRenal clearanceRenal clearance of Ab fragments <70 kDa Hepatobiliary clearance and Fc-receptor-
mediated recycling
Target binding to the extracellular domainEnzymatic pocket of the catalytic domainApical region of the extracellular domainApical region of the extracellular domain
ImmunogenicitySeldomLowExpected
ExamplesPSMA-617, PSMA-I&T, NG001IAB2M, scFvD2B, JVZ-008, PSMA6 and 30J591, 107-1A4, PSMA-TTC
Table 4. Overview of α-emitting radionuclides and PSMA-targeting agents investigated in preclinical studies for targeted prostate cancer therapy. mAb, monoclonal antibody; NA, not available; Fab’, antigen-binding fragment; %ID/g, percent injected dose per gram of tissue. Therapeutic index (TI) is defined as the median survival of the treatment group divided by the median survival of the untreated group.
Table 4. Overview of α-emitting radionuclides and PSMA-targeting agents investigated in preclinical studies for targeted prostate cancer therapy. mAb, monoclonal antibody; NA, not available; Fab’, antigen-binding fragment; %ID/g, percent injected dose per gram of tissue. Therapeutic index (TI) is defined as the median survival of the treatment group divided by the median survival of the untreated group.
Radio-
Nuclide
PSMA Targeting AgentActivityMain ObservationsReferences
Preclinical studies in vitro
213BiJ591 (mAb)0–1.8 MBq/mLAntitumor activity, growth delay of LNCaP-LN3 spheroidsBallangrud et al., 2001 [81]
211AtABCPUPNABinds to PC3 PIP cellsVaidyanathan et al., 2009 [82]
227Ac 0–370 kBq/mLAntitumor activity (LNCaP, Mat-Lu cells)Bandekar et al., 2014 [83]
223RaNA-silane-PEG-D2B (mAb)0–20 kBq/mLAntitumor activity, LD50 ≈ 2.5kBq/mL
(C4-2 cells)
Czerwińska et al., 2020 [84]
Preclinical studies in vivo
149TbPSMA-6172 × 3 MBq37%ID/g (tumor targeting at 1 h, subcutaneous, PC3 PIP cells); antitumor activity, TI ≈ 1.8Umbricht et al., 2019 [85]
211At107-1A4 (mAb)370 kBqPSA decline (intratibial, C4-2B cells)Wilbur et al., 2009 [86]
107-1A4 Fab’740 kBq25%ID/g (tumor targeting at 1 h, subcutaneous, LNCaP cells)Wilbur et al., 2011 [87]
PSMA 6740 kBq14%ID/g (tumor targeting at 1 h, subcutaneous, PC3 PIP cells); antitumor activity, TI ≈ 2.1;
delayed nephropathy dose limiting;
dehalogenation in vivo
Kiess et al., 2016 [88]
131I as a surrogate for 211AtRPS-027NADual targeting to PSMA and albumin; 9%ID/g (tumor targeting at 1 h, subcutaneous, LNCaP cells) Kelly et al., 2017 [89]
16bNA15%ID/g (tumor targeting at 1 h, subcutaneous, C4-2B cells)Li et al., 2020 [90]
212Pb/
212Bi
CA009, CA012NA25%ID/g (tumor targeting at 1 h (203Pb) subcutaneous, C4-2 cells)Dos Santos et al., 2019 [91]
L21.5 and 3.7 MBq22%ID/g (tumor targeting at 1 h (203Pb), subcutaneous, PC3 PIP cells); antitumor activity, TI ≈ 1.9 and TI ≈ 3Banerjee et al., 2020 [92]
NG001320 kBq22%ID/g (tumor targeting at 1 h, subcutaneous, C4-2 cells); antitumor activity, TI * ≈ 2.3Larsen, 2019 [93]; Stenberg et al., 2020 [94,95]
213BiJ591 (mAb)3.3 MBqAntitumor activity, TI ≈ 1.8 (subcutaneous, LNCaP cells)McDevitt et al., 2000 [96]
3.7 MBqAntitumor activity, inhibition of tumor growth in mice (subcutaneous, LN3 cells)Li et al., 2002 [97]
PSMA I&T5.4–6.6 MBq5.8%ID/g (tumor targeting at 1 h, subcutaneous, LNCaP cells)Nonnekens et al., 2017 [98]
JVZ-008 (nanobody)4.5–5.4 MBq2.7%ID/g (tumor targeting at 1 h, subcutaneous, LNCaP cells)
225AcPSMA-61740 kBqAntitumor activity (subcutaneous, C4-2 cells); weight loss at 100 kBqMeyer et al., 2019 [99]
40 kBqAntitumor activity (intravenous, C4-2 cells), TI ≈ 3.9Stuparu et al., 2020 [100]
40 kBqAntitumor activity, (subcutaneous, RM1-PSMA+++ cells)Current et al., 2020 [101]
30 kBqAntitumor activity (subcutaneous, RM1-PGLS cells), TI ≈ 1.2Czernin et al., 2020 [102]
RPS-074148 kBq6%ID/g (tumor targeting at 4 h, subcutaneous, LNCaP cells); antitumor activity, complete response in 86%Kelly et al., 2019 [103]
227ThPSMA-TTC (mAb)100–500 kBq/kg20%ID/g and 37%ID/g (tumor targeting after 3 and 7 days, respectively, subcutaneous, MDA-PCa-2b cells); antitumor activityHammer et al., 2020 [104]
* Therapeutic index (TI) is defined as 75% survival of the treatment group divided by the 75% survival of the untreated group.
Table 5. Overview of α-emitting radionuclides and PSMA-targeting agents investigated in clinical studies for targeted prostate cancer therapy. n, number of patients; PFS, progression-free survival; OS, overall survival; -, activity de-escalation in subsequent cycles.
Table 5. Overview of α-emitting radionuclides and PSMA-targeting agents investigated in clinical studies for targeted prostate cancer therapy. n, number of patients; PFS, progression-free survival; OS, overall survival; -, activity de-escalation in subsequent cycles.
PSMA-
TAT
nActivity per CyclePSA Decline After TAT (Patients)Median PFS/OS (Months)ToxicityReferences
≤0%≥50%
213Bi-
PSMA-
617
1296 MBq 100% (1/1)NANASathekge et al., 2017 [105]
225Ac-
PSMA-
617
2100 kBq/kg 100% (2/2)NAXerostomiaKratochwil et al., 2016 [42]
1450–200 kBq/kg22% (2/9)44% (4/9)NA/8.5XerostomiaKratochwil et al., 2017 [106]
40100 kBq/kg13% (5/40)63% (24/38)NA/>12XerostomiaKratochwil et al., 2018 [107]
18 MBq 100% (1/1)NANASathekge et al., 2019 [108]
178–4 MBq6% (1/17)88% (15/17)NAXerostomiaSathekge et al., 2019 [109]
18–6 MBq 100% (1/1)NAXerostomia xero-
phthalmia
De Medeiros et al., 2019 [110]
738–4 MBq18% (13/73)70% (51/73)15.2/18.0NASathekge et al., 2020 [111]
268–4 MBq11% (3/26)65% (17/26)3.5/7.7Xerostomia, anemia, leucopenia, thrombopeniaFeuerecker et al., 2020 [112]
28100 kBq/kg18% (5/28)39% (11/28)12/17Transient fatigue, xerostomiaYadav et al., 2020 [113]
2NANANANAChronic kidney diseasePelletier et al., 2021[114]
138–6 MBq15% (2/13)69% (9/13)NA/8.5XerostomiaVan der Doelen et al., 2020 [115]
225Ac-
PSMA I&T
18 MBq 100% (1/1)NAXerostomiaIlhan et al., 2020 [116]
147.8 MBq21% (3/14)50% (7/14)NAXerostomiaZacherl etal, 2020 [117]
Table 6. Ongoing clinical trials with targeted α therapies (TAT) targeting PSMA in metastatic castration-resistant prostate cancer (mCRPC) patients registered in https://clinicaltrials.gov/ (accessed on 14 January 2021).
Table 6. Ongoing clinical trials with targeted α therapies (TAT) targeting PSMA in metastatic castration-resistant prostate cancer (mCRPC) patients registered in https://clinicaltrials.gov/ (accessed on 14 January 2021).
Trial IDPhaseTATNumber of PatientsPeriodSponsor
NCT032765721225Ac-J591422017–2021Weill Medical College of Cornell University
NCT045065671/2225Ac-J5911052020–2025Weill Medical College of Cornell University
NCT042259101225Ac-PSMA202019–2021Xinhua Hospital, Shanghai Jiao Tong University School of Medicine
NCT045974111225Ac-PSMA-617302021–2022Novartis Pharmaceuticals
NCT037247471BAY 2315497 (227Th-mAb)1572018–2023Bayer
Table 7. PSMA expression in prostate cancer cell lines. N/A, not applicable; negative, below the lower limit of quantification.
Table 7. PSMA expression in prostate cancer cell lines. N/A, not applicable; negative, below the lower limit of quantification.
Cell LineNumber of PSMA Per CellReferences
DU145Negative[199,200]
PC3Negative[199,200]
22Rv115,000[101]
RM1-h-PSMA19,000[201]
LS174T-PSMA43,000[138]
PSMA++ RM149,000[101]
RM1-PGLS56,000[201]
MDA PCa2b118,000[200]
LNCaP126,000–250,000[96,138,199]
C4-2102,000–255,000[101,199,201]
PC3 PIP552,000[101]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Juzeniene, A.; Stenberg, V.Y.; Bruland, Ø.S.; Larsen, R.H. Preclinical and Clinical Status of PSMA-Targeted Alpha Therapy for Metastatic Castration-Resistant Prostate Cancer. Cancers 2021, 13, 779. https://0-doi-org.brum.beds.ac.uk/10.3390/cancers13040779

AMA Style

Juzeniene A, Stenberg VY, Bruland ØS, Larsen RH. Preclinical and Clinical Status of PSMA-Targeted Alpha Therapy for Metastatic Castration-Resistant Prostate Cancer. Cancers. 2021; 13(4):779. https://0-doi-org.brum.beds.ac.uk/10.3390/cancers13040779

Chicago/Turabian Style

Juzeniene, Asta, Vilde Yuli Stenberg, Øyvind Sverre Bruland, and Roy Hartvig Larsen. 2021. "Preclinical and Clinical Status of PSMA-Targeted Alpha Therapy for Metastatic Castration-Resistant Prostate Cancer" Cancers 13, no. 4: 779. https://0-doi-org.brum.beds.ac.uk/10.3390/cancers13040779

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop