Open Access

Current clinical application of lutetium‑177 in solid tumors (Review)

  • Authors:
    • Tingting Niu
    • Mi Fan
    • Binwei Lin
    • Feng Gao
    • Bangxian Tan
    • Xiaobo Du
  • View Affiliations

  • Published online on: March 26, 2024     https://doi.org/10.3892/etm.2024.12514
  • Article Number: 225
  • Copyright: © Niu et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Radionuclide‑based therapy represents a novel treatment regimen for tumors. Among these therapies, lutetium‑177 (177Lu) has gained significant attention due to its stability and safety, as well as its ability to emit both γ and β rays, allowing for both imaging with single photon emission computed tomography and tumor treatment. As a result, 177Lu can be used for both diagnosis and treatment for diseases such as prostatic and gastric cancer. Therefore, based on the available data, the present review provides a brief overview of the clinical applications of 177Lu‑targeted radionuclide therapy in metastatic prostate cancer, neuroendocrine tumors and other types of solid tumors, and highlights the current therapeutic effect, reduction in damage to normal tissues and future research directions, including the development of new nuclides and the application of more nuclides in different tumors. In the future, such treatments could be used in more tumors.

1. Introduction

The number of total novel cancer cases globally is expected to reach 28.4 million by 2040, which represents a 47% increase compared with 2020(1). The increase is expected to be higher in developing countries (64-95%) compared with developed countries (32-56%) based on demographic changes (1). On the basis of existing chemotherapy, radiotherapy, immunization, targeting and other therapies, novel diagnosis and treatment methods require investigation. Due to its advantages of integrated diagnosis and treatment, targeted radionuclide therapy has become a new strategy to further improve the prognosis of cancer.

Targeted radionuclide therapy (TRT) involves labeling radionuclides on biomolecules with a specific binding ability, which allows the biomolecule to bind to specific tumor targets, and thus the radionuclide is concentrated at the tumor site (2). The α or β particles emitted by radionuclides can then irradiate the tumor tissues specifically and ionize biological effects (low linear energy transfer), resulting in cell senescence and death within the irradiation range (3). This achieves therapeutic results with minimal effects on the surrounding normal tissues. By combining the advantages of targeted therapy and brachytherapy, TRT provides novel methods for the diagnosis and treatment of tumors. Based on current data, the present review briefly introduced the use of different 177Lu drugs in treating several tumors, as well as the issues presented by the use of these drugs and future research directions.

2. Basic characteristics of radionuclide 177Lu

177Lu belongs to the lanthanide metal group (4) and can emit β-rays with a maximum energy of 0.49 MeV and with a half-life of 6.7 days (5). β-rays are mostly used for treatment of diseases such as prostate cancer, and the energy released by the β-particle rays of 177Lu has an average range of 670 µm in soft tissue, significantly reducing the damage caused by radionuclides to nearby healthy cells. In addition, 177Lu can emit two characteristic γ-rays (208 and 113 keV), which can be used as a signal source for single-photon emission computed tomography or positron emission tomography (PET) (6). These are mostly used for imaging and dosimetric analysis; for example, 177Lu-prostate-specific membrane antigen (PSMA)-image and treat (I&T) PET/computed tomography (CT) is used for pre- and post-treatment imaging in patients with metastatic prostate cancer (6,7).

3. Application of 177Lu in prostate cancer

The latest global cancer data results released by the World Health Organization shows that the number of new cases of prostate cancer in 2020 was 1.41 million, ranking it second most common amongst all new tumors and the number of prostate cancer-associated mortalities in men was 380,000, making it the fifth most fatal cancer in men (1). In recent years, the prevalence of metastatic castration-resistant prostate cancer (mCRPC) has been increasing, and its prognosis is generally poor (8). The expression of PSMA in prostate cancer is higher compared with that in other tissues, including prostatic epithelial cells, the small intestine and salivary glands (9), and is 1,000 times higher compared with the lowest expression level in the kidney and small intestine (10). Therefore, precise targeted therapy with PSMA has emerged as a novel therapeutic method, resulting in a decline in prostate specific antigen (PSA) levels and an increase in overall survival (11,12).

Prostate cancer and 177Lu-PSMA-I&T

1,4,7,10-tetraazacyclododececane-1-(glutaric acid)-4,7,10-triaceticacid (DOTAGA) is a small PSMA inhibitor molecule labeled with 177Lu known as 177Lu-PSMA-I&T. In 2015, internal radiation therapy using 177Lu-PSMA-I&T was reported to be effective and safe in two patients with metastatic prostate cancer, with no detectable side effects (13). In a study by Barna et al (14), 177Lu-PSMA-I&T was used to treat mCRPC with a mean injection activity of 7,416±218 MBq. Follow-up imaging using Ga-PSMA PET/CT was used to determine individual tumor molecular volume. The volume of 63 individual tumors in bone, lymph nodes and liver tissues were observed to decrease by 32.3, 84.7 and 72.9% on average, respectively (14). In a trial of 49 patients with mCRPC who received at least three cycles of 177Lu-PSMA-I&T (6.0 GBq), no grade III/IV adverse events were reported, indicating low nephrotoxicity or hematotoxicity, according to the Common Terminology Criteria for Adverse Events (CTCAE v5.0) (15).

Another prospective single-arm trial included 40 patients diagnosed with mCRPC who received 86 cycles of 177Lu-PSMA-I&T at a dose of 3.70-14.43 GBq per cycle from December 2019 to September 2021, with a median follow-up of 8 months (16). The findings revealed that six patients (15%) developed mild reversible dry mouth, and 28 patients (70%) developed grade 1-4 bone marrow dysfunction (anemia, thrombocytopenia and leukopenia) during follow-up (toxicities were assessed following the CTCAE v5.0). Serum PSA is the most significant marker for evaluating therapeutic biochemical response. PSA levels were obtained every 4 weeks before and after treatment. A reduction of ≥30% from baseline is considered a partial response, a >25% increase in PSA above baseline is defined as disease progression, and PSA levels with changes between <-30% and <+25% are considered disease stabilization. PSA levels were assessed after treatment and accompanied by partial remission in 25 patients (62.5%), stable disease in five patients (12.5%) and progression in ten patients (25%). Trials have shown that 177Lu-PSMA-I&T can achieve significant PSA reduction and tumor remission in patients with mCRPC (16,17).

To evaluate the predictive effect after treatment, a trial involving 301 patients demonstrated that the extent of bone metastases and their changes were potential markers for predicting treatment outcomes in patients with mCRPC (18). A previous single-arm phase I study of 177Lu-PSMA-I&T neoadjuvant therapy in high-risk prostate cancer before radical prostatectomy included 14 patients with high-critical-limited-stage prostate cancer, defined as PSA >20 ng/ml, biopsy Gleason score ≥8, or clinical T stage ≥3a, who tested positive for PSMA between December 2019 and December 2021(19). Furthermore, two or three doses of 177Lu-PSMA-I&T (7.4 GBq) were administered at 2-week intervals, and surgery (including lymph node dissection) was performed 4 weeks after the final dose. The main adverse reactions of this treatment were the incidence of perioperative complications and organ functional toxicity, including bleeding, infection, pneumonia and pulmonary embolism. After two doses, the PSA level was reduced by 17%, and after three doses it was reduced by 34%. No severe intraoperative complications were observed in 13 patients (one patient did not undergo surgery due to heart problems), while four (30%) patients developed postoperative complications (including pneumonia, pulmonary embolism, urinary leakage and urinary tract infection) (19).

Preliminary studies have demonstrated that neoadjuvant therapy with 177Lu-PSMA-I&T followed by surgery is safe; however, further data should be obtained from long-term follow-up (19). These findings suggest that patients treated with 177Lu-PSMA-I&T show decreased PSA levels and fewer serious adverse events, contributing to tumor remission (16-18).

Prostate cancer and 177Lu-PSMA-617

177Lu-PSMA-617 is a small molecule inhibitor labeled with 177Lu using 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid (DOTA) as a chelating agent (20,21). In March, 2022, the U.S. Food and Drug Administration (FDA) approved the administration of 177Lu-PSMA-617 for the treatment of adult patients with mCRPC, making it the first targeted radioligand therapy to be approved by the FDA (22).

In a meta-analysis, Kim and Kim (23) included 10 studies involving 455 patients with mCRPC. The analysis showed that 177Lu-PSMA-617 therapy resulted in a combined PSA reduction rate of 68.00% [95% confidence interval (CI), 63.55-72.22%] and with PSA levels reduced by >50% in 34.45% of patients (95% CI, 30.14-38.97%). In phase II, single-arm, single-center trials by Violet et al (24) and Hofman et al (25), 177Lu-PSMA-617 radionuclide therapy had a high response rate, low toxicity and decreased pain in patients with metastatic castration-tolerant prostate cancer who progressed following conventional therapy (including taxane-based chemotherapy and second-generation antiandrogen therapy). The FDA authorized 177Lu-PSMA-617 as a prostate cancer treatment in March 2022 based on a phase III experiment conducted by Sartor et al (26).

An international, open-label, Phase III clinical trial involving 84 centers (52 centers in North America and 32 in Europe) was conducted (26). Patients were randomly assigned in a 2:1 ratio to receive either 177Lu-PSMA-617 (injection of 7.4 GBq once every 6 weeks, lasting 4-6 cycles) plus standard protocol-approved treatment (177Lu-PSMA-617 group) or standard treatment only (e.g., abiraterone and enzalutamide) (control group). Among the 831 patients with metastatic castration-tolerant prostate cancer treated with at least one androgen receptor pathway inhibitor and one or two taxane receptor inhibitors, 581 were included in the analysis set. 177Lu-PSMA-617 + standard therapy significantly extended imaging-based progression-free survival (PFS) time compared with standard therapy (median, 8.7 vs. 3.4 months; hazard ratio for progression or death, 0.40; 99.2% CI, 0.29-0.57; P<0.001) and OS (median, 15.3 vs. 11.3 months; hazard ratio for death, 0.62; 95% CI, 0.52-0.74; P<0.001). The most frequent adverse reactions in the 177Lu-PSMA-617 group were fatigue, dry mouth and nausea; however, these events were usually grade 1 or 2 (Response Evaluation Criteria in Solid Tumors, version 1.1) (27) and had little impact on the quality of life.

177Lu-PSMA-I&T vs. 177Lu-PSMA-617

The distinction between the two PSMA-targeting medications, 177Lu-PSMA-617 and 177Lu-PSMA-I&T, lies in the chelating agents they use. A study with 110 patients with mCRPC from two locations (University Hospital Würzburg and University Hospital Bonn), including 55 who received 177Lu-PSMA-I&T and 55 who received 177Lu-PSMA-617, revealed no significant differences in the harmful effects of the two at a dose of roughly 6.0 GBq per 8 weeks. Survival with 177Lu-PSMA-I&T and 177Lu-PSMA-617 was comparable with a median OS time of 12.0 vs. 13.0 months, respectively, with no serious grade III/IV toxicity (28). In another study that assessed the safety, biological distribution and dosiology in 138 patients, 51 individuals were administered 177Lu-PSMA-I&T at 6.1±1.0 GBq and 87 patients received 177Lu-PSMA 617 at 6.5±1.1 GBq (one injection). The mean dose of 177Lu-PSMA-617 was higher compared with 177Lu-PSMA-I&T (0.04 vs. 0.03 Gy/GBq), and the systemic half-life of 177Lu-PSMA-I&T (35 h) was shorter compared with that of 177lu-PSMA-617 (42 h). Of all the healthy organs, the lacrimal glands had the highest mean absorbed tumor dose of 177Lu-PSMA-I&T and 177Lu-PSMA-617 (5.8 vs. 5.9 Gy/GBq), but patients tolerated the therapy without any acute side effects (29). A study that further evaluated the difference in efficacy between 177Lu-PSMA-617 plus standard therapy (hormone therapy, bisphosphonates and radiotherapy) and 177Lu-PSMA-617 alone showed that combined therapy extended the duration of pain exacerbation compared with 177Lu-PSMA-617 alone (30). It is hoped that future studies can include the combined use of nuclides with other drugs (such as targeted drugs and chemotherapy drugs), and it is hoped that new progress and breakthroughs can be made in the treatment of mCRPC through combination therapy.

Prostate cancer and 177Lu-J591

J591 is a deimmunizing monoclonal antibody that binds to PSMA and has internalized properties (internalization of a putative ligand) (31,32). Participants received 75 mg/m docetaxel once every 21 days for 2 cycles, with two progressive increments of ‘Lu-1591’ at cycle 3 (1.48 GBq/m2, up to 2.96 GBq/m2). Docetaxel was administered in cycle 3 (half of the Lu-J591 dose 2 to 3 days before docetaxel and the other half of the dose 2 weeks after docetaxel), with the fourth cycle of docetaxel given 6-9 weeks after the third cycle. No dose-limiting toxicity was observed at any of the tested dose levels. Grade 4 neutropenia without fever was found in eight patients (53.5%). Subsequently, two patients (13.3%) developed thrombocytopenia, and no grade ≥3 non-hematological toxicity was observed. A PSA decrease of >50% after treatment with 177Lu-J591 was seen in 11 patients (73.3%). The results suggest that a single 177Lu-J591 fractionated course in combination with docetaxel is a viable option for patients with mCRPC (33). A review of the various phase I and II trials of 177Lu-J591 in metastatic prostate cancer showed improved OS, and in almost all cases, myelosuppression is tolerable and reversible (31). While 177Lu-J591 may be considered a potential treatment option, the effectiveness of its combination with docetaxel still requires additional research and clinical trials to establish its efficacy and safety.

Prostate cancer and [177Lu] Ludotadipep

Ludotadipep is a new PSMA inhibitor tagged with 177Lu, which contains a 4-iodophenyl butanoic group that can bind to albumin and prolong circulation time and boost absorption in tumors. A prospective study from Korea evaluated the efficacy of 177Lu Ludotadipep in patients with 18F-PSMA-PET/CT-positive mCRPC. From November 2020 to March 2022, a total of 30 patients were enrolled for single dose of 177Lu Ludotadipep radiopharmaceutical therapy (34). Patients were divided into five groups (n=6) and given an increasing dose of 1.9, 2.8, 3.7, 4.6 and 5.6 GBq for each group. In this study, 29 patients who received 177Lu Ludotadipep (one patient found to deviate from the inclusion criteria after enrollment), 36 treatment-emergent adverse events (58.6%) and four adverse drug reactions (10.3%) were observed. Overall 16 (66.7%) of the 24 participants with complete 12-week follow-up data showed a reduction in PSA levels, and nine (37.5%) of those subjects showed a PSA decline of ≥50%. At the 12th week after receiving a single dosage of 177Lu Ludotadipep, five of the 24 patients (20.8%) displayed disease progression (a ≥25% increase in PSA levels from the baseline), suggesting that 177Lu Ludotadipep may be a promising new treatment for mCRPC (34).

4. Application of 177Lu in neuroendocrine tumors (NETs)

NETs are rare, heterogeneous tumors that originate from cells of the diffuse endocrine system (35). Approximately two-thirds of NETs occur in the gastrointestinal and pancreatic systems, including the stomach, small intestine, colon, appendix, rectum and pancreas. The most common NET subtype is the gastrointestinal pancreatic NET (GEP-NET) (36). An analysis of United States cancer data showed that the incidence of gastrointestinal neuroendocrine tumors has continuously increased each decade from 1977 to 2016(37). Currently, surgery is the primary therapeutic method for GEP-NETs because most GEP-NETs are inert. In addition, severe cases are treated with radiochemotherapy, molecularly targeted medicine.

Most pancreatic and GEP-NETs express somatostatin receptor (SSTR)2 and 5, these receptors can be chelated to a β-emitting radioisotope 177Lu for therapy. The new approach for treating advanced NETs is peptide receptor radionuclide therapy (38).

Somatostatin is a peptide with a potent and broad antisecretory action. SSTRs which belong to the G protein-coupled receptor family, are widely distributed in various tissues of the body and are classified into five subtypes (SSTR1-5) (39). SSTR2 is associated with gastrointestinal neuroendocrine system (40).

NETs and 177Lu-1, 4, 7, 10-tetraazacyclododecane-1, 4, 7, 10-tetraacetic acid-D-phenylalanine 1-tyrosine 3-threonine 8-octreotide (177Lu-DOTA-TATE) also known as DOTA-octreotate

177Lu-DOTA-TATE is a labeled precursor of DOTA-modified octreotide. Octreotide is a somatostatin analogue that is used to control NET progression (41). A 177Lu-labeled SSTR agonist has been used to treat somatostatin receptor positive GEP-NETs (42). The FDA approved the first radiopharmaceutical for the treatment of GET on January 26, 2018.

An open, randomized, phase III clinical trial (NETTER-1) enrolled 231 patients with locally advanced or metastatic, well-differentiated, somatostatin receptor-positive midgut neuroendocrine tumors. Patients were randomly assigned (1:1) to receive intravenous 177Lu-DOTA-TATE 7.4 GBq + intramuscular long-acting octreotide (30 mg every 8 weeks) (experimental group) or high-dose long-acting octreotide (60 mg every 4 weeks) (control group). According to the NETTER-1 trial, which was reported on July 24, 2016, the estimated rate of PFS at 20 months was 65.2% (95% CI, 50.0-76.8%) in the experimental group and 10.8% (95% CI, 3.5-23.0%) in the control group. The response rate (complete plus partial) in the experimental group was 18%, whereas it was only 3% in the control group (P<0.001). Preliminary findings show that using 177Lu-DOTA-TATE markedly increases the PFS (43). In a 2021 follow-up analysis the median OS for the experimental group was 48.0 months (95% CI, 37.4-55.2 months), while the control group OS was 36.3 months (95% CI, 25.9-51.7 months). The OS of the experimental group did not improve significantly compared with that of the control group, and there was no statistically significant difference in OS. However, in absolute terms, there was a difference of 11.7 months in the median OS between 177Lu-DOTA-TATE and octreotide alone groups. In the experimental group, 111 patients (3%) experienced serious adverse events related to treatment that were grade 3 or worse. During the 100 months of follow-up, 2% of patients developed myelodysplastic syndrome, yet no other experimental group patients experienced renal impairment that was grade 3 or worse. Although the OS was not statistically significant, the results showed that the 11.7 monthly difference in the median OS between 177Lu-DOTA-TATE treatment and high-dose long-acting octreotide treatment alone may be considered clinically relevant (44).

A study compared PFS and OS in 177Lu-DOTA-TATE and patients with advanced and unresectable gastrointestinal neuroendocrine tumors treated with everolimus and sunitinib (45). In comparison to everolimus, sunitinib and best supportive care, the results of the primary analysis demonstrated that 177Lu-DOTA-TATE may be a more effective therapy choice. In a study evaluating the efficacy and safety of 177Lu-DOTA-TATE in 30 patients with NET and extensive bone metastases, radiological evaluation at the end of treatment showed partial response in five patients, stable disease in 20 patients, and progressive radiological disease in three patients. Clinical progress was observed in another two patients (46).

NETs and 177Lu-DOTA-JR11 (177Lu-OPS201)

177Lu-OPS201 is a novel somatostatin antagonist with high affinity for SSTR2(47). In situ testing in a SSTR2-positive neuroendocrine model in mice reveals that 177Lu-OPS201 causes a greater decrease in living tumor tissue, a significant delay in tumor growth and increased toxicity compared with 177Lu-DOTATOC (an SSTR agonist that primarily targets SSTR2). Likewise, the use of the 177Lu-OPS201 has been shown to increase tumor uptake of agonist 177Lu-DOTA-TATE in vitro (47); however, it has not been validated at clinical trial stage. According to a recent in vitro study, 177Lu-OPS201 has a high affinity, and 177Lu-OPS201 has at least four times more receptor-binding sites compared with 177Lu-DOTA-TATE. In conclusion, 177Lu-OPS201 has demonstrated faster binding, slower dissociation and a longer cell retention period compared with 177Lu-DOTA-TATE (48).

5. Clinical application of 177Lu in other diseases

Fibroblast activating protein (FAP) overexpression in cancer and 177Lu-FAP-2286

FAP is expressed in a number of malignancies. 177Lu-FAP-2286 is a FAP-binding cyclic peptide consisting of seven amino acids, of which two cysteine residues pass through an aromatic partial ring that is linked to the DOTA chelating agent (49). FAP-2287 (a murine surrogate for FAP-2286) is well targeted for 177Lu targeted radionuclide therapy, rapidly accumulates in tissues and persists in tumors for a long time (50).

A trial has been performed that involved 11 patients with advanced pancreatic, breast, rectal and ovarian cancers receiving 177Lu-FAP-2286. The results showed that the dosage of administration of 177Lu-FAP-2286 (5.8±2.0 GBq; range, 2.4-9.9 GBq) was well tolerated, and no adverse or clinically detectable pharmacological effects were found or reported in any patient. The systemic effective dose was 0.07±0.02 Gy/GBq (range, 0.04-0.1 Gy/GBq). The mean absorbed doses in kidney and red bone marrow were 1.0±0.6 Gy/GBq (range, 0.4-2.0 Gy/GBq) and 0.05±0.02 Gy/GBq (range, 0.03-0.09 Gy/GBq), respectively. No grade 4 adverse events were observed; however, grade 3 adverse reactions occurred in three patients, including one pancytopenia, one leukopenia and one pain response. 177Lu-FAP-2286 is widely used in adenocarcinoma and is well tolerated with few side effects (51). These results indicate that 177Lu-FAP-2286 has encouraging clinical data and deserves further exploration.

Meningioma and 177Lu-DOTA-TATE

At present, there is no evidence-based systemic treatment for patients with progressive meningioma who are unable to receive surgery or external radiotherapy. External radiation therapy was given to 15 patients with meningioma, all of whom had received radiotherapy and 14 of whom had surgery and then received 177Lu-DOTA-TATE (7.5-29.6 GBq). 177Lu-DOTA-TATE was administered with a maximum activity of 7.4 GBq per cycle with a maximum of 4 cycles. In this cohort, the interval between the cycles was a median of 9 weeks (range, 6-14 weeks). Subsequently, six patients (40%) were stable following treatment. The median PFS of the whole cohort was 7.8 months, with a 6-month PFS rate of 60%. The median OS was 13.6 months, with a 12-month OS rate of 60%. Prior to therapy, their average monthly tumor growth rate (TGR) was 4.6% for the surface and 14.8% for the volume. The surface and volume were scanned and determined by magnetic resonance imaging. The TGR decreased to 3.1% in surface (P=0.016) and 5.0% in volume (P=0.013) per month after treatment. The results suggest that 177Lu-DOTA-TATE can control tumor growth (52).

Metastatic salivary gland cancer and 177Lu-PSMA-617

PSMA is expressed on tumor cells or in the tumor neovasculature in salivary gland carcinoma, particularly in certain subtypes such as salivary gland and salivary duct carcinomas (53,54). There have been cases of salivary gland malignancies that show significant uptake in 68Ga-PSMA-11 PET-CT (55-57). Therefore, in view of the encouraging results of patients treated for CRPC, the potential use of 177Lu-PSMA-617 to treat metastatic salivary gland cancer has been assessed.

In a retrospective study by Klein Nulent et al (58), six patients were treated with 177Lu-PSMA-617. This cohort included four adenoid cystic carcinomas, one adenocarcinoma (not otherwise specified) and one acinic cell carcinoma. A total of four patients reported instant reduction of tumor-related symptoms; the most common improvement was a reduction in pain, followed by a reduction in fatigue. Two individuals demonstrated a radiological response, indicating either stable disease or a partial remission. All treatment-related clinical and hematological adverse events were graded using the CTCAE standard version 5.0. This study demonstrated that palliative 177Lu-PSMA-617 for salivary gland cancer is safe and generally well tolerated.

A 56-year-old man with progressive metastatic salivary cancer who received treatment every 6 weeks and was first evaluated after four courses (cumulative activity of 24.3 GBq 177Lu-PSSMA-617) showed stable disease on imaging according to Positron Emission tomography Response Criteria In Solid Tumor (PERCIST) criteria (59).

Metastatic thyroid cancer and 177Lu-DOTA-TATE/177Lu-PSMA-617I

A number of studies have shown that PSMA is expressed in thyroid cancer, and significant uptake can be observed with 68Ga-PSMA-11 PET-CT (60-63). Therefore, 177Lu-PSMA-617 has been used to treat metastatic thyroid cancer. Assadi and Ahmadzadehfar (64) treated a patient with progressing thyroid cancer and neck and lung metastases with 177Lu-PSMA. The patient previously received 25.9 GBq 131Iodine and sorafenib for 6 months, and radioligand therapy targeting the somatostatin receptor using 177Lu-DOTA-TATE. As the patient exhibited persistent disease progression and severe breathing difficulties, the patient was then treated with 7.4 GBq 177Lu-PSMA-617. With whole-body SPE-CT imaging, there was a higher level of whole-body imaging with 177Lu-PSMA-617 compared with receiving 177Lu-DOTA-TATE, suggesting that treatment with 177Lu-PSMA-617 is potentially more effective compared with 177Lu-DOTA-TATE for the treatment of this patient. However, the patient died unexpectedly of cardiac arrest two weeks after treatment.

In an additional study, two patients received 177Lu-PSMA-617 therapy for thyroid carcinoma. One patient showed disease progression on imaging 1 month later, the other patient showed partial, temporary response of lung and liver metastases (65). Side effects were not mentioned in either of these two case reports of thyroid carcinoma.

Advanced renal cell cancer and 177Lu-cG250

Previous studies have shown that carbonic anhydrase IX is almost universally expressed (>90%) in metastatic clear cell renal cell carcinoma (ccRCC), and its expression in healthy tissues has been extensively evaluated, but is limited to gastrointestinal mucosa and gastrointestinal associated structures, with much lower expression levels compared with those in ccRCC (66,67). Thus, 177Lu-girentuximab (cG250) was used to treat ccRCC. Diagnostic Indium111-cG250 imaging confirmed cG250 accumulation in 23 patients with progressive ccRCC metastases (68). In this phase I study, patients received a high-activity dose of 177Lu-cG250. At 3 months after the initial treatment, 74% of patients had stable illness (evaluated by PERCIST), and one patient had a partial response that persisted for 9 months. Mean growth of target tumor lesions was reduced from 40.4% in the 3 months before treatment to 5.5% 3 months after the first treatment cycle. There were no significant non-hematological adverse effects noted (68).

Additionally, in a phase II study, 14 patients with ccRCC were treated with 177Lu-cG250, of which eight patients had stable disease and one had partial regression. The treatment was generally well tolerated (69).

Metastatic bone tumors and 177Lu-EDTMP

A study by Elboga et al (70) found that 177Lu-ethylenediamine tetramethyle nephosphonic acid (EDTMP) effectively relieved pain caused by bone metastases in patients with breast or prostate cancer. Of the 75 patients treated with 177Lu-EDTMP, 59 responded positively, while 16 did not. The pain score was analyzed, and patients who responded had markedly lower pain scores after each radiopharmaceutical treatment. A meta-analysis involving 172 patients revealed that 177Lu-EDTMP had a significant effect on relieving bone pain, suggesting that this agent could be a good choice when other pain-relieving radiopharmaceuticals are not available (71).

Currently, it is proposed that a 177Lu-EDTMP rapid kit be developed based on its success in palliative treatment, which, if successfully implemented, could greatly reduce pain in terminal cancer patients (72).

6. Present problems and future research directions

At present, the main problem with 177Lu TRT is the limited variety of drugs used in clinical practice, and the need for further improvement in clinical efficacy. 177Lu TRT is a promising treatment method for solid tumors, but currently only 177Lu-DOTA-TATE and 177Lu-PSMA-617 have been approved treatment for NETs and prostate cancer, respectively. Moreover, the response rate of 177Lu TRT for mCRPC was from 32.3 to 68%, and 177Lu-DOTA-TATE for neuroendocrine tumors was only 18%.

Future research on 177Lu TRT should focus on expanding the application of 177Lu-DOTA-TATE and 177Lu-PSMA-617 to other diseases that express PSMA or SSTR. There are currently ongoing clinical trials assessing the activity of 177Lu-DOTA-TATE and 177Lu-PSMA-617 on SSTR and PSMA-expressing tumors as listed in Table I. This includes breast cancer, metastatic nasopharyngeal carcinoma, neuroblastoma, glioblastoma and glioma, NETs of the lung and thymus for SSTR-expressing tumors and upper metastatic gastric cancer for PSMA-expressing tumors.

Table I

Recruiting clinical trials of 177Lu targeted radionuclide therapy for tumors expressing SSTR and PSMA (except neuroendocrine tumor and prostate cancer) in the past 5 years.

Table I

Recruiting clinical trials of 177Lu targeted radionuclide therapy for tumors expressing SSTR and PSMA (except neuroendocrine tumor and prostate cancer) in the past 5 years.

NCTStarting datePhaseTargetVectorIsotopeApplicationAdministration RoutePrimary outcome
NCT045290442022-09-01IISSTRDotatateLu-177Recurrent breast cancerI.VORR
NCT051984792023-01IISSTR DOTA0-Tyr3-OctreotateLu-177Metastatic NPCI.VPFS
NCT049038992021-05-19IISSTRDOTATATELu-177NeuroblastomaI.VAssess the response of treatment.
NCT056440802023-03-28N/APSMAPSMA I&TLu-177/Ga-68GliomaI.VAEs/efficacy
NCT059183022023-07IIISSTREdotreotideLu-177Neuroendocrine tumorsI.VPFS
NCT051097282022-05-10ISSTRDOTA-TATELu-177GlioblastomaI.VDLTs
NCT039714612019-05-15IISSTRLutatheraLu-177MeningiomaI.VPFS
NCT052148202022-01-17IIPSMA 68Ga-PSMAGa-68 EndoradiotherapyI.VTumor uptake

[i] SSTR2, somatostatin receptor subtype 2; PSMA, prostate-specific membrane antigen; NPC, metastatic nasopharyngeal cancer; ORR, overall response rate; PFS, progression free survival; DLTs, number of participants with dose limiting toxicities; AEs, adverse events; NCT, national clinical trial; I.V, intravenous; PFS, progression-free survival; N/A, not applicable; I&T, image and treat.

Likewise, future research should focus on the development of more 177Lu TRT targets in solid tumors. In the past 5 years, the ongoing clinical trials of new targets for 177Lu TRT have included fibronectin ED-B, NY108, TLX591, LNC1004, 3PRGD2 and GD2 (Table II).

Table II

New targets of clinical trials of 177Lu targeted radionuclide therapy for solid tumors in the past 5 years.

Table II

New targets of clinical trials of 177Lu targeted radionuclide therapy for solid tumors in the past 5 years.

NCTStarting datePhaseTargetVectorIsotopeApplicationAdministration routePrimary outcome
NCT056238912022-12IED-BB5-IgG4Lu-177Solid TumorsI.VTissue distribution/Dosimetry/AEs
NCT058153942023-03-03INY108DOTALu-177Prostate cancerI.VTissue distribution/AEs
NCT047868472022-01-30IPSMATLX591Lu-177MPCI.VAEs
NCT057236402023-05-10IFAPLNC1004Lu-177Solid tumorI.V DLTs/safety/MDT
NCT054108212022-06-15IFAPDOTA-EB-FAPILu-177Thyroid cancerI.V Safety/tolerability/MDT
NCT056035592023-01-01IPSMAP17-087/088Lu-177mCRPCI.VDosimetry/AEs
NCT050130862021-10-01IIntegrin αvβ3AB-3PRGD2Lu-177NSCLCI.VStandardized uptake value
NCT051302552022-11-17IGD2DOTALu-177Solid tumorsI.VDLTs
NCT049973172021-04-21ISSTR2satoreotideLu-177MeningiomasI.VTherapeutic index/safety
NCT038727782019-07-24I/IIGRPRNeoBLu-177Solid tumorI.VDLTs/MDT
NCT046659472020-12-18IABM-5GDOTALu-177Pancreatic cancerI.VDLTs/RP2D
NCT057061292023-03-14I/IIDPI-4452DOTALu-177/Ga-68Solid tumorsI.VTEAEs/DLTs/ORR
NCT058681742023-05-23ICAIXPeposertibLu-177Solid tumorsI.VDLTs/safety
NCT054321932022-07-13IFAPPNT6555Lu-177Solid tumorsI.VAEs
NCT047111352022-08-31IISSTRLutetiumLu-177GEP-NETsI.VAbsorbed radiation doses/AEs
NCT046475262021-02-25IIIPSMAPNT2002Lu-177mCRPCI.VrPFS
NCT044691272023-01-30I/IIαvβ3DOTALu-177Breast cancerI.VSafety and tolerability
NCT055332422023-06I6A10DOTALu-177GlioblastomaI.VMDT/safety
NCT051786932022-04-25ISSTRASTX727Lu-177NETsI.VTo determine whether pre-treatment with ASTX727 results in re-expression of SSTR2

[i] PSMA, prostate-specific membrane antigen; FAP, fibroblast activation protein; SSTR2, somatostatin receptor subtype 2; GRPR, gastrin-releasing peptide receptor; CAIX, carbonic anhydrase IX; MPC, metastatic prostate cancer; mCRPC, metastatic castration-resistant prostate cancer; NSCLC, non-small cell lung cancer; GEP-NETs, gastroenteropancreatic neuroendocrine tumors; I.V, intravenous; AEs, adverse events; DLTs, number of participants with dose limiting toxicities; MTD, maximum tolerated dose; RP2D, recommended phase 2 dose; TEAEs, treatment emergent adverse events; ORR, overall response rate; rPFS, radiographic progression-free survival; NCT, national clinical trial.

Lastly, the use of 177Lu TRT in combination with other treatments should be explored. For example, in a preclinical model (murine model), 177Lu-FAP-2287 enhanced anti-PD-1-mediated tumor growth inhibition by modulating the tumor microenvironment and increasing the recruitment of tumor-infiltrating CD8+ T cells (50). Another study showed that concurrent rather than sequential blockade of the PD-1/PD-L1 axis combined with 177Lu TRT improves OS and long-term tumor control (73). The study of the metabolism principle of nuclide in the body can further clarify the treatment method in the future to improve the survival period. A Phase I study of the 177Lu-DOTA0-Tyr3-Octreotate combination with nivolumab was well tolerated and showed signs of antitumor activity for patients with neuroendocrine tumors of the lung (74). In the past 5 years, the number of ongoing clinical trials of 177Lu TRT combined with other treatments for solid tumors has increased. A list of the current trials is shown in Table III. The results of these clinical trials will likely improve the current options for 177Lu TRT treatments.

Table III

Ongoing clinical trials of 177Lu targeted radionuclide therapy combined with other solid tumor treatments in the past 5 years.

Table III

Ongoing clinical trials of 177Lu targeted radionuclide therapy combined with other solid tumor treatments in the past 5 years.

NCTStarting datePhaseTargetCombination treatmentApplicationPrimary outcome
NCT053403742022-07-14I/IIPSMACabazitaxel + 177Lu-PSMA-617mCRPCDLTs/MTD/RP2D
NCT045256382020-06-29IISSTRNivolumab + 177Lu-DOTATATENENSORR
NCT057663712023-06-30IIPSMAPembrolizumab + 177Lu-PSMA-617CRPCrPFS
NCT038748842019-07-09IPSMAOlaparib + 177Lu-PSMA-617mCRPCDLTs/MTD/RP2D
NCT051502362022-04-29IIPSMAIpilimumab + Nivolumab + 177Lu-PSMA-617mCRPCPSA-PFS
NCT048869862021-06-30I/IIPSMA 225AC-J591+ 177Lu-PSMA-I&TmCRPCDLTs/MTD
NCT056138942023-06-26IbPSMACabozantinib + 177Lu-PSMA-617mCRPCDLTs
NCT052395332022-02-16IIAnti-carbonic anhydrase IX monoclonal antibodyNivolumab + 177Lu-girentuximabKidney cancerMTD/ORR
NCT058681742023-05-23ICAIXPeposertib + 177Lu-TLX250Solid tumorsDLT/safety
NCT047201572021-06-09IIIPSMASoC + 177Lu-PSMA-617mHSPCrPFS
NCT056637102023-06-30IISSTRCabozantinib + NivolumabRCCIncidence of PFS
NCT042618552020-10-08I/IISSTRAvelumab + 177Lu-DOTATATEMCCPFS
NCT053830792022-09-13I/IIPSMA223Ra + 177Lu-PSMA-I&TmCRPCDLTs/MTD/RP2D
NCT057241082023-03-15IISSTRTriapine + 177Lu-DOTATATENENSORR
NCT052491142022-12-28ISSTRCabozantinib + 177-Lu-DOTATATENENSMTD PFS
NCT056871232023-04-28ISSTRSunitinib Malate + 177Lu-DOTATATEPancreatic NENSAEs
NCT047509542021-06-04ISSTRPeposertib + 177Lu-DOTATATEPancreatic NENSRP2D
NCT056824432023-07-01IIPSMAONC-392 + 177Lu-PSMA-617mCRPCrPFS
NCT052479052022-03-16IISSTRCapecitabine + TemozolomidePancreatic NENSPFS
NCT040864852022-10-03I/IISSTROlaparib + 177Lu-DOTATATEGEP-NETMTD/ORR
NCT046147662022-09-30I/IISSTRLutathera + AzedraSPORE-3MTD/ORR

[i] PSMA, prostate-specific membrane antigen; CAIX, carbonic anhydrase IX; SSTR, somatostatin receptor subtype; SoC, standard of care; mCRPC, metastatic castration resistant prostate cancer; OPC, oligometastatic prostate cancer; NENS, neuroendocrine neoplasms; CRPC, castration resistant prostate cancer; mHSPC, metastatic hormone sensitive prostate cancer; RCC, renal cell carcinoma; MCC, Merkel cell carcinoma; GEP-NET, gastroenteropancreatico neuroendocrine tumor; SPORE-3, mid-gut neuroendocrine tumors; DLTs, number of participants with dose limiting toxicities; MTD, maximum tolerated dose; RP2D, recommended Phase 2 dose; PSA PFS, prostate-specific antigen progression free survival at 1 year; bPFS, biochemical progression free survival; ORR, overall response rate; rPFS, radiographic progression-free survival; AEs, adverse events.

7. Conclusions

177Lu TRT have been successfully applied in patients with NETs or metastatic prostate cancer, resulting in prolonged PFS, OS and improved quality of life with tolerable toxicities. However, 177Lu TRT are rarely used to treat other solid tumors and clinical efficacies need to be improved. The use of 177Lu-DOTA-TATE and 177Lu-PSMA-617 should be expanded to various diseases, and more new targets for 177Lu TRT should be researched and developed. Lastly, 177Lu TRT in combination with other treatments provide further treatment options for solid tumors.

Acknowledgements

Not applicable.

Funding

Funding: This work was financially supported by the NHC Key Laboratory of Nuclear Technology Medical Transformation, Mianyang Central Hospital (grant nos. 2022HYX001 and 2022HYX0015).

Availability of data and materials

Not applicable.

Authors' contributions

TN and MF drafted the manuscript. BL, FG and BT participated in the data review and collection for the study. XD conceived the study and reviewed and edited the manuscript. All authors read and approved the final version of the manuscript. Data authentication is not applicable.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A and Bray F: global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 71:209–249. 2021.PubMed/NCBI View Article : Google Scholar

2 

Al-Toubah T, Strosberg J, Hallanger-Johnson J and El-Haddad G: Targeted radionuclide therapy in endocrine-related cancers: Advances in the last decade. Front Endocrinol (Lausanne). 14(1187870)2023.PubMed/NCBI View Article : Google Scholar

3 

Salih S, Alkatheeri A, Alomaim W and Elliyanti A: Radiopharmaceutical treatments for cancer therapy, radionuclides characteristics, applications, and challenges. Molecules. 27(5231)2022.PubMed/NCBI View Article : Google Scholar

4 

Park UJ, Lee JS, Choi KH, Nam SS and Yu KH: Lu-177 preparation for radiotherapy application. Appl Radiat Isot. 115:8–12. 2016.PubMed/NCBI View Article : Google Scholar

5 

Zakaly HMH, Mostafa MYA, Deryabina D and Zhukovsky M: Comparative studies on the potential use of 177Lu-based radiopharmaceuticals for the palliative therapy of bone metastases. Int J Radiat Biol. 96:779–789. 2020.PubMed/NCBI View Article : Google Scholar

6 

Dash A, Pillai MR and Knapp FF Jr: Production of (177)Lu for targeted radionuclide therapy: Available options. Nucl Med Mol Imaging. 49:85–107. 2015.PubMed/NCBI View Article : Google Scholar

7 

Tan HY, Yeong CH, Wong YH, McKenzie M, Kasbollah A, Shah MNM and Perkins AC: Neutron-activated theranostic radionuclides for nuclear medicine. Nucl Med Biol. 90-91:55–68. 2020.PubMed/NCBI View Article : Google Scholar

8 

Cao J, Chen Y, Hu M and Zhang W: 177Lu-PSMA-RLT of metastatic castration-resistant prostate cancer: Limitations and improvements. Ann Nucl Med. 35:861–870. 2021.PubMed/NCBI View Article : Google Scholar

9 

Boegemann M, Schrader AJ and Rahbar K: 177Lu-PSMA therapy: Current evidence for use in the treatment of patients with metastatic prostate cancer. Urologe A. 56:1440–1444. 2017.PubMed/NCBI View Article : Google Scholar : (In German).

10 

Ghosh A and Heston WD: Tumor target prostate specific membrane antigen (PSMA) and its regulation in prostate cancer. J Cell Biochem. 91:528–539. 2004.PubMed/NCBI View Article : Google Scholar

11 

Fendler WP, Rahbar K, Herrmann K, Kratochwil C and Eiber M: 177Lu-PSMA radioligand therapy for prostate cancer. J Nucl Med. 58:1196–1200. 2017.PubMed/NCBI View Article : Google Scholar

12 

Sadaghiani MS, Sheikhbahaei S, Werner RA, Pienta KJ, Pomper MG, Gorin MA, Solnes LB and Rowe SP: 177 Lu-PSMA radioligand therapy effectiveness in metastatic castration-resistant prostate cancer: An updated systematic review and meta-analysis. Prostate. 82:826–835. 2022.PubMed/NCBI View Article : Google Scholar

13 

Weineisen M, Schottelius M, Simecek J, Baum RP, Yildiz A, Beykan S, Kulkarni HR, 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. 56:1169–1176. 2015.PubMed/NCBI View Article : Google Scholar

14 

Barna S, Haug AR, Hartenbach M, Rasul S, Grubmüller B, Kramer G and Blaickner M: Dose calculations and dose-effect relationships in 177Lu-PSMA I&T radionuclide therapy for metastatic castration-resistant prostate cancer. Clin Nucl Med. 45:661–667. 2020.PubMed/NCBI View Article : Google Scholar

15 

Hartrampf PE, Weinzierl FX, Serfling SE, Pomper MG, Rowe SP, Higuchi T, Seitz AK, Kübler H, Buck AK and Werner RA: Hematotoxicity and nephrotoxicity in prostate cancer patients undergoing radioligand therapy with [177Lu]Lu-PSMA I&T. Cancers (Basel). 14(647)2022.PubMed/NCBI View Article : Google Scholar

16 

Bu T, Zhang L, Yu F, Yao X, Wu W, Zhang P, Shi L, Zang S, Meng Q, Ni Y, et al: 177Lu-PSMA-I&T radioligand therapy for treating metastatic castration-resistant prostate cancer: A single-centre study in East Asians. Front Oncol. 12(835956)2022.PubMed/NCBI View Article : Google Scholar

17 

Emmett L, John N, Pathmanandavel S, Counter W, Ayers M, Sharma S, Agrawal S, Poole A, Hovey E, Pranavan G, et al: Patient outcomes following a response biomarker-guided approach to treatment using 177Lu-PSMA-I&T in men with metastatic castrate-resistant prostate cancer (Re-SPECT). Ther Adv Med Oncol. 15(17588359231156392)2023.PubMed/NCBI View Article : Google Scholar

18 

Karimzadeh A, Heck M, Tauber R, Knorr K, Haller B, D'Alessandria C, Weber WA, Eiber M and Rauscher I: 177Lu-PSMA-I&T for treatment of metastatic castration-resistant prostate cancer: Prognostic value of scintigraphic and clinical biomarkers. J Nucl Med. 64:402–409. 2023.PubMed/NCBI View Article : Google Scholar

19 

Golan S, Frumer M, Zohar Y, Rosenbaum E, Yakimov M, Kedar D, Margel D, Baniel J, Steinmetz AP, Groshar D, et al: Neoadjuvant 177Lu-PSMA-I&T radionuclide treatment in patients with high-risk prostate cancer before radical prostatectomy: A single-arm phase 1 trial. Eur Urol Oncol. 6:151–159. 2023.PubMed/NCBI View Article : Google Scholar

20 

Stasiuk GJ and Long NJ: The ubiquitous DOTA and its derivatives: The impact of 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid on biomedical imaging. Chem Commun (Camb). 49:2732–2746. 2013.PubMed/NCBI View Article : Google Scholar

21 

Keam SJ: Lutetium Lu 177 vipivotide tetraxetan: First approval. Mol Diagn Ther. 26:467–475. 2022.PubMed/NCBI View Article : Google Scholar

22 

Liu X, Fang GC, Lu H, Shi ZD, Chen ZS and Han CH: Lutetium Lu 177 vipivotide tetraxetan for prostate cancer. Drugs Today (Barc). 59:37–49. 2023.PubMed/NCBI View Article : Google Scholar

23 

Kim YJ and Kim YI: Therapeutic responses and survival effects of 177Lu-PSMA-617 radioligand therapy in metastatic castrate-resistant prostate cancer: A meta-analysis. Clin Nucl Med. 43:728–734. 2018.PubMed/NCBI View Article : Google Scholar

24 

Violet J, Sandhu S, Iravani A, Ferdinandus J, Thang SP, Kong G, Kumar AR, Akhurst T, Pattison DA, Beaulieu A, et al: Long-term follow-up and outcomes of retreatment in an expanded 50-patient single-center phase II prospective trial of 177Lu-PSMA-617 theranostics in metastatic castration-resistant prostate cancer. J Nucl Med. 61:857–865. 2020.PubMed/NCBI View Article : Google Scholar

25 

Hofman MS, Violet J, Hicks RJ and Sandhu S: [177Lu]-PSMA-617 radionuclide therapy in patients with metastatic castration-resistant prostate cancer-Author's reply. Lancet Oncol. 19(e373)2018.PubMed/NCBI View Article : Google Scholar

26 

Sartor O, de Bono J, Chi KN, Fizazi K, Herrmann K, Rahbar K, Tagawa ST, Nordquist LT, Vaishampayan N, El-Haddad G, et al: Lutetium-177-PSMA-617 for metastatic castration-resistant prostate cancer. N Engl J Med. 385:1091–1103. 2021.PubMed/NCBI View Article : Google Scholar

27 

Schwartz LH, Litière S, de Vries E, Ford R, Gwyther S, Mandrekar S, Shankar L, Bogaerts J, Chen A, Dancey J, et al: RECIST 1.1-Update and clarification: From the RECIST committee. Eur J Cancer. 62:132–137. 2016.PubMed/NCBI View Article : Google Scholar

28 

Hartrampf PE, Weinzierl FX, Buck AK, Rowe SP, Higuchi T, Seitz AK, Kübler H, Schirbel A, Essler M, Bundschuh RA and Werner RA: Matched-pair analysis of [177Lu]Lu-PSMA I&T and [177Lu]Lu-PSMA-617 in patients with metastatic castration-resistant prostate cancer. Eur J Nucl Med Mol Imaging. 49:3269–3276. 2022.PubMed/NCBI View Article : Google Scholar

29 

Schuchardt C, Zhang J, Kulkarni HR, Chen X, Müller D and Baum RP: Prostate-specific membrane antigen radioligand therapy using 177Lu-PSMA I&T and 177Lu-PSMA-617 in patients with metastatic castration-resistant prostate cancer: Comparison of safety, biodistribution, and dosimetry. J Nucl Med. 63:1199–1207. 2022.PubMed/NCBI View Article : Google Scholar

30 

Fizazi K, Herrmann K, Krause BJ, Rahbar K, Chi KN, Morris MJ, Sartor O, Tagawa ST, Kendi AT, Vogelzang N, et al: Health-related quality of life and pain outcomes with [177Lu]Lu-PSMA-617 plus standard of care versus standard of care in patients with metastatic castration-resistant prostate cancer (VISION): A multicentre, open-label, randomised, phase 3 trial. Lancet Oncol. 24:597–610. 2023.PubMed/NCBI View Article : Google Scholar

31 

Niaz MO, Sun M, Ramirez-Fort MK and Niaz MJ: Review of lutetium-177-labeled anti-prostate-specific membrane antigen monoclonal antibody j591 for the treatment of metastatic castration-resistant prostate cancer. Cureus. 12(e7107)2020.PubMed/NCBI View Article : Google Scholar

32 

Liu H, Rajasekaran AK, Moy P, Xia Y, Kim S, Navarro V, Rahmati R and Bander NH: Constitutive and antibody-induced internalization of prostate-specific membrane antigen. Cancer Res. 58:4055–4060. 1998.PubMed/NCBI

33 

Batra JS, Niaz MJ, Whang YE, Sheikh A, Thomas C, Christos P, Vallabhajosula S, Jhanwar YS, Molina AM, Nanus DM, et al: Phase I trial of docetaxel plus lutetium-177-labeled anti-prostate-specific membrane antigen monoclonal antibody J591 (177Lu-J591) for metastatic castration-resistant prostate cancer. Urol Oncol. 38:848.e9–848.e16. 2020.PubMed/NCBI View Article : Google Scholar

34 

Shin D, Ha S, O JH, Rhew SA, Yoon CE, Kwon HJ, Moon HW, Park YH, Park SY, Park C, et al: A single dose of novel PSMA-targeting radiopharmaceutical agent [177Lu]ludotadipep for patients with metastatic castration-resistant prostate cancer: Phase i clinical trial. Cancers (Basel). 14(6225)2022.PubMed/NCBI View Article : Google Scholar

35 

Jacoba IM and Weber HC: Biomarkers in gastroenteropancreatic neuroendocrine neoplasms. Curr Opin Endocrinol Diabetes Obes. 30:175–180. 2023.PubMed/NCBI View Article : Google Scholar

36 

Fernandes CJ, Leung G, Eads JR and Katona BW: Gastroenteropancreatic neuroendocrine tumors. Gastroenterol Clin North Am. 51:625–647. 2022.PubMed/NCBI View Article : Google Scholar

37 

Liu M, Wei L, Liu W, Chen S, Guan M, Zhang Y, Guo Z, Liu R and Xie P: Trends in incidence and survival in patients with gastrointestinal neuroendocrine tumors: A SEER database analysis, 1977-2016. Front Oncol. 13(1079575)2023.PubMed/NCBI View Article : Google Scholar

38 

Harris PE and Zhernosekov K: The evolution of PRRT for the treatment of neuroendocrine tumors; What comes next? Front Endocrinol (Lausanne). 13(941832)2022.PubMed/NCBI View Article : Google Scholar

39 

Theodoropoulou M and Stalla GK: Somatostatin receptors: From signaling to clinical practice. Front Neuroendocrinol. 34:228–252. 2013.PubMed/NCBI View Article : Google Scholar

40 

Ichikawa Y, Kobayashi N, Takano S, Kato I, Endo K and Inoue T: Neuroendocrine tumor theranostics. Cancer Sci. 113:1930–1938. 2022.PubMed/NCBI View Article : Google Scholar

41 

Pokuri VK, Fong MK and Iyer R: Octreotide and lanreotide in gastroenteropancreatic neuroendocrine tumors. Curr Oncol Rep. 18(7)2016.PubMed/NCBI View Article : Google Scholar

42 

Das S, Al-Toubah T, El-Haddad G and Strosberg J: 177Lu-DOTATATE for the treatment of gastroenteropancreatic neuroendocrine tumors. Expert Rev Gastroenterol Hepatol. 13:1023–1031. 2019.PubMed/NCBI View Article : Google Scholar

43 

Strosberg J, El-Haddad G, Wolin E, Hendifar A, Yao J, Chasen B, Mittra E, Kunz PL, Kulke MH, Jacene H, et al: Phase 3 trial of 177Lu-Dotatate for midgut neuroendocrine tumors. N Engl J Med. 376:125–135. 2017.PubMed/NCBI View Article : Google Scholar

44 

Strosberg JR, Caplin ME, Kunz PL, Ruszniewski PB, Bodei L, Hendifar A, Mittra E, Wolin EM, Yao JC, Pavel ME, et al: 177Lu-Dotatate plus long-acting octreotide versus high-dose long-acting octreotide in patients with midgut neuroendocrine tumours (NETTER-1): Final overall survival and long-term safety results from an open-label, randomised, controlled, phase 3 trial. Lancet Oncol. 22:1752–1763. 2021.PubMed/NCBI View Article : Google Scholar

45 

Khan MS, Stamp E, Sammon C, Brabander T, de Herder WW and Pavel ME: Matching-adjusted indirect treatment comparison of [177Lu]Lu-DOTA-TATE, everolimus and sunitinib in advanced, unresectable gastroenteropancreatic neuroendocrine tumours: Relative effectiveness of [177Lu]Lu-DOTA-TATE in gastroenteropancreatic neuroendocrine tumours. EJC Suppl. 16:5–13. 2021.PubMed/NCBI View Article : Google Scholar

46 

Alsadik S, Gnanasegaran G, Chen L, Quigley AM, Mandair D, Toumpanakis C, Caplin M and Navalkissoor S: Safety and efficacy of 177 Lu-DOTATATE in neuroendocrine tumor patients with extensive bone disease. Clin Nucl Med. 48:667–672. 2023.PubMed/NCBI View Article : Google Scholar

47 

Albrecht J, Exner S, Grötzinger C, Prasad S, Konietschke F, Beindorff N, Kühl AA, Prasad V, Brenner W and Koziolek EJ: Multimodal imaging of 2-cycle PRRT with 177Lu-DOTA-JR11 and 177Lu-DOTATOC in an orthotopic neuroendocrine xenograft tumor mouse model. J Nucl Med. 62:393–398. 2021.PubMed/NCBI View Article : Google Scholar

48 

Mansi R, Plas P, Vauquelin G and Fani M: Distinct in vitro binding profile of the somatostatin receptor subtype 2 antagonist [177Lu]Lu-OPS201 compared to the agonist [177Lu]Lu-DOTA-TATE. Pharmaceuticals (Basel). 14(1265)2021.PubMed/NCBI View Article : Google Scholar

49 

Zboralski D, Hoehne A, Bredenbeck A, Schumann A, Nguyen M, Schneider E, Ungewiss J, Paschke M, Haase C, von Hacht JL, et al: Preclinical evaluation of FAP-2286 for fibroblast activation protein targeted radionuclide imaging and therapy. Eur J Nucl Med Mol Imaging. 49:3651–3667. 2022.PubMed/NCBI View Article : Google Scholar

50 

Zboralski D, Osterkamp F, Christensen E, Bredenbeck A, Schumann A, Hoehne A, Schneider E, Paschke M, Ungewiss J, Haase C, et al: Fibroblast activation protein targeted radiotherapy induces an immunogenic tumor microenvironment and enhances the efficacy of PD-1 immune checkpoint inhibition. Eur J Nucl Med Mol Imaging. 50:2621–2635. 2023.PubMed/NCBI View Article : Google Scholar

51 

Baum RP, Schuchardt C, Singh A, Chantadisai M, Robiller FC, Zhang J, Mueller D, Eismant A, Almaguel F, Zboralski D, et al: Feasibility, biodistribution, and preliminary dosimetry in peptide-targeted radionuclide therapy of diverse adenocarcinomas using 177Lu-FAP-2286: First-in-humans results. J Nucl Med. 63:415–423. 2022.PubMed/NCBI View Article : Google Scholar

52 

Minczeles NS, Bos EM, de Leeuw RC, Kros JM, Konijnenberg MW, Bromberg JEC, de Herder WW, Dirven CMF, Hofland J and Brabander T: Efficacy and safety of peptide receptor radionuclide therapy with [177Lu]Lu-DOTA-TATE in 15 patients with progressive treatment-refractory meningioma. Eur J Nucl Med Mol Imaging. 50:1195–1204. 2023.PubMed/NCBI View Article : Google Scholar

53 

Nishida H, Kondo Y, Kusaba T, Kadowaki H and Daa T: Immunohistochemical reactivity of prostate-specific membrane antigen in salivary gland tumors. Head Neck Pathol. 16:427–433. 2022.PubMed/NCBI View Article : Google Scholar

54 

van Boxtel W, Uijen MJM, Verhaegh GW, Willems SM and Jonker MA: PALGA Group. Schalken JA, van Engen-van Grunsven ICH and van Herpen CML: Prognostic value of PSMA, c-MET and E-cadherin in salivary duct carcinoma. Oral Oncol. 110(105018)2020.PubMed/NCBI View Article : Google Scholar

55 

van Boxtel W, Lütje S, van Engen-van Grunsven ICH, Verhaegh GW, Schalken JA, Jonker MA, Nagarajah J, Gotthardt M and van Herpen CML: 68Ga-PSMA-HBED-CC PET/CT imaging for adenoid cystic carcinoma and salivary duct carcinoma: A phase 2 imaging study. Theranostics. 10:2273–2283. 2020.PubMed/NCBI View Article : Google Scholar

56 

Uijen MJM, Derks YHW, Merkx RIJ, Schilham MGM, Roosen J, Privé BM, van Lith SAM, van Herpen CML, Gotthardt M, Heskamp S, et al: PSMA radioligand therapy for solid tumors other than prostate cancer: Background, opportunities, challenges, and first clinical reports. Eur J Nucl Med Mol Imaging. 48:4350–4368. 2021.PubMed/NCBI View Article : Google Scholar

57 

Lütje S, Sauerwein W, Lauenstein T, Bockisch A and Poeppel TD: In vivo visualization of prostate-specific membrane antigen in adenoid cystic carcinoma of the salivary gland. Clin Nucl Med. 41:476–477. 2016.PubMed/NCBI View Article : Google Scholar

58 

Klein Nulent TJW, van Es RJJ, Willems SM, Braat AJAT, Devriese LA, de Bree R and de Keizer B: First experiences with 177Lu-PSMA-617 therapy for recurrent or metastatic salivary gland cancer. EJNMMI Res. 11(126)2021.PubMed/NCBI View Article : Google Scholar

59 

Terroir M, Lamesa C, Krim M, Vija L, Texier JS, Cassou-Mounat T, Delord JP, Vallot D and Courbon F: Radioligand therapy with [177Lu]Lu-PSMA-617 for salivary gland cancers: Literature review and first compassionate use in France. Pharmaceuticals (Basel). 16(754)2023.PubMed/NCBI View Article : Google Scholar

60 

Lütje S, Gomez B, Cohnen J, Umutlu L, Gotthardt M, Poeppel TD, Bockisch A and Rosenbaum-Krumme S: Imaging of prostate-specific membrane antigen expression in metastatic differentiated thyroid cancer using 68Ga-HBED-CC-PSMA PET/CT. Clin Nucl Med. 42:20–25. 2017.PubMed/NCBI View Article : Google Scholar

61 

Lawhn-Heath C, Yom SS, Liu C, Villanueva-Meyer JE, Aslam M, Smith R, Narwal M, Juarez R, Behr SC, Pampaloni MH, et al: Gallium-68 prostate-specific membrane antigen ([68Ga]Ga-PSMA-11) PET for imaging of thyroid cancer: A feasibility study. EJNMMI Res. 10(128)2020.PubMed/NCBI View Article : Google Scholar

62 

Sollini M, Kirienko M, di Tommaso L, Pini C, Gelardi F, Ariano S, Lania AG, Mazziotti G, Mercante G and Chiti A: The complementary role of PSMA expression and [18F]FDG PET/CT in predicting thyroid cancer outcome: From black and white to shades of gray, in the era of precision oncology. EJNMMI Res. 13(54)2023.PubMed/NCBI View Article : Google Scholar

63 

Pitalua-Cortes Q, García-Perez FO, Vargas-Ahumada J, Gonzalez-Rueda S, Gomez-Argumosa E, Ignacio-Alvarez E, Soldevilla-Gallardo I and Torres-Agredo L: Head-to-head comparison of 68Ga-PSMA-11 and 131I in the follow-up of well-differentiated metastatic thyroid cancer: A new potential theragnostic agent. Front Endocrinol (Lausanne). 12(794759)2021.PubMed/NCBI View Article : Google Scholar

64 

Assadi M and Ahmadzadehfar H: 177Lu-DOTATATE and 177Lu-prostate-specific membrane antigen therapy in a patient with advanced metastatic radioiodine-refractory differentiated thyroid cancer after failure of tyrosine kinase inhibitors treatment. World J Nucl Med. 18:406–408. 2019.PubMed/NCBI View Article : Google Scholar

65 

de Vries LH, Lodewijk L, Braat AJAT, Krijger GC, Valk GD, Lam MGEH, Borel Rinkes IHM, Vriens MR and de Keizer B: 68Ga-PSMA PET/CT in radioactive iodine-refractory differentiated thyroid cancer and first treatment results with 177Lu-PSMA-617. EJNMMI Res. 10(18)2020.PubMed/NCBI View Article : Google Scholar

66 

Pastoreková S, Parkkila S, Parkkila AK, Opavský R, Zelník V, Saarnio J and Pastorek J: Carbonic anhydrase IX, MN/CA IX: Analysis of stomach complementary DNA sequence and expression in human and rat alimentary tracts. Gastroenterology. 112:398–408. 1997.PubMed/NCBI View Article : Google Scholar

67 

Steffens MG, Oosterwijk-Wakka JC, Zegwaart-Hagemeier NE, Boerman OC, Debruyne FM, Corstens FH and Oosterwijk E: Immunohistochemical analysis of tumor antigen saturation following injection of monoclonal antibody G250. Anticancer Res. 19:1197–1200. 1999.PubMed/NCBI

68 

Stillebroer AB, Boerman OC, Desar IME, Boers-Sonderen MJ, van Herpen CML, Langenhuijsen JF, Smith-Jones PM, Oosterwijk E, Oyen WJ and Mulders PF: Phase 1 radioimmunotherapy study with lutetium 177-labeled anti-carbonic anhydrase IX monoclonal antibody girentuximab in patients with advanced renal cell carcinoma. Eur Urol. 64:478–485. 2013.PubMed/NCBI View Article : Google Scholar

69 

Muselaers CH, Boers-Sonderen MJ, van Oostenbrugge TJ, Boerman OC, Desar IM, Stillebroer AB, Mulder SF, van Herpen CM, Langenhuijsen JF, Oosterwijk E, et al: Phase 2 study of lutetium 177-labeled anti-carbonic anhydrase IX monoclonal antibody girentuximab in patients with advanced renal cell carcinoma. Eur Urol. 69:767–770. 2016.PubMed/NCBI View Article : Google Scholar

70 

Elboga U, Kilbas B, Sahin E, Cayırlı YB, Eryilmaz K, Begec T, Bakar HE, Mercanoglu G and Celen YZ: An automated synthesis of 177Lu-EDTMP as an efficient bone-seeking therapeutic radiopharmaceutical. Eur Rev Med Pharmacol Sci. 25:4829–4834. 2021.PubMed/NCBI View Article : Google Scholar

71 

Askari E, Harsini S, Vahidfar N, Divband G and Sadeghi R: 177Lu-EDTMP for metastatic bone pain palliation: A systematic review and meta-analysis. Cancer Biother Radiopharm. 36:383–390. 2021.PubMed/NCBI View Article : Google Scholar

72 

Mercanoglu G, Zilbeyaz K and Arslan N: Synthesis and ready to use kit formulation of EDTMP for the preparation of 177Lu-EDTMP as a bone palliation radiopharmaceutical. Curr Radiopharm. 16:38–43. 2023.PubMed/NCBI View Article : Google Scholar

73 

Chen H, Zhao L, Fu K, Lin Q, Wen X, Jacobson O, Sun L, Wu H, Zhang X, Guo Z, et al: Integrin αvβ3-targeted radionuclide therapy combined with immune checkpoint blockade immunotherapy synergistically enhances anti-tumor efficacy. Theranostics. 9:7948–7960. 2019.PubMed/NCBI View Article : Google Scholar

74 

Kim C, Liu SV, Subramaniam DS, Torres T, Loda M, Esposito G and Giaccone G: Phase I study of the 177Lu-DOTA0-Tyr3-octreotate (lutathera) in combination with nivolumab in patients with neuroendocrine tumors of the lung. J Immunother Cancer. 8(e000980)2020.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

May-2024
Volume 27 Issue 5

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Niu T, Fan M, Lin B, Gao F, Tan B and Du X: Current clinical application of lutetium‑177 in solid tumors (Review). Exp Ther Med 27: 225, 2024
APA
Niu, T., Fan, M., Lin, B., Gao, F., Tan, B., & Du, X. (2024). Current clinical application of lutetium‑177 in solid tumors (Review). Experimental and Therapeutic Medicine, 27, 225. https://doi.org/10.3892/etm.2024.12514
MLA
Niu, T., Fan, M., Lin, B., Gao, F., Tan, B., Du, X."Current clinical application of lutetium‑177 in solid tumors (Review)". Experimental and Therapeutic Medicine 27.5 (2024): 225.
Chicago
Niu, T., Fan, M., Lin, B., Gao, F., Tan, B., Du, X."Current clinical application of lutetium‑177 in solid tumors (Review)". Experimental and Therapeutic Medicine 27, no. 5 (2024): 225. https://doi.org/10.3892/etm.2024.12514