Next Article in Journal
Sesquiterpenoids from Inula britannica and Their Potential Effects against Triple-Negative Breast Cancer Cells
Next Article in Special Issue
Production Review of Accelerator-Based Medical Isotopes
Previous Article in Journal
Comparison of the Performance of Different Bile Salts in Enantioselective Separation of Palonosetron Stereoisomers by Micellar Electrokinetic Chromatography
Previous Article in Special Issue
Production and Quality Control of [177Lu]Lu-PSMA-I&T: Development of an Investigational Medicinal Product Dossier for Clinical Trials
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Radiopharmaceutical Treatments for Cancer Therapy, Radionuclides Characteristics, Applications, and Challenges

1
Radiology and Medical Imaging Department, Fatima College of Health Sciences, Abu Dhabi 3798, United Arab Emirates
2
National Cancer Institute, University of Gezira, Wad Madani 2667, Sudan
3
Nuclear Medicine Division of Radiology Department, Faculty of Medicine, Universitas Andalas, Padang 25163, Indonesia
*
Author to whom correspondence should be addressed.
Molecules 2022, 27(16), 5231; https://doi.org/10.3390/molecules27165231
Submission received: 26 June 2022 / Revised: 9 August 2022 / Accepted: 10 August 2022 / Published: 16 August 2022
(This article belongs to the Special Issue Radiopharmaceuticals)

Abstract

:
Advances in the field of molecular biology have had an impact on biomedical applications, which provide greater hope for both imaging and therapeutics. Work has been intensified on the development of radionuclides and their application in radiopharmaceuticals (RPS) which will certainly influence and expand therapeutic approaches in the future treatment of patients. Alpha or beta particles and Auger electrons are used for therapy purposes, and each has advantages and disadvantages. The radionuclides labeled drug delivery system will deliver the particles to the specific targeting cell. Different radioligands can be chosen to uniquely target molecular receptors or intracellular components, making them suitable for personal patient-tailored therapy in modern cancer therapy management. Advances in nanotechnology have enabled nanoparticle drug delivery systems that can allow for specific multivalent attachment of targeted molecules of antibodies, peptides, or ligands to the surface of nanoparticles for therapy and imaging purposes. This review presents fundamental radionuclide properties with particular reference to tumor biology and receptor characteristic of radiopharmaceutical targeted therapy development.

1. Introduction

In the early 1900s, Henri Becquerel and Marie Curie discovered radioactivity. Therapeutic applications immediately followed this discovery [1,2]. For many years radionuclide therapy was limited to the use of Iodide-131 (131I) for thyroid cancer and hyperthyroidism and phosphate-32 (32P) for polycythemia vera [2,3,4,5,6]. Radionuclides labeled molecules such as a drug, a protein, or a peptide that operate as a delivery vehicle that accumulates and binds to specific targets such as tumors or other undesirable cell proliferation [3,7,8]. The development of radionuclide use has been growing exponentially with the introduction of more new radiopharmaceuticals (RPs) for therapy and imaging.
In recent times, RPs use in nuclear medicine has become popular in theranostics. These are used in therapeutic interventions after imaging verifies the presence of a biological target [6,9,10]. Unlike radiotherapy, RPs are administrated intravenously to be delivered to a target tumor or associated structure. RPs have advantages in treating systemic malignancy in areas such as the bone or brain, which are impossible to treat using external radiotherapy [2]. The targeted tumor cell absorbs a dose of radiation from an RP which exponentially decreases over time (Figure 1a). On the other hand, in external radiotherapy, radiation beams are directed at tumor tissue and cannot avoid healthy cells (Figure 1b).
Radiopharmaceutical therapy (RPT) is a novel modality that can be effective with minimal toxicity [6,7]. The advantages of RPT are, firstly, it can be targeted at tumors, including metastasis sites. The RPs can be used in radiotracer imaging to determine the uptake of the RP in the target tissues before administering a therapeutic dose. Secondly, a wide variety of radionuclides are now available emitting different types of radiation at different energies. For instance, high linear energy transfer (LET) radionuclides are used effectively to kill resistant hypoxic cells. Thirdly, this therapy allows for a relatively lower whole-body absorbed dose [7,10,11,12,13].
RPT can be used as adjuvant therapy with or after other treatment options such as chemotherapy and surgery [2]. It is being used to control symptoms and shrink and stabilize tumors in systemic metastatic cancer, where conventional therapy or chemotherapy is impossible. RPT can be a good choice, especially for patients who no longer respond to other treatments [2,3,7,10]. This review describes some fundamental radionuclide properties with particular reference to tumor biology and the receptor characteristics of radiopharmaceutical targeted therapy development.

2. Radionuclide Emission Properties

The physical characteristics of a radionuclide should be considered when selecting it for therapy purposes. These include physical half-life, radiation energy, type of emissions, daughter product(s), production method, and radionuclide purity [2,9]. Ideally, the physical half-life of the radionuclide should be between 6 h and seven days [14]. The RPs with a long half-life will expose the target tumor and surrounding environment to radiation for longer. However, RPs with a very short physical half-life have limitations due to the delivery time. There must be sufficient retention time for the emission to be delivered to the tumor target [15].
Furthermore, in vivo stability, toxicity, and the biological half-life within the patient’s body must be considered [7,16], along with the type and size of the tumor, method of administration, and uptake mechanism [1,2,6,15]. The tumor uptake mechanism is specific to the target cell. It depends on processes such as antigen–antibody reactions, physical particle trapping, receptor binding sites, removal of damaged cells from circulation, and transportation of a chemical species across a cell membrane and metabolic cycle [2,17]. The condition will influence the ratio of the concentration of radionuclides in the tumor to that in normal tissues. This ratio should be optimized [2]. The other factors that must be considered are radionuclide particle size, toxicity, specific gravity for optimal flow and distribution, and clearance rate [2,6,18,19,20,21,22].
Radionuclides used in RPT are primarily beta (β)-particle (0.2 keV/μm) or alpha (α)-particle (50–230 keV/μm) emitters [2,9,11,15,23], and Auger electrons (AE) (4–26 keV/μm) [2,9,11,15,23,24]. Various radionuclides and their characteristics are summarized in Table 1. Each of these radiation types results in ionization along the travel length, and they are fully deposited in the cell [16]. The radiation destroys the cell directly and indirectly [6,25]. The distance traveled by particles and the energy deposited in cells must be considered to ensure optimal targeted cell destruction and minimize ionization interaction with healthy cells [2,6,7,15].

2.1. Beta Particles

Beta particles have been used in cancer therapy over the last 40 years [6]. They are the product of the β decay process, wherein an unstable nucleus is converted to a proton, and a β particle, a high-energy electron [7,26]. β particles are the most frequently used radiation in RPT agents and are widely available [7]. β particles are negatively charged. They have a relatively long path from 0.0 to 12 mm, and some emit a gamma (γ) ray such as 32P, 89Sr, 90Y, and 169Er [3]. They emit γ ray <10%, which is acceptable for imaging to confirm the tumor uptake and biodistribution and dosimetric calculations [2,3]. They have a low linear energy transfer (LET) of approximately 0.2 keV/μm, so more β particles are required to deliver a similar absorbed dose compared to alpha particles.
The most familiar and frequently used β particle is iodine-131 (131I). Hertz and Roberts used radioiodine I-130 (130I) for hyperthyroid therapy in 1941, which rose at the birth of nuclear medicine [27,28,29]. In August 1946, 130I was replaced by 131I because it was much cheaper [27,29]. 131I is a β and γ emitter with a half-life of 8.05 days. The β particle has a peak energy of 0.606 MeV, with a maximum range of ~3 mm in the tissue, and it is used for therapy. The peak energy of the γ ray is 0.364 MeV and is used for imaging [27]. Since then, 131I has been used countless times for therapy for hyperthyroid and thyroid cancer [3,6,27,28,29,30]. In 1981, 131I-iobenguane (meta-iodobenzylguanidine, MIBG) was introduced as a diagnostic agent, and in 1984, it was used for treating malignant phaeochromocytoma [31]. Monoclonal antibodies are used to label with 131I, and, in 2003, FDA approved 131I-tositumomab (Bexxar) for the treatment of refractory non-Hodgkin’s lymphoma (NHL) [2,6,7]. Several studies have reported the monoclonal antibodies labeled on other beta particle emitters, including Yttrium-90 (90Y) and Lutetium-177 (177Lu), for more effective therapy purposes [2,7,31,32,33].
The high-energy β from Yttrium-90 (90Y) or Rhenium-188 (188Re) is preferable for treating higher volume solid and poorly perfused tumors and is less suited for targeting micro-metastases to avoid crossfire doses to neighbor cells [9,11,34]. 90Y, widely available like 131I, is a popular radionuclide for liver cancer and metastases [35,36]. Neuroendocrine tumors (NETS) have been treated with radionuclide therapy (PPRT) targeting peptide receptors with radiopharmaceuticals labeled with 90Y. Antibodies also labeled with 90Y, have been introduced for ovarian and hematological cancers [7,26,37,38,39]. Low-energy ẞ, like those seen with lutetium-177 (177Lu), is more efficient for small tumors [1,9]; hence, 177Lu is becoming a popular ẞ-particle source for treating small tumors [7,9]. 177Lu has a half-life of 6.73 days and is compatible with antibodies and peptides [40,41]. Furthermore, it also emits gamma-rays and can be detected externally as a theranostic agent [1,7,40]. Samarium-153 (153Sm) is used to treat palliative bone metastases and other primary cancers [3,42,43]. Ethylenediamine-tetra-methylene-phosphonic acid (EDTMP) chelator binds with 153Sm through six ligands (four phosphate groups and two amines). It has been widely used since FDA approval in various osteoblastic metastatic lesions, especially in prostate and breast cancer [44]. However, not all possible β particle sources have been widely adopted because of the complexity of the radiochemistry or the absence of commercial availability. The decision to use one β-particle source over another must consider the absorbed dose ratio between tumor to non-tumor tissue [7].

2.2. Alpha Particles

The application of targeted α particle therapy (TAT) gained approval in 2013 [19]. Alpha particles are high energy and have shorter path lengths, resulting in higher efficacy in some applications [2,8,15,25,26]. TAT is an attractive therapeutic option for multiple micro-metastases. It is easy to administer and can be used to treat multiple lesions simultaneously. It is also possible to combine it with other therapeutic approaches, primarily for cancer treatment [45,46].
An alpha particle is a 4He nucleus without its surrounding electrons (sometimes denoted as (He2+)) [26,45]. Alpha radiation is emitted from the nucleus of a radioactive atom undergoing decay with an energy is 4–9 MeV, and the particles travel only 1–3 cell diameters (40–100 μm) in tissue [7,15,32,45,46]. The particles have high LET (60–230 keV/μm) throughout their range, peaking to three times the initial value at the end of the path range (the Bragg peak) [16,26,32]. Most alpha particles also emit gamma-ray. However, treatment planning or post-therapeutic imaging using alpha particles is not performed yet in clinical settings due to technical limitations [45].
Furthermore, intracellular accumulation of the α particles effectively creates double-strand breaks (DSBs) in DNA, and numerous clusters of DSBs in target cells, making cellular repair systems ineffectual [7,32,47]. The cytotoxicity of α-particles is much higher than that of β-particles due to the particle deposit energy per unit path length, which is 1500 times more than beta particles [45,48]. In addition, the short travel distance of α particles reduces the damage to surrounding healthy tissue [15,49]. The particle radiation has been demonstrated to be independent of cell oxygen concentration [15,32,45,50]. The physical and biological characteristics of alpha, beta particles, and Auger electrons are summarized in Table 2, and DNA damage by that radiations are illustrated in Figure 2a,b.
Improvements in understanding molecular tumor biology, labeling techniques, technology development, and other related disciplines have paved the way for significant new clinical applications of α radiation as a novel therapeutic agent [7,15,51]. Alpha particle-labeled biological molecules such as monoclonal antibodies (mAb) allow close radiation targeting and selectively deliver high radiation to the target, with limited toxicity to normal tissues [15]. The mAbs are labeled radionuclides that bind to the extracellular domain of PSMA, demonstrating promising results in imaging and therapy of prostate cancers [9]. The monoclonal antibodies are labeled with bismuth-213 (213Bi) and astatine-211 (211At) and are used to treat leukemia and brain tumors [11,52]. The monoclonal antibody MX35 labeled 213Bi successfully treated ovarian cancer in animal models with no signs of toxicity [53]. 213Bi has a short half-life and is produced using a generator and labeling to produce TAT compounds is therefore completed on-site [26,54]. Because of its short half-life, 213Bi needs to be delivered directly into tumor tissue, and it can be given at a high dose over a short period, which is more effective than low dose rates given over a more extended period [26,32,55]. 213Bi has been used to label DOTA peptides in preclinical and clinical trials with >99% purity [15,26]. In preclinical and clinical studies, 213Bi and 225Ac have been used to label somatostatin receptors [15,26,32].
Radium-223 dichloride (Xofigo), a α particle emitter used for bone pain palliation in prostate and breast cancer patients, was approved by FDA in 2013 [7,11,26,32,45]. The emission energy of 223Ra can generate irreparable DNA double-strand breaks in the adjacent osteoblasts and osteoclasts, which has a detrimental effect on the adjacent cells and inhibits abnormal bone formation [7]. 223Ra is being studied as a radioactive label for other cytotoxic agents such as poly (ADP-ribose) polymerase inhibitors (olaparib), docetaxel (DORA trial), and new androgen axis inhibitors as enzalutamide and abiraterone citrate. The recently high number of 223Ra and in combination with other therapeutics, showed promising results [7].
Another alpha particle attracting increasing interest is 225Ac, the parent of 213Bi, which is relatively long-lived, with a half-life of 9.9 days [54]. 255Ac is produced via the neutron transmutation of 225Ra or decay of 233U [26,54,55]. 225Ac can be used to treat neuroendocrine tumors. It has been used to label PSMA with a radiochemical purity of >98% for prostate cancer therapy [26,54,56]. It also labeled antibodies to test for myeloid malignancy [9] and shows a potential for therapy, and post-therapy imaging, even though the images are suboptimal [26,32,55]. Results of clinical trials using TAT results indicate that this treatment strategy presents a promising alternative to targeted cancer therapy [52]. Lately, 225Ac-labeled PSMA-ligands have gained popularity as an alternative to 177Lu-PSMA [26,54,56]. However, 225Ac may damage the healthy cells due to daughter radionuclides such as 221Fr, 217At, and 213Bi [47]. Danger from radiation from daughter radionuclides needs to be carefully evaluated.
Table 2. Physical and biological characteristics of α, β particles, and Auger electron.
Table 2. Physical and biological characteristics of α, β particles, and Auger electron.
Alpha ParticleBeta ParticleAuger Electron
Type of particles 4He nucleusEnergetic electronLow energy electron; electron capture (ec) and/or internal conversion (ic)
Particle energy4–9 MeV50–2300 keV25–80 keV
Particle path length40–100 μm0.05–12 mmNanomicrometers
Linear energy transfer~80 keV/μm~0.2 keV/μm4–26 keV/μm
Hypoxic tumorsEffectiveLess effectiveEffective
ToxicityEffective in creating double-strand breaks in DNAHigh dose rates (tumor survival rates close to linear exponential). Low dose rates (single-strand breaks), repairable with shouldering the dose-response curvePotential creation of double-strand breaks DNA, and cell membrane
Bystander effect/crossfireYes/lowYesYes
Tumor sizeMicro/smallHigher volume solid tumorMicro
Ref: [7,8,13,24,25,26,32,47,50,55,57].
Figure 2. High and intermediate LET radiation (alpha particle and Auger electron, respectively), cause double-strand breaks in DNA (a). Single-strand breaks in DNA due to radiation by low LET (beta particle) (b).
Figure 2. High and intermediate LET radiation (alpha particle and Auger electron, respectively), cause double-strand breaks in DNA (a). Single-strand breaks in DNA due to radiation by low LET (beta particle) (b).
Molecules 27 05231 g002

2.3. Auger Electrons

Auger electrons (AE) have an even shorter range than alpha particles delivering radiation of only 1–1000 nm through the tissue causing potent tumor cell death if they can be conjugated with suitable ligands that effectively target micro-metastasis, particularly of DNA and cell membranes [2,11,24,32,34,52]. AEs are generated from suborbital transitions, and their range depends on their energy. They have an intermediate LET (4–26 keV/μM) [4,32,58]. Bromine-77 (77Br), indium-111 (111In), iodine-123 (123I), and iodine-125 (125I) are the most commonly used radionuclides sources [24,59,60]. Human studies using locoregional administration have shown promising results in therapy [7].
Despite the short range of AE, local cross-dose effects occur in cells adjacent to the radionuclide decay mediated by the several micrometer ranges of higher energy AEs and internal conversion (IC) that causes the death of distant non-irradiated cells through the bystander effect [16,24,25,27,32]. Lethally damaged tumor cells may release mediators that cause the death of distant non-irradiated cells [24,25]. Radiation also releases heat shock protein-70 and high mobility group box-1, which activate the dendritic cells (DCs). The activated DCs activate cytotoxic T cells that result in tumor regression at distant sites [11,61]. It has been observed that the effects of ionizing radiation can work synergistically with targeted immune treatment observed at the site(s) distant from targeted tissues/organs. This phenomenon is suggestive of the role of the abscopal effect [11,25,61,62]. Attention has been focused on delivering AE to the nucleus/DNA as the primary cellular radiation target to maximize toxic effects. However, cell membrane targeting has also been proven to be an effective strategy for killing cancer cells [24,63]. Cell membrane damage further induces γH2AX foci in the nucleus of the cells exposed to 125I-anti-CEA mAbs and in non-exposed cells through a bystander effect. 125I-labeled anti-CEA 35A7 was also found to be effective in vivo for treating small peritoneal tumors in mice [24,63]. Toxicity may also be induced indirectly by free radical-mediated pathways [24,25,57].
So, AE nuclear targeting is essential but not always required for RPT [24]. The abscopal and indirect killing effects suggest that targeting cell surface antigens overexpressed on tumor cells that are recognized by monoclonal antibodies (Mabs) or other ligands may be effective [25,64]. AE therapy has not been widely adopted yet. The fact that auger electron agents are incorporated into the DNA, and the cell membrane results in unfavorable pharmacokinetics, might be the reason for the lack of efficacy. Technological developments could overcome obstacles and increase interest in AE for therapy development [7].

3. Therapy Application

Radiopharmaceutical or radioligand therapy includes systemic radiation therapy, molecular radiotherapy, targeted radiation therapy, or peptide receptor radionuclide therapy (PRRT) and there are examples of where RPT is applied in optimizing and balancing the therapeutic index (TI). Various radioligands are being developed and investigated to target molecular receptors or intracellular components in personal therapy [58].

3.1. Antibodies

The monoclonal antibodies (mAbs) are labeled with radionuclides. The smaller fragments and new fusion proteins are directed against tumor antigens to deliver radionuclides to the targeted tumor [2,9]. The FDA has approved these agents for the clinical management of liquid malignancies (ibritumomab tiuxetan (Zevalin) labeled with 90Y, and tositumomab (Bexxar) labeled with 131I) is used for lymphoma therapy [2,6,7,55,64,65], and some the RPT in optimizing and balancing the therapeutic index (TI) [65]. The therapeutic benefit is achieved when the cells absorb continuous radiation emitted by radionuclides tagged to mAbs while minimizing toxicities in non-target tissues.
The effect of the RPT depends on the radiation’s energy and the antibody’s affinity, antigen target concentration on the cells, tissue vascularity, and antibody/antigen rate constants [64]. Novel antibody engineering techniques have enabled the development of antibodies that bind to antigens expressed in target cancer cells. An antibody that binds to a particular antigen will allow for a higher RP uptake within tumor tissue. However, antibodies are larger molecules, limiting the tumor penetration and distribution of the radiolabeled antibody within the tumor. Furthermore, antibodies have a prolonged circulation time and slow biological clearance, leading to larger radiation-absorbed doses to healthy organs and blood. Pre-infusion of a certain mass of non-radiolabeled antibody (cold antibody) may be used before the infusion of radiolabeled (hot antibody) to saturate antigenic sites in normal cells to avoid unnecessary radiation to healthy cells [64], reducing the binding of the hot antibody and decreasing the radiation doses to healthy organs. However, a pre-infusion time before administration of the hot antibody must be determined and optimized for every therapy [64].
Patient selection for RPT should be based on the predetermined expression of specific tumor antigens or diagnostic results [65]. Several antigens or receptors are expressed on the surface of the membrane of tumor cells, such as human epidermal growth factor receptor 2+ (HER2+), epidermal growth factor receptor (EGFR), CD20, prostate-specific membrane antigen (PSMA), vascular endothelial growth factor (VEGF), mucin 1 (MUC1) and tumor necrosis factor (TNF). Any of these can be labeled with various radionuclides [9]. Beta particle emitters have often been labeled with antibodies because they emit β and γ rays and have a longer half-life of 8 days. Lately, alpha particles have rapidly gained interest and have been used to label antibodies to deliver radiation to tumors, such as 227Th-anti CD22 and 225Ac-PSMA-617 [55]. However, α particles cannot be imaged unless they emit γ rays as 223Ra and 227Th do [64]. Unfortunately, these radionuclides only emit γ rays in low concentrations, which is not optimal for assessment. This imaging limitation may lead to noncompliance, and other radionuclides imaging may be required to establish lesion targeting and dosimetry [45,64].
Radionuclide-labeled mAbs demonstrate more efficacy in inducing cancer remissions than unlabeled molecules and are also more effective than chemotherapy [9]. They have been shown to benefit lung, pancreatic, stomach, ovarian, breast, colorectal, leukemia, and high-grade brain glioma cancers [2]. Fortunately, the application of the RPT in giant solid tumors is less successful than in small volume tumors such as malignant lymphoma due to poor perfusion, increased intratumoral hydrostatic pressure, and various radionuclides uptakes by the cells [8,64,65].

3.2. Prostate-Specific Membrane Antigen (PSMA)

131I-labeled prostate-specific membrane antigen (PSMA) ligands showed promise for prostate cancer therapy and were further developed to ”the 177Lu-PSMA” introduced in 2015 [60]. PSMA is a transmembrane protein that is over-expressed in prostate cancer (PC) cells, and its expression increases progressively in higher-grade cancers such as metastatic castration-resistant prostate cancer (mCRPC) PC [56,66,67,68]. Its benefits remain high even after multiple lines of therapy [56,66]. Radionuclide PSMA is a promising therapeutic approach for mCRPC patients for whom chemotherapy has been ineffective [55,56,66,69]. Early reports show that 177Lu-PSMA is safe and effectively reduces the tumor burden. It has low toxicity [69] and has become popular, with more than a thousand therapy cycles performed [66,69]. Severe hematological side effects are rare. Organs at risk after treatment with 177Lu-PSMA, including the salivary glands and the kidneys. However, the radiation dose to bone marrow, spleen, and liver is below critical limits [68].
Currently, the two most frequently used PSMA ligands are PSMA-617 and PSMA-I&T (imaging and therapy), labeled with 177Lu [68]. PSMA- targeting ligands using 225Ac maybe have an advantage compared to PSMA-targeting ligands using β particles. Clinical studies using 225Ac-labeled PSMA-ligands (PSMA-617 or PSMA-I&T) have demonstrated remarkable therapeutic results recently. Data on treatment with 225Ac-PSMA-617 indicate an excellent effect on tumor control in both early and late-stage mCRPC [70]. A novel α particle treatment with a 227Th-PSMA has shown potency in in vitro studies and efficacy in xenograft models of prostate cancer [8,67]. However, α particles have a more significant radiobiological effect on the organs at risk [56]. Concerns have been raised about treatment-associated, mostly permanent xerostomia, frequently leading to treatment discontinuation in many patients [56,68]. Combining α particles with β particle emitters is called “tandem therapy” and may reduce these significant adverse effects compared to using α particles alone [56,71,72].

3.3. Peptide Receptor Radionuclide Therapy (PRRT)

Receptor-based radionuclide therapies (PRRT) targeting the somatostatin receptor (SSTR), have since early 1990 been an important treatment modality for neuroendocrine tumors [7,26]. The efficacy of PPRT therapy might be due to the somatostatin receptor ligand that binds the specific receptor (SSTR1–5) [30,73]. Peptide receptors expressed in various tumor cells, including NETs, are significantly higher than in normal tissues or cells. NETs overexpress the SSTR2 potential for SSTR2 targeted therapies such as synthetic somatostatin analogs (SSAs) and radio-peptides or PRRT [30,73], and SSTR2 is primarily targeted by PRRT [73]. Octreotide and lanreotide are two SSAs developed and employed for clinical practice, which bind primarily to SSTR2 and SSTR5 [73]. Peptides have been labeled with several radionuclides, such as beta particles emitter 177Lu and 90Y. 177Lu–SSTR ligand is more effective in small-sized tumors, whereas, for larger tumor volumes, 90Y might be a better choice [30,73]. The first agent used was 90Y-labeled DOTATOC and DOTATATE. However, significant permanent kidney damage has been reported [34,74]. 177Lu-labeled DOTATATE or DOTATOC was the next PRRT radiopharmaceutical, causing less nephrotoxicity compared to 90Y [26] and a more negligible crossfire effect, particularly on small and metastatic tumors [74]. 177Lu-DOTATATE (Lutathera®) has also become the most widely used PRRT radiopharmaceutical at present [34].
Overall, α-emitters PRRT has shown good results. However, crossfire effects on small-size tumors have a significant impact. Additionally, hypoxia tumor tissue could be resistant to β-emitters treatment. α particles with high LET over a short range can minimize damage to surrounding healthy tissue. 213Bi and 225Ac have been clinically tested for brain tumors, neuroendocrine tumors, and prostate cancer therapy [26]. 213Bi and 225Ac-DOTA chelated peptides have been developed for peptide receptor radiotherapies, such as DOTA-Substance P targeting the neurokinin-1 receptor and somatostatin-analogs (e.g., DO-TATOC, DOTATATE) [74]. However, the results from these agents need to be confirmed in further studies.

3.4. Radioiodine Concentration via Sodium Iodide Symporter

131I has been used for adjuvant therapy to manage well-differentiated thyroid cancer (DTC) for more than 60 years. It is used to destroy remaining thyroid cells post-thyroidectomy, including in metastases, and is relatively inexpensive and widely available [14,75]. It increases the 10-year survival rate to 80% and decreases mortality by 12% [75]. One-third of advanced DTC metastases show low uptake of iodine. Losing the ability to accumulate iodine can occur during the progression of the disease due to dysfunction and loss of sodium iodide symporter (NIS) expression [75,76], indicating a status of dedifferentiation known as a radioiodine refractory disease [75,76,77].
A sodium iodide symporter (NIS) transports iodine through the cell membrane. Iodine is transported into follicular thyroid cells against the electrochemical gradient [27,75,76]. In a normal condition, the gradient between a thyroid cell and the extracellular environment is 100:1 [27,75]. The expression of NIS provides the molecular basis of radioiodine for diagnostic and therapeutic use in patients with thyroid disease [76,77,78]. It resides in the thyroid in the basolateral membrane of epithelial cells and transports two cations of sodium (Na+) and one anion of iodide (I-) into the cells. This process is facilitated by an enzyme Na+/K+ ATPase [27,29,75,76].
Genetic alteration causes the mitogen-activated protein kinase (MAPK) and phosphoinositide 3-kinase (PI3K) pathways associated with the silencing of solute carrier family five-member 5 (SLC5A5), which encodes NIS. The condition causes the cancer cell failure to take radioiodine [77]. A clinical trial of kinase inhibitors targeting the MAPK or PI3K pathways has shown promising effects in redifferentiation therapy. It brings hope to future therapy using either kinase inhibitors with different targets or kinase inhibitors and 131I in managing radioiodine refractory disease in DTC [77].
Furthermore, NIS transgene has been successfully transferred selectively into extra-thyroidal tumor cells or cells in the tumor environment using various gene delivery systems [78]. An advanced endogenous PDAC mouse model study indicated genetically engineered mesenchymal stem cells (MSC) as NIS gene delivery vehicles demonstrate high stromal targeting of NIS by selective recruitment of NIS-MSCs after systemic application resulting in an impressive 131I therapeutic effect [78].

3.5. Nanotargeted Radionuclides

In the last three decades, there has been a rapid increase in the use of new nanomaterials and radionuclides to enhance cancer diagnosis and therapies [2,79]. Many organic and inorganic materials can be used as nanoparticles [58,80,81]. Nanoparticle (NP) delivery systems have enhanced imaging and therapeutic efficacy by targeting the delivery of radio-labeled drugs to the tumor site and reducing their toxic side effects [79,81,82]. The significant advantages of nanoparticles are that they can be prepared in sizes <100 nm. This increases the localization of the drugs and radionuclides and the permeability and retention (EPR) effect of passive targeting tumor cells and facilitates uptake by active targeting tumor cells [81,82]. The surface of nanomaterials is usually coated with polymers or ligands to improve biocompatibility and the selection of specific targets [80]. A nanomaterial’s final size and structure depend on the salt concentrations, surfactant additives, reactant concentrations, reaction temperatures, and solvent conditions used during synthesis [79,80]. Two mechanisms of nanoparticle delivery system for diagnostics and therapy to tumor sites are (i) specific passive targeting cells and (ii) specific active targeting cells [81,82].
Nanotargeted radionuclides have three main components, the nanoparticle core, the targeting biomolecule (which must be able to recognize a specific biological target), and the radionuclide [80]. Nanoparticles drug delivery systems can be made from polymers (polymeric nanoparticles, micelles, or dendrimers), lipids (liposomes), viruses (viral nanoparticles), organometallic compounds (nanotubes), inorganic nanoparticles (fullerenes, carbon nanotubes, quantum dots, or magnetic nanoparticles) [47,80,81,82]. The physical and chemical properties of nanoparticles play a critical role in determining particle–cell interactions, cellular trafficking mechanisms, biodistribution, pharmacokinetics, and optical properties [80]. Each nanoparticle type shows certain advantages and disadvantages that are inherent features of a particular material, such as solubility, thermal conductivity, ability to bind biomolecules or linkers, chemical stability, and capacity to incorporate and release compounds, as well as biocompatibility, toxicity, immunogenicity, and controlled drug release rate [47,80].
The targeting biomolecule must have a high affinity for the targeted epitopes. For ligands to bind effectively, each radionuclide can be conjugated directly on the nanoparticle surface, with or without a spacer, or can be attached to the nanoparticle during chemical synthesis. The spacer groups between the nanoparticle surface, the radionuclide, or the biomolecule can be a simple hydrocarbon chain, a peptide sequence, or a PEG linker [80,81,82]. In some cases, a bifunctional chelating group (BFC), such as 1, 4, 7, 10-tetraazadodecane-DOTA, must be conjugated to the nanoparticle, and then a radioactive metal needs to be attached. This requires modification of nanoparticles before radiolabeling [80].
There are many passive and active targeted nanoparticle therapies being developed. Most development is still at the in vitro or animal study stage. The most significant development is of 131I labeled nanoparticles for targeted therapy of different tumor types, according to the targeting strategy of the prepared NPs in which 131I is incorporated. The targeting strategy of these NPs depends on either passive or active targeting. 131I labeled NPs (silver or polymeric) shows 131I accumulation in different tumor types [79]. 131I labeled NPs targeting integrin have been studied. This protein is essential in regulating angiogenesis processes and tumor progression. Radionuclide labeled arginine–glycine–aspartate (RGD) can specifically target tumor integrin receptors [79]. Other β-particles such as 188Re, Holmium-166 (166Ho), 90Y, and gold-198 (198Au)-NPs have also been investigated for tumor-targeted radiotherapy [79,82]. 88Re-liposome has been shown to have a therapeutic effect in various animal models and translational clinical research [83]. 166Ho nanoparticles have also been prepared and studied in radionuclide tumor therapy for skin cancer and ovarian cancer metastases [80,82]. Liposome labeled with 90Y has been investigated for colon and melanoma tumors in animal models [79]. Gum arabic, functionalized peptide and protein, coated 198Au NP have been shown to be potential prostate cancer therapeutic agents in an animal model [79].
Nanoparticles labeled with α-particle emitters have been synthesized to enhance therapeutic efficiency with minimum danger to healthy tissues. 211At has been studied as a prospective NP’s alpha particle emitter, but the main disadvantage of 211At for NTR is low in vivo stability [79]. The sodium form of A-type nano-zeolites targeting peptides has been labeled with 223Ra, showing a cytotoxic effect on glioma cells [79]. A preliminary study reported that 225Ac-Au@TADOTAGA administrated intratumoral delayed tumor growth in glioma xenografts, and it is the first reported study using 225Ac-labeled gold nanoparticles [84]. However, 225Ac use remains challenging for insufficient retention of its daughter’s products due to the α recoil effect observed upon release of an α particle [84]. So, the use of α particles still has challenges related to incorporation into useful targeting vectors, such as in vivo stability, the weakness of α emitter–biomolecule bond, organ toxicity of inappropriate leakage of radionuclides from the bioconjugate, and uncoupling (or trans-chelated) than distribution to off-target areas [47].

4. Challenges in Radiopharmaceutical Therapy

In the last three decades, there has been a growing interest in radioiodine, a beta and gamma emitter, as new RPs are introduced for therapy and imaging (theranostics) for specific target tumor cells. However, one must be aware of issues related to the crossfire effect and toxicity of ß particles. The high LET and short range of α particles enable effective and rapid cancer therapies but are hindered by short half-lives and rarely emit gamma radiation for imaging [9,15,55]. Combining α and β particle emitters may reduce some of these obstacles [56,64]. Some issues with RPs are related to tumor-targeting uptake, biocompatibility, side effects, nonspecific uptake and distribution, and the radiation exposure effect in healthy tissues.
The development of intelligent drug delivery agents such as peptides, small molecules, mAb, and mAb fragments, and especially nanoparticle cores offer the promise of better diagnostic and therapeutic options [55,81]. However, the heterogeneity of RP uptake by tumor cells is challenging when using radiolabeled antibodies. The larger size of whole antibodies may limit penetration into the tumor tissue and crossfire effects, which occur when radiation interacts with cells away from the site of actual binding of the antibody agent [64].
Radiopharmaceuticals provide effective cancer treatment, particularly when other standard therapeutic approaches have failed. However, even after more than four decades of clinical investigation, RPs have still not become a standard part of cancer management therapy, which is peculiar, especially in light that other “targeted therapy” have clinical trial failure rates of 97% and is more popular than RPs that [7,8,85]. Furthermore, changing the fear of the public perception of radioactivity and the perceived complexity of the treatment are challenges in developing and applying RPs for therapy and imaging.

5. Conclusions and Future Direction

Radiopharmaceutical therapy can be a safe and effective targeted approach to treating many types of cancer. RPT has shown high efficacy with minimal toxicity compared to other systemic cancer treatment options. Different radioligands can be chosen to uniquely target molecular receptors or intracellular components, making them suitable for personal patient-tailored therapy in modern cancer therapy management. Further research is still needed regarding specific targets, radioligand stability in vivo, toxic effects, crossfire, dosimetry, and bond stability with daughter nuclides, particularly for alpha emitters.
However, new particle drug delivery systems continue to enhance targeted therapy efficacy and safety, including the use of nanoparticles. The number of successful studies exploring new drug delivery agents’ different delivery systems of radionuclide particles will probably increase the effectiveness and range of applicants. With a growing positive track record, public understanding and perception of the safety and success of RPT may improve.
If this occurs, then RPT will be adopted as an increasingly mainstream cancer therapy approach and the investment needed to resolve issues of radionuclides supply. In the coming decades, RPT may provide an increasing variety of rapid, personalized, practical, effective, and affordable treatments that offer new hope to cancer patients.

Author Contributions

Conceptualization, A.E.; writing—draft preparation, S.S., A.A. and W.A.; writing—review, A.E. and S.S.; editing A.E.; funding acquisition, S.S. All authors have read and agreed to the published version of the manuscript.

Funding

Article publication fund from Fatima College of Health Sciences, the United Arab Emirates.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

Thanks to Fatima College of Health Sciences, the United Arab Emirates, for fund of the article publication processing and Fay Farley for English editing assistance.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Goldsmith, S.J. Targeted Radionuclide Therapy: A Historical and Personal Review. Semin. Nucl. Med. 2020, 50, 87–97. [Google Scholar] [CrossRef] [PubMed]
  2. Yeong, C.H.; Cheng, M.H.; Ng, K.H. Therapeutic Radionuclides in Nuclear Medicine: Current and Future Prospects. J. Zhejiang Univ. Sci. B 2014, 15, 845–863. [Google Scholar] [CrossRef] [PubMed]
  3. Ercan, M.T.; Caglar, M. Therapeutic Radiopharmaceuticals. In Current Pharmaceutical Design; Bentham Science Publishers: Sharjah, United Arab Emirates, 2000; Volume 6, pp. 1085–1121. [Google Scholar]
  4. Gabriel, M. Radionuclide therapy beyond radioiodine. Wien. Med. Wochenschr. 2012, 162, 430–439. [Google Scholar] [CrossRef] [PubMed]
  5. Hillegonds, D.J.; Franklin, S.; Shelton, D.K.; Vijayakumar, S.; Vijayakumar, V. The management of painful bone metastases with an emphasis on radionuclide therapy. J. Natl. Med. Assoc. 2007, 99, 785–794. [Google Scholar] [PubMed]
  6. Asadian, S.; Mirzaei, H.; Kalantari, B.A.; Davarpanah, M.R.; Mohamadi, M.; Shpichka, A.; Nasehi, L.; Es, H.A.; Timashev, P.; Najimi, M.; et al. β-radiating radionuclides in cancer treatment, novel insight into promising approach. Pharmacol. Res. 2020, 160, 105070. [Google Scholar] [CrossRef] [PubMed]
  7. Sgouros, G.; Bodei, L.; McDevitt, M.R.; Nedrow, J.R. Radiopharmaceutical therapy in cancer: Clinical advances and challenges. Nat. Rev. Drug Discov. 2020, 19, 589–608. [Google Scholar] [CrossRef] [PubMed]
  8. Elliyanti, A. Radiopharmaceuticals in Modern Cancer Therapy. In Radiopharmaceutical Current Research for Better Diagnosis, Therapy, Environmental and Pharmaceutical Applications, 1st ed.; Badria, F.A., Ed.; Intechopen: London, UK, 2021. [Google Scholar] [CrossRef]
  9. Kramer-Marek, G.; Capala, J. The role of nuclear medicine in modern therapy of cancer. Tumor Biol. 2012, 33, 629–640. [Google Scholar] [CrossRef] [PubMed]
  10. Herrmann, K.; Schwaiger, M.; Lewis, J.S.; Solomon, S.B.; McNeil, B.J.; Baumann, M.; Gambhir, S.S.; Hricak, H.; Weissleder, R. Radiotheranostics: A roadmap for future development. Lancet Oncol. 2020, 21, e146–e156. [Google Scholar] [CrossRef]
  11. Kumar, C.; Shetake, N.; Desai, S.; Kumar, A.; Samuel, G.; Pandey, B.N. Relevance of radiobiological concepts in radionuclide therapy of cancer. Int. J. Radiat. Biol. 2016, 92, 173–186. [Google Scholar] [CrossRef]
  12. Wulbrand, C.; Seidl, C.; Gaertner, F.C.; Bruchertseifer, F.; Morgenstern, A.; Essler, M.; Senekowitsch-Schmidtke, R. Alpha-particle emitting 213Bi-anti- EGFR immunoconjugates eradicate tumor cells independent of oxygenation. PLoS ONE 2013, 8, e64730. [Google Scholar] [CrossRef]
  13. Calais, P.J.; Turner, J.H. Outpatient 131I-rituximab radoimmunotherapy for non-Hodgkin lymphoma: A study in safety. Clin. Nucl. Med. 2012, 37, 732–737. [Google Scholar] [CrossRef] [PubMed]
  14. Qaim, S.M. Therapeutic radionuclides and nuclear data. Radiochim. Acta 2001, 89, 297–304. [Google Scholar] [CrossRef]
  15. 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] [PubMed]
  16. Baskar, R.; Dai, J.; Wenlong, N.; Yeo, R.; Yeoh, K.W. Biological response of cancer cells to radiation treatment. Front. Mol. Biosci. 2014, 1, 24. [Google Scholar] [CrossRef]
  17. Komal, S.; Nadeem, S.; Faheem, Z.; Raza, A.; Sarwer, K.; Umer, H.; Roohi, S.; Naqvi, S.A.R. Localization Mechanisms of Radiopharmaceuticals. In Medical Isotopes, 1st ed.; Naqvi, S.A.R., Imran, M.B., Eds.; Intechopen: London, UK, 2021. [Google Scholar] [CrossRef]
  18. Elliyanti, A. An introduction to nuclear medicine in oncological molecular imaging. In Proceedings of the AIP Conference Proceedings, Padang, Indonesia, 10 December 2019. [Google Scholar] [CrossRef]
  19. Arslan, N.; Emi, M.; Alagöz, E.; Üstünsöz, B.; Oysul, K.; Arpacı, F.; Uğurel, Ş.; Beyzadeoğlu, M.; Ozgüven, M.A. Selective intraarterial radionuclide therapy with Yttrium-90 (Y-90) microspheres for hepatic neuroendocrine metastases: Initial experience at a single center. Vojnosanit. Pregl. 2011, 68, 341–348. [Google Scholar] [CrossRef]
  20. Kucuk, O.N.; Soydal, C.; Lacin, S.; Ozkan, E.; Bilgic, S. Selective intraarterial radionuclide therapy with yttrium-90 (Y-90) microspheres for unresectable primary and metastatic liver tumors. World J. Surg. Oncol. 2011, 9, 86. [Google Scholar] [CrossRef]
  21. Houle, S.; Yip, T.K.; Shepherd, F.A.; Rotstein, L.E.; Sniderman, K.W.; Theis, E.; Cawthorn, R.H.; Richmond-Cox, K. Hepatocellular carcinoma: Pilot trial of treatment with Y-90 microspheres. Radiology 1989, 172, 857–860. [Google Scholar] [CrossRef]
  22. Thamboo, T.; Tan, K.B.; Wang, S.C.; Salto-Tellez, M. Extrahepatic embolisation of Y-90 microspheres from selective internal radiation therapy (SIRT) of the liver. Pathology 2003, 35, 351–353. [Google Scholar]
  23. Widel, M.; Przybyszewski, W.M.; Cieslar-Pobuda, A.; Saenko, Y.V.; Rzeszowska-Wolny, J. Bystander normal human fibroblasts reduce damage response in radiation targeted cancer cells through intercellular ROS level modulation. Mutat. Res. 2012, 731, 117–124. [Google Scholar] [CrossRef]
  24. Ku, A.; Facca, V.J.; Cai, Z.; Reilly, R.M. Auger electrons for cancer therapy—A review. EJNMMI Radiopharm. Chem. 2019, 4, 27. [Google Scholar] [CrossRef]
  25. Elliyanti, A. Molecular Radiobiology and Radionuclides Therapy Concepts. In The Evolutionof Radionanotargeting towards Clinical Precission Oncology: A Festschrift in Honor of Kalevi Kairemo; Jekunen, A., Ed.; Bentham Science: Sharjah, United Arab Emirates, 2022; pp. 395–408. [Google Scholar] [CrossRef]
  26. 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]
  27. Elliyanti, A. Radioiodine for Graves’ Disease Therapy. In Graves’ Diseas, 1st ed.; Gensure, R., Ed.; Intechopen: London, UK, 2021. [Google Scholar] [CrossRef]
  28. Slonimsky, E.; Tulchinsky, M. Radiotheragnostics Paradigm for Radioactive Iodine (Iodide) Management of Differentiated Thyroid Cancer. Curr. Pharm. Des. 2020, 26, 3812. [Google Scholar] [CrossRef] [PubMed]
  29. Luster, M.; Pfestroff, A.; Hänscheid, H.; Verburg, F.A. Radioiodine Therapy. Semin. Nucl. Med. 2017, 47, 126–134. [Google Scholar] [CrossRef]
  30. Kendi, A.T.; Moncayo, V.M.; Nye, J.A.; Galt, J.R.; Halkar, R.; Schuster, D.M. Radionuclide therapies in molecular imaging and precision medicine. PET Clin. 2017, 12, 93–103. [Google Scholar] [CrossRef] [PubMed]
  31. Koziorowski, J.; Ballinger, J. Theragnostic radionuclides: A clinical perspective. Q. J. Nucl. Med. Mol. Imaging 2021, 65, 306–314. [Google Scholar] [CrossRef] [PubMed]
  32. Kassis, I.A.; Adelstein, S.J. Radiobiologic principles of radionuclide therapy. J. Nucl. Med. 2005, 46, 4S–12S. [Google Scholar] [PubMed]
  33. Gholami, Y.H.; Maschmeyer, R.; Kuncic, Z. Radio-enhancement effects by radiolabeled nanoparticles. Sci. Rep. 2019, 9, 14346. [Google Scholar] [CrossRef]
  34. Pouget, J.P.; Navarro-Teulon, I.; Bardiès, M.; Chouin, N.; Cartron, G.; Pèlegrin, A.; Azria, D. Clinical Radioimmunotherapy—The role of radiobiology. Nat. Rev. Clin. Oncol. 2011, 8, 720–734. [Google Scholar] [CrossRef]
  35. Jia, Z.; Wang, W. Yttrium-90 radioembolization for unresectable metastatic neuroendocrine liver tumor: A systematic review. Eur. J. Radiol. 2018, 100, 23–29. [Google Scholar] [CrossRef]
  36. Filippi, L.; Schillaci, O.; Cianni, R.; Bagni, O. Yttrium-90 resin microspheres and their use in the treatment of intrahepatic cholangiocarcinoma. Future Oncol. 2018, 14, 809–818. [Google Scholar] [CrossRef]
  37. Oei, A.L.; Verheijen, R.H.; Seiden, M.V.; Benigno, B.B.; Lopes, A.D.B.; Soper, J.T.; Epenetos, A.A.; Massuger, L.F. Decreased intraperitoneal disease recurrence in epithelial ovarian cancer patients receiving intraperitoneal consolidation treatment with yttrium-90-labeled murine HMFG1 without improvement in overall survival. Int. J. Cancer 2007, 120, 2710–2714. [Google Scholar] [CrossRef] [PubMed]
  38. Waldmann, T.; White, J.; Carrasquillo, J.; Reynolds, J.; Paik, C.; Gansow, O.; Brechbiel, M.; Jaffe, E.; Fleisher, T.; Goldman, C. Radioimmunotherapy
of interleukin-2R alpha-expressing adult T-cell leukemia with yttrium-90-labeled anti-Tac. Blood 1995, 86, 4063–4075. [Google Scholar] [CrossRef] [PubMed]
  39. Nisa, L.; Savelli, G.; Giubbini, R. Yttrium-90 DOTATOC therapy in GEP-NET and other SST2 expressing tumors: A selected review. Ann. Nucl. Med. 2011, 25, 75–85. [Google Scholar] [CrossRef] [PubMed]
  40. Kang, L.; Li, C.; Rosenkrans, Z.T.; Huo, N.; Chen, Z.; Ehlerding, E.B.; Huo, Y.; Ferreira, C.A.; Barnhart, T.E.; Engle, J.W.; et al. CD38-Targeted Theranostics of Lymphoma with 89Zr/177 Lu-Labeled Daratumumab. Adv. Sci. 2021, 8, 2001879. [Google Scholar] [CrossRef]
  41. Da Silva, T.N.; van Velthuysen, M.L.F.; van Eijck, C.H.J.; Teunissen, J.J.; Hofland, J. Successful neoadjuvant peptide receptor radionuclide therapy for an inoperable pancreatic neuroendocrine tumour. Endocrinol. Diabetes Metab. Case Rep. 2018, 11, 18-0015. [Google Scholar] [CrossRef]
  42. Sartor, O. Overview of samarium Sm 153 lexidronam in the treatment of painful metastatic bone disease. Rev. Urol. 2004, 6, S3–S12. [Google Scholar]
  43. Sgouros, G. Alpha-particles for targeted therapy. Adv. Drug Deliv. Rev. 2008, 60, 1402–1406. [Google Scholar] [CrossRef]
  44. Manafi-Farid, R.; Masoumi, F.; Divband, G.; Saidi, B.; Ataeinia, B.; Hertel, F.; Schweighofer-Zwink, G.; Morgenroth, A.; Beheshti, M. Targeted Palliative Radionuclide Therapy for Metastatic Bone Pain. J. Clin. Med. 2020, 9, 2622. [Google Scholar] [CrossRef]
  45. Guerra Liberal, F.D.C.; 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]
  46. Jurcic, J.G.; Levy, M.; Park, J.; Ravandi, F.; Perl, A.; Pagel, J.; Smith, B.D.; Orozco, J.; Estey, E.; Kantarjian, H.; et al. Trial in progress: A phase I/II study of lintuzumab-Ac225 in older patients with untreated acute myeloid leukemia. Clin. Lymphoma Myeloma Leuk. 2017, 17, S277. [Google Scholar] [CrossRef]
  47. Kleynhans, J.; Sathekge, M.; Ebenhan, T. Obstacles and Recommendations for Clinical Translation of Nanoparticle System-Based Targeted Alpha-Particle Therapy. Materials 2021, 14, 4784. [Google Scholar] [CrossRef] [PubMed]
  48. Goyal, J.; Antonarakis, E.S. Bone-targeting radiopharma ceuticals for the treatment of prostate cancer with bone metastases. Cancer Lett. 2012, 323, 135. [Google Scholar] [CrossRef] [PubMed]
  49. Filippi, L.; Chiaravalloti, A.; Schillaci, O.; Cianni, R.; Bagni, O. Theranostic approaches in nuclear medicine: Current status and future prospects. Expert Rev. Med. Devices 2020, 17, 331–343. [Google Scholar] [CrossRef] [PubMed]
  50. Bertolet, A.; Ramos-Méndez, J.; Paganetti, H.; Schuemann, J. The relation between microdosimetry and induction of direct damage to DNA by alpha particles. Phys. Med. Biol. 2021, 66, 155016. [Google Scholar] [CrossRef] [PubMed]
  51. Liberini, V.; Huellner, M.W.; Grimaldi, S.; Finessi, M.; Thuillier, P. The Challenge of Evaluating Response to Peptide Receptor Radionuclide Therapy in Gastroenteropancreatic Neuroendocrine Tumors: The Present and the Future. Diagnostics 2020, 10, 1083. [Google Scholar] [CrossRef]
  52. McDevitt, M.R.; Sgouros, G.; Sofou, S. Targeted and nontargeted α-particle therapies. Annu. Rev. Biomed. Eng. 2018, 20, 73–93. [Google Scholar] [CrossRef]
  53. Gustafsson-Lutz, A.; Bäck, T.; Aneheim, E.; Hultborn, R.; Palm, S.; Jacobsson, L.; Morgenstern, A.; Bruchertseifer, F.; Albertsson, P.; Lindegren, S. Therapeutic efficacy of α-radioimmunotherapy with different activity levels of the 213Bi-labeled monoclonal antibody MX35 in an ovarian cancer model. EJNMMI Res. 2017, 7, 38. [Google Scholar] [CrossRef]
  54. Ahenkorah, S.; Cassells, I.; Deroose, C.; Cardinaels, T.; Burgoyne, A.; Bormans, G.; Ooms, M.; Cleeren, F. Bismuth-213 for Targeted Radionuclide Therapy: From Atom to Bedside. Pharmaceutics 2021, 3, 599. [Google Scholar] [CrossRef]
  55. Silindir-Gunay, M.; Karpuz, M.; Ozer, A.Y. Targeted alpha therapy and Nanocarrier approach. Cancer Biother. Radiopharm. 2020, 35, 446–458. [Google Scholar] [CrossRef]
  56. Rosar, F.; Krause, J.; Bartholomä, M.; Maus, S.; Stemler, T.; Hierlmeier, I.; Linxweiler, J.; Ezziddin, S.; Khreish, F. Efficacy and safety of [225Ac] Ac-PSMA-617 augmented [177Lu] Lu-PSMA-617 Radioligand therapy in patients with highly advanced mCRPC with poor prognosis. Pharmaceutics 2021, 13, 722. [Google Scholar] [CrossRef]
  57. Reissig, F.; Wunderlich, G.; Runge, R.; Freudenberg, R.; Lühr, A.; Kotzerke, J. The effect of hypoxia on the induction of strand breaks in plasmid DNA by alpha-, beta- and Auger electron-emitters 223Ra, 188Re, 99mTc and DNA-binding 99mTc-labeled pyrene. Nucl. Med. Biol. 2020, 80–81, 65–70. [Google Scholar] [CrossRef] [PubMed]
  58. Stéen, E.J.L.; Edem, P.E.; Nørregaard, K.; Jørgensen, J.T.; Shalgunov, V.; Kjaer, A.; Herth, M.M. Pretargeting in nuclear imaging and radionuclide therapy: Improving efficacy of theranostics and nanomedicines. Biomaterials 2018, 179, 209–245. [Google Scholar] [CrossRef] [PubMed]
  59. Kennel, S.J.; Mirzadeh, S.; Eckelman, W.C.; Waldmann, T.A.; Garmestani, K.; Yordanov, A.T.; Stabin, M.G.; Brechbiel, M.W. Vascular-targeted radioimmunotherapy with the alpha- particle emitter 211At. Radiat. Res. 2002, 157, 633–641. [Google Scholar] [CrossRef]
  60. Persson, L. The Auger electron effect in radiation dosimetry. Health Phys. 1994, 67, 471–476. [Google Scholar] [CrossRef] [PubMed]
  61. Widel, M. Radionuclides in radiation-induced bystander effect; may it share in radionuclide therapy? Neoplasma 2017, 64, 641–654. [Google Scholar] [CrossRef] [PubMed]
  62. Kirsch, D.G.; Diehn, M.; Kesarwala, A.; Maity, A.; Morgan, M.A.; Schwarz, J.K.; Bristow, R.; DeMaria, S.; Eke, I.; Griffin, R.J.; et al. The Future of Radiobiology. J. Natl. Cancer Inst. 2018, 110, 329–340. [Google Scholar] [CrossRef]
  63. Paillas, S.; Ladjohounlou, R.; Lozza, C.; Pichard, A.; Boudousq, V.; Jarlier, M.; Sevestre, S.; Le Blay, M.; Deshayes, E.; Sosabowski, J.; et al. Localized irradiation of cell membrane by auger electrons is cytotoxic through oxidative stress-mediated nontargeted effects. Antioxid. Redox Signal. 2016, 25, 467–484. [Google Scholar] [CrossRef]
  64. Pandit-Taskar, N. Targeted Radioimmunotherapy and Theranostics with Alpha Emitters. J. Med. Imaging Radiat. Sci 2019, 50, S41–S44. [Google Scholar] [CrossRef]
  65. White, J.M.; Escorcia, F.E.; Viola, N.T. Perspectives on metals-based radioimmunotherapy (RIT): Moving forward. Theranostics 2021, 11, 6293–6314. [Google Scholar] [CrossRef]
  66. Fendler, W.P.; Rahbar, K.; Herrmann, K.; Kratochwil, C.; Eiber, M. 177Lu-PSMA Radioligand Therapy for Prostate Cancer. J. Nucl. Med. 2017, 58, 1196–1200. [Google Scholar] [CrossRef]
  67. Nevedomskaya, E.; Baumgart, S.J.; Haendler, B. Recent advances in prostate Cancer treatment and drug discovery. Int. J. Mol. Sci. 2018, 19, 1359. [Google Scholar] [CrossRef] [PubMed]
  68. Lunger, L.; Tauber, R.; Feuerecker, B.; Gschwend, J.E.; Eiber, M. Narrative review: Prostate-specific membrane antigen-radioligand therapy in metastatic castration-resistant prostate cancer. Transl. Androl. Urol. 2021, 10, 3963–3971. [Google Scholar] [CrossRef] [PubMed]
  69. Kairemo, K.; Joensuu, T. Lu-177-PSMA treatment for metastatic prostate cancer: Case examples of major responses. Clin. Transl Imaging 2018, 6, 223–237. [Google Scholar] [CrossRef]
  70. Sathekge, M.; Bruchertseifer, F.; Knoesen, O.; Reyneke, F.; Lawal, I.; Lengana, T.; Davis, C.; Mahapane, J.; Corbett, C.; Vorster, M.; et al. 225Ac-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] [PubMed]
  71. Khreish, F.; Ebert, N.; Ries, M.; Maus, S.; Rosar, F.; Bohnenberger, H.; Stemler, T.; Saar, M.; Bartholomä, M.; Ezziddin, S. 225Ac-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]
  72. Kratochwil, C.; Haberkorn, U.; Giesel, F.L. 225Ac-PSMA-617 for Therapy of Prostate Cancer. Semin. Nucl. Med. 2020, 50, 133–140. [Google Scholar] [CrossRef]
  73. Basu, S.; Parghane, R.V.; Chakrabarty, S. Peptide Receptor Radionuclide Therapy of Neuroendocrine Tumors. Semin. Nucl. Med. 2020, 50, 447–464. [Google Scholar] [CrossRef]
  74. Kunikowska, J.; Królicki, L. Targeted α-Emitter Therapy of Neuroendocrine Tumors. Semin. Nucl. Med. 2020, 50, 171–176. [Google Scholar] [CrossRef]
  75. Elliyanti, A.; Rustam, R.; Tofrizal, T.; Yenita, Y.; Susanto, Y.D.B. Evaluating the Natrium iodide Symporter expressions in thyroid Tumors. Open Access Maced. J. Med. Sci. 2021, 9, 18–23. [Google Scholar] [CrossRef]
  76. Elliyanti, A.; Rusnita, D.; Afriani, N.; Susanto, Y.D.B.; Susilo, V.Y.; Setiyowati, S.; Harahap, W.A. Analysis natrium iodide symporter expression in breast cancer subtypes for radioiodine therapy response. Nucl. Med. Mol. Imaging 2020, 54, 35–42. [Google Scholar] [CrossRef]
  77. Liu, J.; Liu, Y.; Lin, Y.; Liang, J. Radioactive Iodine-Refractory Differentiated Thyroid Cancer and Redifferentiation Therapy. Endocrinol. Metab. 2019, 34, 215–225. [Google Scholar] [CrossRef]
  78. Schug, C.; Gupta, A.; Urnauer, S.; Steiger, K.; Cheung, P.F.Y.; Neander, C.; Savvatakis, K.; Schmohl, K.A.; Trajkovic-Arsic, M.; Schwenk, N. A Novel Approach for Image-Guided 131I Therapy of Pancreatic Ductal Adenocarcinoma Using Mesenchymal Stem Cell-Mediated NIS Gene Delivery. Mol. Cancer Res. 2019, 17, 310–320. [Google Scholar] [CrossRef]
  79. Bayoumi, N.A.; El-Kolaly, M.T. Utilization of nanotechnology in targeted radionuclide cancer therapy: Monotherapy, combined therapy and radiosensitization. Radiochim. Acta 2021, 109, 459–475. [Google Scholar] [CrossRef]
  80. Mirshojaei, S.F.; Ahmadi, A.; Morales-Avila, E.; Ortiz-Reynoso, M.; Reyes-Perez, H. Radiolabelled nanoparticles: Novel classification of radiopharmaceuticals for molecular imaging of cancer. J. Drug Target. 2016, 24, 91–101. [Google Scholar] [CrossRef]
  81. Ting, G.; Chang, C.H.; Wang, H.E.; Lee, T.W. Nanotargeted radionuclides for cancer nuclear imaging and internal radiotherapy. J. Biomed. Biotechnol. 2010, 2010, 953537. [Google Scholar] [CrossRef]
  82. Farzin, L.; Sheibani, S.; Moassesi, M.E.; Shamsipur, M. An overview of nanoscale radionuclides and radiolabeled nanomaterials commonly used for nuclear molecular imaging and therapeutic functions. J. Biomed. Mater. Res. A 2019, 107, 251–285. [Google Scholar] [CrossRef]
  83. Chang, C.H.; Chang, M.C.; Chang, Y.J.; Chen, L.C.; Lee, T.W.; Ting, G. Translating Research for the Radiotheranostics of Nanotargeted 188Re-Liposome. Int. J. Mol. Sci. 2021, 22, 3868. [Google Scholar] [CrossRef]
  84. Salvanou, E.-A.; Stellas, D.; Tsoukalas, C.; Mavroidi, B.; Paravatou-Petsotas, M.; Kalogeropoulos, N.; Xanthopoulos, S.; Denat, F.; Laurent, G.; Bazzi, R.; et al. A Proof-of-Concept Study on the Therapeutic Potential of Au Nanoparticles Radiolabeled with the Alpha-Emitter Actinium-225. Pharmaceutics 2020, 12, 188. [Google Scholar] [CrossRef]
  85. Wong, C.H.; Siah, K.W.; Lo, A.W. Estimation of clinical trial success rates and related parameters. Biostatistics 2018, 20, 273–286. [Google Scholar] [CrossRef]
Figure 1. The cell’s radiation distribution by RPT (a) and external radiotherapy (b). Radiopharmaceuticals are administrated intravenously to be delivered to a target tumor. The targeted tumor cell absorbs a dose of radiation which exponentially decreases over time. The tumor mass’s periphery cells will receive absorbed and crossfire doses from other target cells (a). Radiation beams are directed at tumor tissue in external radiotherapy and can also affect healthy cells (b).
Figure 1. The cell’s radiation distribution by RPT (a) and external radiotherapy (b). Radiopharmaceuticals are administrated intravenously to be delivered to a target tumor. The targeted tumor cell absorbs a dose of radiation which exponentially decreases over time. The tumor mass’s periphery cells will receive absorbed and crossfire doses from other target cells (a). Radiation beams are directed at tumor tissue in external radiotherapy and can also affect healthy cells (b).
Molecules 27 05231 g001
Table 1. Characteristics of radionuclides used in radiotherapy.
Table 1. Characteristics of radionuclides used in radiotherapy.
RadionuclidesEmittingPhysical
Half-Life
Mean
Eα/β- (MeV)
Primary Eα/β- (MeV) (%)Mean Range in Soft Tissue (mm)IndicationReferences
MaxMinMean
131Iβ8.02 d0.606 MeV0.069 MeV 0.356 MeV0.3645 MeV (81%)0.4 mmHyperthyroid, thyroid cancer, Radioimmunotherapy (RIT) for NHL and neuroblastoma, pheochromocytoma, carcinoid, medullary thyroid cancer[2,3,6,8,22,24]
32Pβ14.26 d1.71 MeV0.695 MeV1.015 MeV-2.6 mmPolycythemia vera, keloid, cystic craniopharyngioma,[2,3,23]
89Srβ50.53 d1.491 MeV0.583 MeV0.908 MeV0.91 MeV (0.01%)2.4 mmBone pain palliation[2,3,6,8,23]
90Yβ64.10 d2.284 MeV0.935 MeV1.349 MeV(0.01%)3.6 mmLiver metastasis, hepatocellular carcinoma, RIT for NHL, neuroendocrine tumor[2,3,6,8,22,23]
153Smβ46.50 h0.8082 MeV--0.1032 MeV (29.8%)0.7 mmBone pain palliation, synovitis[2,3,6,8]
169Erβ9.4 d0.35 MeV--0.084 MeV (0.16%)0.3 mmSynovitis[2,3]
177Luβ6.73 d0.497 MeV0.047 MeV0.208 MeV0.208 MeV (11%)0.28 mmSynovitis and RIT for various cancer[2,6,8,22,23,24]
186Reβ3.72 d1.077 MeV0.308 MeV0. 769 MeV0.137 MeV (9.4%)1.2 mmBone pain palliation, arthritis[2,6,8,23]
188Reβ17 h2.12 MeV0.528 MeV1.592 MeV0.155 MeV
(15%)
2.1 mmBone pain palliation, RIT for various cancer, rheumatoid arthritis[2,3,8,22,23]
223Raα11.44 d5.9792 MeV-6.59 MeV0.154 MeV (5.59%)0.054 mmBone pain palliation[2,5,13]
211Atα7.2 h--6.79 MeV(5.87%)0.057 mmRIT leukemia, brain tumor, RLT prostate cancer[2,3,23,25]
213Biα46 mins--8.32MeV(26%)0.078 mmRIT leukemia, brain tumor[3,22,23,25]
225Acα10 d--0.218MeV(11.4%)0.05–0.08 mmRadioligand (RLT) prostate cancer[2,8,24]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Salih, S.; Alkatheeri, A.; Alomaim, W.; Elliyanti, A. Radiopharmaceutical Treatments for Cancer Therapy, Radionuclides Characteristics, Applications, and Challenges. Molecules 2022, 27, 5231. https://doi.org/10.3390/molecules27165231

AMA Style

Salih S, Alkatheeri A, Alomaim W, Elliyanti A. Radiopharmaceutical Treatments for Cancer Therapy, Radionuclides Characteristics, Applications, and Challenges. Molecules. 2022; 27(16):5231. https://doi.org/10.3390/molecules27165231

Chicago/Turabian Style

Salih, Suliman, Ajnas Alkatheeri, Wijdan Alomaim, and Aisyah Elliyanti. 2022. "Radiopharmaceutical Treatments for Cancer Therapy, Radionuclides Characteristics, Applications, and Challenges" Molecules 27, no. 16: 5231. https://doi.org/10.3390/molecules27165231

Article Metrics

Back to TopTop