Skip to main content
Cell Reports Medicine logoLink to Cell Reports Medicine
. 2023 Mar 31;4(4):100960. doi: 10.1016/j.xcrm.2023.100960

A 211At-labelled mGluR1 inhibitor induces cancer senescence to elicit long-lasting anti-tumor efficacy

Lin Xie 1,5,, Lulu Zhang 1,2,5, Kuan Hu 1,4,5, Masayuki Hanyu 1, Yiding Zhang 1, Masayuki Fujinaga 1, Katsuyuki Minegishi 1, Takayuki Ohkubo 1, Kotaro Nagatsu 1, Cuiping Jiang 1,2, Takashi Shimokawa 3, Kazuma Ashisuke 3, Noriyuki Okonogi 3, Shigeru Yamada 3, Feng Wang 2, Rui Wang 4, Ming-Rong Zhang 1,6,∗∗
PMCID: PMC10140459  PMID: 37003259

Summary

Metabotropic glutamate receptor 1 (mGluR1), a key mediator of glutamatergic signaling, is frequently overexpressed in tumor cells and is an attractive drug target for most cancers. Here, we present a targeted radiopharmaceutical therapy strategy that antagonistically recognizes mGluR1 and eradicates mGluR1+ human tumors by harnessing a small-molecule alpha (α)-emitting radiopharmaceutical, 211At-AITM. A single dose of 211At-AITM (2.96 MBq) in mGluR1+ cancers exhibits long-lasting in vivo antitumor efficacy across seven subtypes of four of the most common tumors, namely, breast cancer, pancreatic cancer, melanoma, and colon cancers, with little toxicity. Moreover, complete regression of mGluR1+ breast cancer and pancreatic cancer is observed in approximate 50% of tumor-bearing mice. Mechanistically, the functions of 211At-AITM are uncovered in downregulating mGluR1 oncoprotein and inducing senescence of tumor cells with a reprogrammed senescence-associated secretory phenotype. Our findings suggest α-radiopharmaceutical therapy with 211At-AITM can be a useful strategy for mGluR1+ pan-cancers, regardless of their tissue of origin.

Keywords: metabotropic glutamate receptor 1, oncoprotein, alpha-emitting radiopharmaceutical, 211At-AITM, targeted alpha radiopharmaceutical therapy, pan-cancer, glutamine metabolism, α−particle, senescence, reprogrammed senescence-associated secretory phenotype

Graphical abstract

graphic file with name fx1.jpg

Highlights

  • Most cancer patients, across 32 different tumor types, aberrantly express mGluR1

  • α-Radiopharmaceutical 211At-AITM binds to mGluR1-expressing multiple human cancers

  • A single dose of 211At-AITM elicits durable antitumor effect in mGluR1+ pan-cancers

  • 211At-AITM decreases mGluR1 and modulates cancer senescence with reprogrammed SASP


Xie et al. report a targeted radiopharmaceutical therapy strategy that antagonistically recognizes mGluR1, a key mediator of glutamatergic signaling, and eradicates mGluR1+ human tumors by harnessing an α-radiopharmaceutical, 211At-AITM. A single injection of 211At-AITM in mGluR1+ pan-cancers elicits long-lasting antitumor efficacy and even cures, regardless of their tissue origins.

Introduction

Metabolic reprogramming has been recognized as one of the 14 hallmarks of cancer.1 Cancer cells alter their glucose and glutamine metabolism to maintain bioenergetics, redox status, cell signaling, and biosynthesis.2,3,4,5 The discovery of the Warburg effect, increasing aerobic glycolysis, led to the extensive use of 18F-fluorodeoxyglucose (18F-FDG) as the main positron emission tomography (PET) tracer to map glucose metabolism for cancer detection, staging, and response monitoring in the clinic.6,7,8 In addition to glucose metabolism, proliferating cancer cells rely on increased glutamine metabolism (glutaminolysis) to maintain the functioning of the tricarboxylic acid cycle and produce glutathione as a precursor for nucleotide and lipid synthesis.2,9 Notably, glutamate is an immediate product of glutamine metabolism catalyzed by glutaminase.10 The central role of glutamine/glutamate metabolism in cancer bioenergetics and biosynthesis has reinforced the interest in key receptors of glutamatergic signaling.

Indeed, the overexpression of glutamate receptors, triggered by glutamate, has been detected in several cancer types and contributes to tumor cell growth via paracrine or autocrine signaling.11,12,13 Among these tumor-associated glutamate receptors, metabotropic glutamate receptor 1 (protein: mGluR1; gene: GRM1), a seven-transmembrane domain G protein-coupled receptor,14 has been shown to be overexpressed in a variety of cancers (such as melanoma,11 breast cancer,15 prostate cancer,16 chondromyxoid fibroma,17 and lung cancer18). mGluR1 is normally expressed in the CNS and is involved in memory and learning.19 However, its aberrant overexpression led to neoplastic transformation in vitro and tumorigenesis in vivo.20 Binding of glutamate, the endogenous ligand of mGluR1, activates the mitogen-activated protein kinase (MAPK), phosphatidylinositol-3 kinase/Ak strain transforming (AKT), and AKT/mammalian target of rapamycin (mTOR)/hypoxia-inducible factor 1 pathways and is involved in numerous aspects of tumor cell growth, metastasis, angiogenesis, and survival.21,22 The oncogenic effect of mGluR1 in mammary tumors was revealed when full-length wild-type GRM1 exogenously introduced in melanocytes and epithelial cells independently drove carcinogenesis with 100% penetrance.11,23

Genetic and pharmacological interventions of mGluR1 have been performed and have resulted in clinically tested drug candidates.22,24,25 Genetically, mGluR1 silencing using GRM1 small hairpin RNA diminished cell proliferation, migration, and invasion in triple-negative breast cancers (TNBCs).23 Pharmacologically, the inhibition of mGluR1 activity using antagonists such as riluzole,26 LY367385,27 and BAY36-762015 inhibited tumor growth, decreased blood vessel density, and induced DNA damage in mGluR1-overexpressing (mGluR1+) melanoma and TNBCs, but not in mGluR1-negative (mGluR1) cancers. A phase 2 clinical trial of riluzole for antitumor therapy in stage III and IV melanoma patients yielded short-term stable disease in 30%–46% of patients.22 Nevertheless, the single agent riluzole is unlikely to have a long-lasting benefit in melanoma patients. Combinatorial therapies with riluzole and γ-irradiation28 or immunotherapy29 have also been studied; however, these approaches failed to induce durable remission in any type of cancer.

As the majority of conventional mGluR1-targeting strategies are devoted to transiently modulating mGluR1 activity through antagonistic binding, the subsequent consequence is usually revealed to be moderate and susceptible to metabolic compensatory regulation. To overcome this bottleneck associated with mGluR1 antagonists, there is an urgent need for therapeutic strategies that can hinder the proliferation potential of mGluR1+ tumor cells via direct DNA double-strand breaks (DSBs) and reduce the oncoprotein expression to simultaneously block tumorigenesis. Tumor targeted alpha (α) radiopharmaceutical therapy (α-RPT), which refers to the targeted delivery of lethal α-particles to cancerous cells mediated by specific tumor-binding ligands, thus eliminating tumor cells by inducing DNA DSBs, has been demonstrated to be a remarkable therapeutic approach.30,31,32 In this context, we envisioned that mGluR1 ligand-based RPT might represent a remedy for conventional molecular targeted therapy that fails in early clinical trials. For targeted α-RPT, whether the oncoprotein phenotype would be altered after radiation has been scarcely studied. Moreover, tumor cell responses against radiation exposure are usually cellular and genetic type and dosimetry dependent. To develop effective targeted RPT, it is essential to fully understand these issues, especially what will occur to the oncoprotein and the tumor cells after radiation and why it occurs.

Herein, we report a targeted α-RPT strategy for mGluR1+ human tumors using a small molecule-based α-emitting radiopharmaceutical (termed 211At-AITM) (Figures 1A and 1B).33 We verified the therapeutic efficacy of 211At-AITM in a variety of human tumors with different tissue origins and distinct mGluR1 expression levels, including breast cancer, pancreatic cancer, melanoma, and colon cancer. Furthermore, we analyzed the phenotypic variations and exact cell fate of tumor cells after 211At-AITM exposure. We found functions of 211At-AITM in downregulating mGluR1 expression and inducing senescence of tumor cells with a reprogrammed senescence-associated secretory phenotype (SASP), except for its classical role in causing DNA DSBs. Consequently, the divergent outcomes showed that synergism in expanding the therapeutic index and eliciting a durable damage signal led to long-lasting benefit and even cure in multiple human solid cancers. Taken together, 211At-AITM based RPT represents a two-birds-one-stone strategy (211At-AITM is the stone, which causes phenotypic and genetic variations) for tackling mGluR1+ pan-cancers.

Figure 1.

Figure 1

Construction of a mGluR1 targeted 211At-AITM RPT strategy for the tumor-agnostic treatment of cancers

(A) Left: Chemical structure of 211At-AITM. Right: Radio-characteristics of 211At-AITM.

(B) 211At-AITM docking to the human mGluR1 seven-transmembrane crystal structure. See also Figure S2.

(C) The percentage of GRM1+ in human cancers from different tissues of origin. Data were collected from TCGA. CESC, cervical squamous cell carcinoma and endocervical adenocarcinoma. See also Figure S1.

(D) Validation of mGluR1 protein expression levels in seven representative cancer cell lines from four solid cancer types using immunofluorescence staining. Green, Alexa Fluor 488-labeled anti-mGluR1 antibody. Scale bar, 50 μm. Representative images are shown from three independent experiments.

(E) Quantification of GRM1 mRNA expression using qRT-PCR.

(F) Specific binding ability of 211At-AITM in a panel of human cancers with and without mGluR1 expression after 1, 3, and 21 h of treatment.

(G) Pearson correlation analysis between the level of GRM1 expression and the uptake of 211At-AITM.

(H) Biospecificity of 211At-AITM based on competitive co-incubation with the unlabeled mGluR1 antagonist FITM (10 μmol/L) for 3 h.

(I) Cellular localization of 211At-AITM action after 1, 3, and 21 h of treatment. (E, F, H, and I) Results are expressed as mean ± SEM, n = 3–4. All comparisons were performed using an unpaired two-tailed Student’s t test or two-way ANOVA followed by Tukey’s multiple comparison test.

Results

GRM1 is widely expressed in human cancers

Pan-cancer analysis of GRM1 expression in The Cancer Genome Atlas (TCGA) database data revealed that GRM1 expression was universally present in 27.03%–100% of human cancers across distinct histological subtypes (to date, 32 major tumor types covering 10,967 clinical samples have been identified) (Figures 1C, S1A, and S1B). A high percentage of GRM1 positivity was observed in most common solid cancers, such as breast cancer (96.13% [1,042/1,084]), pancreatic cancer (88.04% [162/184]), melanoma (97.32% [436/448] in skin melanoma and 87.50% [70/80] in uveal melanoma), and colon cancer (63.80% [379/594]). Mutation at Thr8157.38 and Pro7565.43 of GRM1 only represented 0.23% (1/442) of all human tumor samples, which were previously reported to reduce the affinity and potency of small-molecule mGluR1 antagonists, including 4-fluoro-N-[4-[6-(isopropylamino)pyrimidin-4-yl]-1,3-thiazol-2-yl]-N-methylbenzamide (FITM).14 Our findings indicate that most cancer patients, across 32 different tumor types, aberrantly express mGluR1 and could be good candidates for the mGluR1-targeting α-RPT.

In this study, we profiled and treated a panel of the most common solid cancers, including seven cancer cell lines of four cancer types (MDA-MB231 TNBC; MIA PaCa2 and PANC1 pancreatic cancers; A375, Bowes and A2058 melanomas; and DLD1 colon cancer). mGluR1 expression in these cell lines was assessed at the protein level using immunocytochemistry (Figure 1D) and confirmed at the mRNA level using quantitative real-time reverse transcriptase-polymerase chain reaction (qRT-PCR) (Figure 1E). Five tumor cell lines (MDA-MB231, MIA PaCa2, Bowes, A375, and DLD1) exhibited differential mGluR1 expression (Figures 1D and 1E). In comparison, mGluR1 expression was negligible in A2058 and PANC1 cell lines.

211At-AITM specifically binds to mGluR1-expressing multiple human cancer cells

Ligand receptor-mediated targeting is a facile and effective approach for radiopharmaceutical design. Based on this approach, we previously designed the α-emitting radiopharmaceutical 211At-AITM (Figures 1A and 1B),33 which was derived from a high-affinity, small-molecule mGluR1 antagonist, FITM (half-maximal inhibitory concentration = 5.1 nM),34 whereas the two compounds are distinguished by the type of halogen atoms. 211At-AITM retains high binding affinity and selectivity to murine and human mGluR1 as revealed by molecular dynamics simulations (Figures S2A–S2D). Notably, the bulky 211At atom can prevent 211At-AITM from entering the mGluR1-rich brain,33,35,36,37 thus avoiding detrimental radiation to the CNS, which is a major concern associated with the clinical translation of mGluR1-targeted radiotherapies.

The binding characteristics of 211At-AITM were measured by incubating a subset of the human cancer cells with 211At-AITM for 1, 3, and 21 h. 211At-AITM displayed a high binding ability to mGluR1+ MDA-MB231, MIA PaCa2, Bowes, A375, and DLD1 cells after 1 h and maintained consistently high uptake at 3 and 21 h, while exhibiting only low uptake in mGluR1 A2058 and PANC1 cells at all time points (Figure 1F). A close positive correlation was found between the GRM1 expression and the uptake of radioactivity at 1, 3, and 21 h after 211At-AITM incubation (Figure 1G). To clarify the specificity of 211At-AITM for mGluR1, we used excess unlabeled FITM (10 μmol/L), which competes for the same receptor, in these cancer cells. As expected, the uptake of 211At-AITM was significantly decreased in mGluR1+ MDA-MB231 and MIA PaCa2 cells but not in mGluR1 A2058 and PANC1 cells (Figure 1H). The cellular localization of 211At-AITM action was also investigated38; more than half of the radioactivity was anchored on the membrane of MDA-MB231 cells (68.71%–73.25% CD; the value is quantified for a percentage of cell-associated radioactivity dose, similarly hereinafter) and MIA PaCa2 cells (49.42%–61.45% CD), while the remaining radioactivity was rapidly internalized by cancer cells (Figure 1I). These results indicate that 211At-AITM specifically enters cancer cells expressing mGluR1 at high and stable concentrations and could act on the membrane, as well as in the interior of cancer cells, thus potentially providing double benefits for targeted α-RPT.

211At-AITM induces mGluR1 downregulation and locks cancer cells into senescence

Given the reported oncogenic role of mGluR1 when aberrantly expressed in cells, we next assessed the effects of 211At-AITM on mGluR1-expressing cancer cells. We treated mGluR1+ MDA-MB231 cells with different doses of 211At-AITM (Figures S3A and S3B), then chose a dose of 18.5 kBq/mL for subsequent cell culture experiments. MDA-MB231 and MIA PaCa2 cancer cells were incubated with 211At-AITM for 1 day (day 1) followed by 7 days of culture without 211At-AITM (days 2–7). Real-time cell survival rate measurements showed that 211At-AITM did not trigger immediate and massive cell death (Figures 2A and S3A). Most cancer cells remained viable on day 7 post-therapy (Figure 2A); however, cell proliferation was obviously impaired as the cell number doubling time was significantly prolonged in MDA-MB231 (vehicle vs. treatment = 38.22 ± 0.75 h vs. 71.18 ± 3.97 h; p < 0.0001) and MIA PaCa2 (vehicle vs. treatment = 27.54 ± 0.63 h vs. 37.31 ± 0.95 h; p < 0.0001) cells after 211At-AITM treatment. Cell cycle analysis showed that treatment with 211At-AITM in MDA-MB231 and MIA PaCa2 cancer cells induced a rapid shift from the G0/G1 phase and S phase to the G2/M phase and caused cell arrest in the G2/M phase over a long period of time after α-irradiation (Figures 2B, 2C, S3B–S3D). As shown in Figures 2D and 2G, a few multinucleated giant cells were observed, which is likely the result of mitotic catastrophe,39 consistent with a small fraction of cells dying after 211At-AITM exposure (Figure 2A).

Figure 2.

Figure 2

211At-AITM downregulates mGluR1 and induces senescence in mGluR1+ cancer cells

(A) Survival rate of mGluR1+ MDA-MB231 and MIA PaCa2 cells. Cancer cells were treated with 211At-AITM or vehicle (PBS) for 1 day (day 1), incubated for 7 days without drug (days 2–7), and pretreated on day 0. Survival rate (to vehicle) of cells measured using Muse Cell Analyzer for up to 7 days.

(B and C) Cell cycle state distribution. Data show the results from cells in (A). See also Figures S3C and S3D.

(D) Representative fluorescence images of mGluR1 (green) and DNA (blue) in MDA-MB-231 and MIA PaCa2 cells. Scale bar, 50 μm.

(E and F) Quantification of mGluR1 expression.

(G and I) Representative fluorescence images and quantification of γH2AX (red) and DNA (blue) to assess DNA DSBs. Scale bar, 50 μm.

(J) Representative fluorescence images of SA-β-gal (green) to assess senescence. Scale bar, 50 μm.

(K and L) Percentages of cells that showed SA-β-gal positive activity.

(M and N) Pearson correlation analysis of the mGluR1 expression and the marker of DNA DSBs γH2AX or the marker of senescence SA-β-Gal in MDA-MB231 and MIA PaCa2 cells. (A–C, E, F, H, I, K, and L) Results are expressed as mean ± SEM, n = 4. All comparisons were performed using an unpaired two-tailed Student’s t test or two-way ANOVA followed by Tukey’s multiple comparison test. Asterisks indicate statistical significance (p < 0.05, ∗∗p < 0.01), ns indicates no significance. (D, G, and J) Representative images are shown from three independent experiments.

To assess why 211At-AITM triggered long-term growth arrest of cancer cells, we first evaluated the mGluR1 protein expression associated with oncogenic activity and confirmed the mRNA level. We observed a significant and stable reduction in mGluR1 protein expression in both MDA-MB231 and MIA PaCa2 cells after 1–7 days (Figures 2D–2F). Compared with the observed following vehicle treatment, the GRM1 mRNA expression was not detectable in either MDA-MB231(Figure S3E) or MIA PaCa2 cells (Figure S3F) after 2–7 days of treatment, although there was no significant GRM1 mRNA change between vehicle or treated cell samples at the 1-day timepoint. These results indicated that 211At-AITM treatment induced the mGluR1 downregulation not only at the protein level (Figures 2D–2F), but also at the mRNA level (Figures S3E and S3F) in mGluR1+ cancer cells.

Next, we assessed the protein expression associated with DNA damage after treatment with 211At-AITM. The results showed that the signal of γH2AX, a marker of DNA DSBs, was notable and not lost over time after α-irradiation, indicating that 211At-AITM caused clustered DNA lesions and persistent DNA damage response (DDR) activation in most MDA-MB231 and MIA PaCa2 cancer cells, which was not observed in vehicle-treated cells (Figures 2G and 2I). Then, we assessed senescence-associated beta-galactosidase (SA-β-gal), a biomarker of senescence, in the treated cancer cells. The SA-β-gal+ cells were enriched in MDA-MB231 and MIA PaCa2 cells on day 1 and robustly upregulated on days 2–7 after 211At-AITM treatment, but not in the vehicle-treated cells (Figures 2J–2L). Simultaneously, the cell morphology apparently changed as the cells became enlarged and flattened with increasing SA-β-gal concentration (Figure 2J). Figures 2M and 2N showed scatterplots and fitted curves of mGluR1 expression versus γH2AX or SA-β-Gal positive cells on days 1–7 after 211At-AITM treatment. Significant correlations were indicated between the mGluR1 expression and the γH2AX-positive cells, and between the mGluR1 expression and the SA-β-Gal+ cells for each timepoint in MDA-MB231 and MIA PaCa2 cancer cells. Overall, our results demonstrate that 211At-AITM downregulated mGluR1 expression, induced DSBs, and triggered cell senescence, which cooperatively governed cancer cell fates.

211At-AITM is a safe and effective precision radiopharmaceutical for mGluR1-expressing cancers

To explore whether the 211At-AITM characteristics observed in vitro could be translated into therapeutic efficacy in vivo, we carried out 211At-AITM therapeutic experiments in multiple mouse xenograft models. First, the pharmacokinetics of intravenously injected 211At-AITM were evaluated in human cancer-bearing mice (Figures 3A and 3B). 211At-AITM showed peak tumor accumulation of radioactivity with 4.05%–5.01% ID/g (the value is quantified for a mean percentage of the injected radioactivity dose per gram of tissue, similarly hereinafter) at 1 h post-injection, which decreased to 3.66%–4.57% ID/g at 3 h and 0.22%–0.57% ID/g at 21 h post-injection in mGluR1+ human cancers. In contrast, 211At-AITM constantly showed low accumulation with 1.80%–2.44% ID/g at 1 h and 0.45%–1.05% ID/g at 3 and 21 h in mGluR1 human cancers (Figure 3A). A correlation analysis identified a significantly positive association between the mGluR1 expression and the uptake in vivo at 1 and 3 h, but no association at 21 h post-injection in tumor xenografts (Figure S4A). The radiation doses absorbed were 3.79–4.62 Gy/MBq for mGluR1+ human cancers and 1.16–2.05 Gy/MBq for mGluR1 human cancers (Figure 3C), based on a standard method using the Medical Internal Radiation Dose formula.40 The biological effective doses to mGluR1+ human cancers were 18.95–23.09 Sv/MBq and to mGluR1 human cancers were 5.8–10.26 Sv/MBq (Figure 3C), based on the quality factor of 5.41,42 Minimal radioactivity was observed in the thyroid (<0.13% ID) at the three time points, thus indicating high in vivo stability of 211At-AITM with a low level of deastatination.43 A high accumulation with 10.17–6.28 %ID/g was observed in the stomach, probably because of the physiological mGluR1 expression in the tissue.44

Figure 3.

Figure 3

211At-AITM accumulates rapidly into mGluR1+ tumor xenografts in human cancer-bearing mice

(A) Quantitative measurement of radioactivity in human cancers following ex vivo biodistribution analysis at 1, 3, and 21 h after 211At-AITM injection.

(B) Quantitative measurement of radioactivity in major organs of the representative MDA-MB231-bearing mice after 211At-AITM injection. Note that thyroid values are presented as a percentage of the injected radioactivity dose (% ID).

(C) Absorbed doses and biological effective doses in human cancers. Absorbed doses were estimated in accordance with the standard method using the medical internal radiation dose formula, expressed as the mean dose per unit of injected activity (gray per MBq; Gy/MBq).40 The biological effective dose was calculated by using a quality factor of 5, expressed as sievert per MBq (Sv/MBq).41,42

(D) Absorbed doses in major organs of the representative MDA-MB231-bearing mice.

(E) Biological effective doses in major organs of the representative MDA-MB231-bearing mice. (A and B) Data are expressed as mean percentage of the injected radioactivity dose per gram of tissue (% ID/g) ± SEM, n = 3 for each time point. Comparisons were performed using an unpaired two-tailed Student’s t test or two-way ANOVA followed by Tukey’s multiple comparison test.

As expected, extremely low radioactivity was observed in the brain of not only 8-week-old adult mice, but also 115-week-old aged mice after 211At-AITM injection (Figures 3B and S4C), although normal brain tissue exhibits high mGluR1 expression; therefore, we concluded that chemical modifications and the use of the biggest halogen atom 211At minimized the radiation exposure of the brain. Although high radioactivity was observed in the lung (22.39% ID/g) and spleen (5.86% ID/g) at 1 h (Figures 3B, 3D, and 3E), rapid clearance of radioactivity from blood and these tissues began at 3 h after injection. At 21 h post-injection, except for a low level of radioactivity in the targeted tumor tissues, no considerable amounts of radioactivity were detected in the nontargeted tissues of mice. The good pharmacokinetic profile of 211At-AITM ensures minimized radiation exposure in healthy off-target organs.

Next, the in vivo antitumor efficacy of 211At-AITM was tested in xenograft mice bearing seven subtypes of cells characteristic to four types of solid tumors. Given the complex relationship between p53 status and radiation damage responses in tumors,45 we defined the p53 status of the panel of human solid cancer cells mutant-type p53 (p53mt) or wild-type p53 (p53wt) allele using the PubMed database. The xenograft mice were treated with a single injection of 211At-AITM (2. 96 MBq), a dose that was chosen based on previous studies33 and our pilot data (Figures S5A and S5B). In contrast with its minimal effect against mGluR1 cancers (A2058 with p53mt and PANC1 with p53mt), a single administration of 211At-AITM in mGluR1+ (MDA-MB231 with p53mt, MIAPaCa2 with p53mt, Bowes with p53 wt, A375 with p53 wt, and DLD1 with p53mt) cancers exhibited unequivocal and durable antitumor efficacy across breast cancer, pancreatic cancer, melanoma, and colon cancer, regardless of their tissue of origin (Figures 4A–4C). A significant inverse correlation was verified between the mGluR1 expression and the tumor volumes on day 30 post-injection (Figure S4B) (Pearson r = −0.6379, p = 0.0019). Furthermore, the antitumor efficacies of 211At-AITM were independent of the p53 status, unlike conventional radiotherapy.45 Notably, complete regression was observed in 42.11% (8/19) of MDA-MB231 TNBC-bearing mice between 13 and 28 days and 54.55% (6/11) of MIA PaCa2 pancreatic cancer-bearing mice between 13 and 16 days after a single 211At-AITM treatment (Figures 4A and 4B). No recurrence was observed until the end of this investigation (Figures 4A and S6A–S6C). To clarify the specificity of therapeutic effect of 211At-AITM in vivo, the xenograft mice bearing mGluR1+ MIA PaCa2 cancers were treated with unlabeled FITM (1 mg/kg) or different doses of 211At-AITM. No significant anti-tumor effect was observed in the FITM-treated group compared with the vehicle-treated group (Figure S5C). However, we found that a single dose of 211At-AITM at 0.37 MBq resulted in a 22.48% reduction in tumor volume, 82.19% at 1.48 MBq, and 95.53% at 2.96 MBq, compared with the 0 MBq (saline)-treated group at 30 days after administration (Figure S5D). The results confirmed the specificity and dose-response effects of 211At-AITM RPT from α-particles of 211At in the mGluR1+ xenograft mice.

Figure 4.

Figure 4

A single treatment with 211At-AITM suppresses tumor growth in multiple models of mGluR1+ cancers

(A) Individual tumor growth curves of mice treated with vehicle (saline) or 211At-AITM. Multiple human cancer cells were grown as tumor xenografts in male and female BALB/c nude mice. Longitudinal progression of tumor volume in tumor-bearing mice treated with a single 211At-AITM or vehicle (injection = day 0).

(B) Mouse tumor volumes on day 30 post-injection. MDA-MB231: n = 8 for vehicle group, n = 19 for 211At-AITM group (male: 16, female: 3); MIA PaCa2: n = 6 for vehicle group, n = 11 for 211At-AITM group (male: 8, female: 3); Bowes: n = 6 for vehicle group, n = 4 for 211At-AITM group; A375: n = 5 for vehicle group, n = 5 for 211At-AITM group; DLD1: n = 4 for vehicle group, n = 4 for 211At-AITM group; A2058: n = 5 for vehicle group, n = 7 for 211At-AITM group; PANC1: n = 5 for vehicle group (male: 4, female: 1), n = 7 for 211At-AITM group (male: 4, female: 3). All comparisons were performed using an unpaired two-tailed Student’s t test.

(C) Changes in body weight. Results are expressed as mean ± SEM, n = 4–11.

Finally, the potential toxicity of intravenously injected 211At-AITM was investigated in multiple human cancer-bearing mice. There was no apparent weight loss (Figure 4C), and no clinical signs of toxicity were observed throughout the examination period. Leukocyte, erythrocyte, and platelet counts were transiently decreased at 5–10 days (Figure 5A). Although blood cell counts recovered to baseline after day 15, management of hematologic functions after 211At-AITM therapy should be considered in clinical trials. Liver enzyme (glutamic oxaloacetic transaminase and glutamic pyruvic transaminase) levels (Figure 5B) and kidney function (blood urea nitrogen and creatinine) index (Figure 5C) did not show significant changes on days 2, 7, and 30 after exposure to 211At-AITM. Histological analyses did not show noticeable histological and structural lesions in the liver, kidney, lung, spleen, and stomach at 30 days after a single dose of 211At-AITM, compared with the vehicle-treated mice (Figure 5D). These results were consistent with the results of enzyme assays (Figures 5B and 5C). 211At-AITM physiologically accumulated in the stomach, leading to a high stomach-absorbed dose with 12.38 Gy/MBq (Figure 3D). In patients, the maximum tolerated dose in the stomach is postulated to be 50 Gy.46 Although stomach histology showed no significant differences in vehicle vs treated mice, stomach protectants during 211At-AITM RPT are advised, especially when high treatment activities are used. These data suggest that the mGluR1-targeted α-RPT strategy for treating multiple human cancers is safe, can be effective, and can induce complete tumor regression after a single treatment.

Figure 5.

Figure 5

211At-AITM is a safe radiopharmaceutical in human cancer-bearing mice

(A) Changes in leukocyte, erythrocyte, and platelet counts. Results are expressed as mean ± SEM, n = 6 per group.

(B) The values of glutamic oxaloacetic transaminase (GOT) and glutamic pyruvic transaminase (GPT).

(C) The values of creatinine and blood urea nitrogen (BUN). (B and C) Results are expressed as mean ± SEM, n = 3 per group. All comparisons were performed using an unpaired two-tailed Student’s t test or two-way ANOVA followed by Tukey’s multiple comparison test.

(D) Histological analysis of liver, kidney, lung, spleen, and stomach on 30 days post-injection. Tissue sections (5 μm) were stained with hematoxylin-eosin staining. Scale bars, 50 μm. Representative images are shown from three independent experiments.

211At-AITM downregulates mGluR1 and modulates cancer senescence with reprogrammed SASP

To gain insight into the mechanism underlying the antitumor effects of 211At-AITM, we performed multiple analyses to dissect the phenotypic changes in tumor tissues after radiation. The expression levels of mGluR1 (Figures 6A and 6B), the cell proliferation marker Ki67 (Figures 6A and 6C), the senescence marker SA-β-gal (Figures 6D, 6E, and 6M), and the tumor neovascularization marker CD31 (Figure 6N) were assessed on days 2, 7, and 30 after 211At-AITM treatment. Immunohistological staining of tumor sections showed that 211At-AITM effectively decreased mGluR1 and Ki67 protein levels in both MDA-MB231 and MIA PaCa2 tumors on day 2 and retained the low levels on day 7 post-injection compared with those observed in the vehicle-treated xenografts (Figures 6A–6C). High Ki67 expression and negligible mGluR1 expression were observed in A2058 and PANC1 xenografts pre- or post-treatment, which is consistent with the result of mGluR1+-selective antitumor efficacy of 211At-AITM. Moreover, 211At-AITM-treated MDA-MB231 and MIA PaCa2 tumors showed increased numbers of senescent SA-β-gal+ cells on day 2, constitutively upregulated on day 7 (Figures 6D and 6E), and still higher levels on days 30 and 45 post-injection compared with the tumors treated with the vehicle (Figures 6M, S6D, and S6E). In contrast, A2058 and PANC1 tumors under the same conditions did not display significant SA-β-gal staining. The results indicated that 211At-AITM treatment induces stable senescence of mGluR1+ cancer cells in vivo, which were consistent with the long-lasting antitumor consequences.

Figure 6.

Figure 6

211At-AITM therapy reduces mGluR1 expression and triggers cancer senescence by reprogramming the SASP variants

(A) Representative fluorescence images of mGluR1 (green), Ki67 (red), and DAPI (blue) performed on frozen sections from mGluR1+ (MDA-MB231 and MIA PaCa2) and mGluR1- (A2058 and PANC1) xenografts treated either with vehicle (saline) or 211At-AITM for 2 and 7 days. Scale bar, 50 μm.

(B and C) Quantification of mGluR1 and Ki67 expression.

(D and E) Representative images and quantification of SA-β-gal (green) expression. Scale bar, 50 μm.

(F and G) Quantification of the trigger of cell senescence 53BP1 and CDKN1A using qRT-PCR.

(H–L) Quantification of SASP components using qRT-PCR.

(M) Representative images (left) and quantification (right) of SA-β-gal (green) expression in the representative mGluR1+ MIA PaCa2 and mGluR1- PANC1 cancers on 30-day after 211At-AITM treatment. Scale bar, 50 μm.

(N) Representative images (left) and quantification (right) of CD31 (green) and DNA (blue) on 30-day following 211At-AITM treatment to assess tumor angiogenesis. Scale bar, 50 μm.

(O and P) Quantification of VEGFA and MET using qRT-PCR on day 30 post-injection. (B, C, F, G, and E–P) Results are expressed as mean ± SEM, n = 4 per group. All comparisons were performed using an unpaired two-tailed Student’s t test or two-way ANOVA followed by Tukey’s multiple comparison test. (A, D, M, and N) Representative images are shown from three independent experiments.

We also assessed the gene expression associated with DDR after treatment with 211At-AITM. The expression of 53BP1 (a biomarker of DDR) and CDKN1A (the gene encoding p21) was significantly upregulated on day 2 and persisted at high levels 7 days after 211At-AITM treatment in mGluR1+ cancers compared with that in mGluR1 cancers (Figures 6F and 6G), although p53 is mutated in both MDA-MB231 and MIA PaCa2 cancer cells. These results suggested that DDR represents a trigger of cell senescence after 211At-AITM treatment, and persistent DDR activation maintains long-term and stable cancer senescence by upregulating p21 expression.

Activated MAPK and mTOR pathways, which are modulated by mGluR1, play important roles in stimulating the production of SASP factors.21,22,47 We hypothesized that reduced mGluR1 expression elicits an extensive range of effects on SASP components. To verify this hypothesis, the expression levels of several indicators of SASP, including pro-inflammatory interleukin-6 (IL-6), CXC chemokine ligand 8 (CXCL8), tumor necrosis factor alpha (TNF-α), vascular endothelial growth factor A (VEGFA), and MET, were measured using qRT-PCR in mGluR1+ (MDA-MB231 and MIA PaCa2) and mGluR1 (A2058 and PANC1) xenografts after 211At-AITM treatment. Consistent with the senescence expansion, the expression levels of the key SASP components IL-6, CXCL8 and TNF-α were significantly increased in 211At-AITM-treated MDA-MB231 and MIA PaCa2 tumors, but not in 211At-AITM-treated mGluR1 tumors (Figures 6H–6J). Surprisingly, a significant decrease in the expression of pro-angiogenic VEGFA and pro-metastatic MET was observed on days 2 and 7 (Figures 6K and 6L), and constitutively low levels on day 30 after α-irradiation in 211At-AITM-treated mGluR1+ cancers, compared with vehicle-treated cancers and 211At-AITM-treated mGluR1 cancers (Figures 6O and 6P). The SASP variants had not been identified in conventional X-ray treatment (Figures S7A–S7J). Furthermore, we investigated tumor angiogenesis by immunohistological staining with an antibody against CD31 in 30 and 45-day tumor xenografts after 211At-AITM RPT.48 The CD31-stained blood vessels showed much fewer signals in MIA PaCa2 and MDA-MB231 tumor tissues than their corresponding control tumors (Figures 6N and S6F–S6G), suggesting that reprogrammed SASP variants suppressed tumor angiogenesis and remodeled the tumor microenvironment into angiogenesis antagonizing and metastasis antagonizing. Overall, these results suggested that 211At-AITM therapy reprogrammed the phenotypes of mGluR1+ cancers, including but not limited to downregulating mGluR1 expression, inducing cell senescence, secreting angiogenesis-antagonizing as well as metastasis-antagonizing SASP components, and suppressing tumor angiogenesis. In coordination with its DNA DSB effects, 211At-AITM and SASP work together to govern the fate of tumor cells.

Discussion

The success of metabolic imaging with 18F-FDG in clinical oncology spurred the exploration of therapeutic strategies targeting mGluR1, a key mediator of glutamatergic signaling, for tumor-agnostic treatment of cancers. However, mGluR1 antagonists are usually associated with drug resistance and unsatisfactory clinical outcomes; thus, we sought to develop alternative therapeutic strategies that can overcome the bottleneck of conventional molecular targeted therapy. Receptor-targeted α-RPT, which is impervious to conventional cellular resistance mechanisms,49 is considered an ideal approach for mGluR1-targeted therapy owing to the metabolism-independent cell-killing effects of α-rays.

In this study, we showed that a single intravenous administration of 211At-AITM resulted in unequivocal and durable tumor growth inhibition in multiple models of mGluR1+ human cancers with little toxicity, regardless of their tissue of origin. Moreover, mGluR1-targeting 211At-AITM therapy led to complete regression of mGluR1+ breast cancer and pancreatic cancer in approximate 50% of tumor-bearing mice. Interestingly, mechanistic studies uncovered the functions of 211At-AITM in reshaping the phenotypic patterns of tumor cells, such as decreasing mGluR1 expression, inducing cell senescence, releasing angiogenesis-antagonizing as well as metastasis-antagonizing SASP variants, and suppressing tumor angiogenesis, beyond its classical function in DNA DSBs. These divergent effects of 211At-AITM cooperatively governed tumor cell fate (Figure 7). Thus, our results facilitate the understanding of the tumor cells cascade responses against α-radiation and pave the way for developing precise α-RPT for mGluR1+ pan-cancers.

Figure 7.

Figure 7

Schematic representation of the potential mechanism through which 211At-AITM kills cancer cells

It is proposed that 211At-AITM RPT inhibits mGluR1 function by decreasing its expression levels, which results in downregulation of the MAPK, phosphatidylinositol-3 kinase (PI3K)/AKT, and AKT/mTOR/hypoxia-inducible factor 1 (HIF1) pathways, leading to the suppression of mGluR1-mediated tumor growth, angiogenesis, and metastasis. Furthermore, the α-particles can trigger irreparable DSBs in targeted cancer cells, which activate a persistent DDR that drives cancer senescence. Lastly, mGluR1 ablation and senescent cancer co-reprogram the SASP components into angiogenesis-antagonizing and metastasis-antagonizing variants to improve the long-term outcome of patients and even cure the disease by co-promoting cancer cell death.

In RPT, radiation is delivered systemically to cancer cells using high-affinity ligands as carriers of radionuclides—primarily β-emitters (β-RPT) or high LET α-emitters (α-RPT).50 The α-RPT is currently attracting interest owing to the superior biological radiation effects of α-rays compared to those of β-rays. The 223Ra dichloride (Xofigo), the first α-radiopharmaceutical approved by the US Food and Drug Administration,32 is used for treating bone metastases in patients with advanced castration-resistant prostate cancer. The mGluR1-targeting α-RPT is an example of targeting cancer metabolism-associated targets for cancer treatment. Notably, 18F-FITM is an appropriate companion diagnostic tracer for 211At-AITM,51 as the two radiopharmaceuticals share similar pharmacokinetics and in vivo biodistribution profiles. For example, both showed rapid tumor uptake and non-target normal tissue clearance. In the scenario of imaging-guided α-RPT, the target engagement of 211At-AITM could be accurately predicted using 18F-FITM PET to provide valuable information for dosimetry and treatment planning. This precision treatment mode provides substantial advantages over conventional antagonistic therapy.

As an oncoprotein that is aberrantly overexpressed on the cell membranes of most cancers, mGluR1 shows only limited background expression in normal peripheral organs,11,19,51 thus making it a potential target for α-RPT. As a result, an elevated administration dose could be considered a feasible method to improve therapeutic outcomes, largely expanding the therapeutic window of tumors with relatively low mGluR1 protein expression. Notably, even though mGluR1 is highly expressed in normal brain tissue, 211At-AITM showed little blood-brain barrier penetrance and extremely low brain activity accumulation. This mitigates any concern of radiation toxicity to the normal human brain. Another safety concern of translating 211At-AITM α-RPT to the clinic is the exposure of patients’ healthy off-target organs to α-radiation. In this regard, it should be noted that the temporarily high radioactivity in the blood, lung, spleen, stomach, liver, and kidneys did not induce body weight reduction, severe hematologic toxicity, apparent liver and kidney functional side effects, or obvious histological and structural lesions of the major organs during the 211At-AITM treatment. Because of the remarkable α-particle flux emitted by 211At (t1/2 = 7.21 h), 211At-AITM led to irreparable DSBs in targeted cancer cells, and effective cell killing can be expected even in the hypoxic regions of solid cancers.52 Furthermore, 211At-AITM used for treating multiple human solid cancers achieved consistently high response rates. Moreover, in MDA-MB231 and MIA PaCa2 tumor-bearing mice, 211At-AITM cured approximate 50% of mice and no recurrence was observed until the end of the investigation. Therefore, the outstanding therapeutic efficacy and favorable safety profile of 211At-AITM highlight its remarkable clinical and translational value associated with systemically targeted therapy of a wide range of human cancers, regardless of their tissue of origin.

Based on the classical theory of radiation biology, α-particles directly or indirectly cause DNA DSBs to induce cell death. In addition to the classical function, our mechanistic investigation into the cellular response against 211At-AITM exposure led to the identification of two roles of 211At-AITM through which it affects tumor cell fate. Specifically, 211At-AITM treatment reshaped the phenotypic features of tumor cells. One is the reduction in mGluR1 expression, while the other is the induction of cancer senescence with reprogrammed SASP variants. The phenotypic variations synergize with genetic alterations to cooperatively govern cell fate.

In summary, we have demonstrated that mGluR1-targeted small-molecule α-RPT could deliver long-lasting therapeutic benefits and even cure a wide range of human cancers, regardless of their tissue of origin. Furthermore, mechanistic studies revealed two roles of 211At-AITM RPT in reshaping the phenotype of tumor cells, including the reduction in mGluR1 expression and induction of cancer senescence with programmed SASP variants. Given the prevalence of glutamine/glutamate dysregulation and mGluR1 overexpression in cancers, α-RPT with 211At-AITM may have broad application prospects and bring us one step closer to winning the fight against cancers.

Limitations of the study

There are several limitations of this study. First, our understanding of the molecular mechanisms underlying the biological effects of 211At-AITM remains insufficient. In the future, research efforts should focus on more extensive in-parallel dosimetry control experiments and longitudinal monitoring of changes in mGluR1 expression within tumor tissues. Second, whether α-radiation or reduced mGluR1 expression alone or both coordinately contribute to the reprogramming of SASP in senescent cancers is currently unclear. Third, the changes in glutamate metabolism induced by mGluR1 downregulation remain unexplored. Furthermore, the correlation between glutamine metabolism variation and senescence in our study was unclear. It should be emphasized that these questions are critical for further clinical application of 211At-AITM RPT in patients with cancer that will, in turn, provide valuable insights for the development of effective combinations of α-RPT with immunotherapies or senolytic therapies against cancer. Following this proof-of-concept study, further studies on the exact mechanism and the translational clinical trial of 211At-AITM RPT with long-term follow-up for the treatment of multiple cancers are ongoing.

STAR★Methods

Key resources table

REAGENT or RESOURCE SOURCE IDENTIFIER
Antibodies

Rabbit anti-mGluR1 LSBio Cat#LS-A882; RRID:AB_592206
Mouse anti-Ki67 BD Pharmingen Cat#550609; RRID:AB_393778
Rat anti-CD31 BD Pharmingen Cat#550274; RRID:AB_393571
Alexa Fluor® 488 goat anti-rat IgG Invitrogen Cat#A-11006; RRID:AB_141373
Alexa Fluor® 488 goat anti-rabbit IgG Invitrogen Cat#A-11008; RRID:AB_143165
Alexa Fluor® 546 goat anti-mouse IgG Invitrogen Cat#A-11003; RRID:AB_141370

Chemicals, peptides, and recombinant proteins

211At-AITM Previously developed in our Lab (Xie et al.33) N/A
DAPI Vector Laboratories Cat#H-1200
DMEM, with glucose, L-glutamine, sodium pyruvate, and sodium bicarbonate Sigma-Aldrich Cat#D6429
MEM Sigma-Aldrich Cat#M4655
RPMI-1640 Sigma-Aldrich Cat#R8758
Trypsin-EDTA solution Gibco Cat# 25300054
Fetal bovine serum Sigma-Aldrich Cat#12103C
Penicillin/streptomycin Gibco Cat#15140122
DAPI Vector Laboratories Cat#H-1200
Matrigel Corning Cat#356234
FITM MedChemExpress Cat#932737-65-0
Sepasol-RNA I Super Nacalai Tesque Cat#09379-84

Critical commercial assays

γH2AX DNA Damage Detection Kit Dojindo Cat#G266
Senescence-associated β-galactosidase staining kits Dojindo Cat#SG02
Muse® Count & Viability Kit Luminex Cat#MCH100102
Muse® Cell Cycle kit Luminex Cat#MCH100106
RNeasy Mini Kit Qiagen Cat#74904
Protein assay kit Bio-Rad Cat#5000111
DNA-free™ DNA Removal kit Invitrogen Cat#AM1906
PrimeScript™ RT reagent Kit Takara Cat#RR037A
FUJI DRI-CHEM SLIDE GOT/AST-PIII Fujifilm Cat#4902520-77484-2
FUJI DRI-CHEM SLIDE GPT/ALT-PIII Fujifilm Cat#4902520-77485-9
FUJI DRI-CHEM SLIDE CRE-PIII Fujifilm Cat#4902520-77481-1
FUJI DRI-CHEM SLIDE BUN-PIII Fujifilm Cat#4902520-89062-7

Deposited data

TCGA RNA –Seq This paper Mendeley Data (https://data.mendeley.com/datasets/xfh84npb2h/2) with the database https://doi.org/10.17632/xfh84npb2h.2.

Experimental models: Cell lines

MDA-MB231 ATCC HTB-26
MIA PaCa2 RIKEN RCB2094
PANC1 RIKEN RCB2095
DLD1 ATCC CCL-221
A375 ATCC CRL-1619
Bowes ATCC CRL-9607
A2058 JCRB IFO50276

Experimental models: Organisms/strains

Mouse: Balb/c-nu/nu Japan SLC N/A

Oligonucleotides

Q-PCR prime: human GRM1 Applied Biosystems Hs00168250_m1
Q-PCR prime: human 53BP1 Applied Biosystems Hs00996827_m1
Q-PCR prime: human CDKN1A Applied Biosystems Hs00355782_m1
Q-PCR prime: human IL-6 Applied Biosystems Hs00174131_m1
Q-PCR prime: human CXCL8 Applied Biosystems Hs00174103_m1
Q-PCR prime: human TNF-α Applied Biosystems Hs00174128_m1
Q-PCR prime: human VEGFA Applied Biosystems Hs00900055_m1
Q-PCR prime: human MET Applied Biosystems Hs01565584_m1
Q-PCR prime:18S ribosomal RNA Applied Biosystems Hs99999901_s1

Software and algorithms

GraphPad Prism 8 GraphPad Software https://www.graphpad.com/scientific-software/prism/
Molecular Operating Environment Chemical Computing Group https://www.chemcomp.com/index.htm
Visual Molecular Dynamics University of Illinois https://www.ks.uiuc.edu/Research/vmd/
AMBER18 HPC Systems https://www.hpc.co.jp/chem/software/amber/
PyMol Schrödinger https://pymol.org/2/
Specialized Hybrid Cell Count software Keyence https://www.keyence.co.jp/
Medical internal radiation dose formula Stabin40 https://jnm.snmjournals.org/content/37/3/538.long

Resource availability

Lead contact

Further information and requests for resources and reagents should be directed to and will be fulfilled by the lead contact, Dr. Ming-Rong Zhang (zhang.ming-rong@qst.go.jp).

Materials availability

This study did not generate new unique reagents. The radiopharmaceutical in this study would be available from the lead contact without restriction.

Experimental model and subject details

Cell lines

The following cell lines were used in this study: MDA-MB231 human TNBCs (ATCC, Manassas, VA, USA), MIA PaCa2 (RIKEN, Tsukuba, Japan) and PANC1 (RIKEN) human pancreatic cancers, DLD1 human colon cancers (ATCC), A375 (ATCC), Bowes (ATCC), and A2058 (Japanese Collection of Research Bioresources, Osaka, Japan) human melanomas. MDA-MB231, MIA PaCa-2, A375, and A2058 cells were maintained and passaged in Dulbecco’s modified Eagle’s medium (DMEM; Sigma-Aldrich, St. Louis, MO, USA) supplemented with 10% fetal bovine serum (FBS; Sigma-Aldrich) and 1% penicillin/streptomycin (50 U/mL; Gibco, Grand Island, NY, USA). Bowes cells were maintained and passaged in minimum Eagle’s medium (MEM; Sigma-Aldrich) supplemented with 10% FBS and 1% penicillin/streptomycin. PANC1 and DLD1 cells were maintained and passaged in RPMI-1640 medium (Sigma-Aldrich) supplemented with 10% FBS and 1% penicillin/streptomycin. All cells were cultured at 37°C in a humidified atmosphere with 5% CO2 and were regularly tested to ensure the absence of mycoplasma contamination.

Mice and mouse models

Animal experiments were performed on 6−8-week-old male and female Balb/c-nu/nu mice (body weight, 21.18 ± 0.25 g) (Japan SLC, Shizuoka, Japan). The xenograft tumor models were created by injecting a single-cell suspension of human cancer cells (5×106) into the left flanks of mice with Matrigel (Corning, Teterboro, NJ, USA). All animal experiments were approved by the Animal Ethics Committee of National Institutes for Quantum Science and Technology (QST). Animals were maintained and handled under a SPF condition (12-h light/dark cycle, 50% relative humidity, between 25°C and 27°C) with free access to food and tap water, in accordance with the recommendations of the National Institute of Health and the institutional guidelines of QST.

Method details

Study design

The major objective of this study was to evaluate the therapeutic efficacy of a mGluR1-based RPT strategy using a small-molecule α-emitting radiopharmaceutical (211At-AITM) against multiple human cancers and to define the mechanisms underlying the antitumor effects of 211At-AITM. The binding and specific studies of 211At-AITM for mGluR1 were carried out using parallel control experiments including comparative experiments in multiple mGluR1+ and mGluR1 human cancer cells and competition experiments with unlabeled mGluR1-competitive antagonist FITM. In vitro effects of 211At-AITM on mGluR1-expressing cancer cells were tested using two mGluR1+ human cancer cells treated with the testing α-radiopharmaceutical (211At-AITM) or vehicle (PBS). End points for cell morphology, viability and phenotypic analyses were selected on the basis of a time course ranging from 1 to 7 days after addition of 211At-AITM to cells in culture. All cellular experiments were conducted in triplicates (technical replicates) and repeated independently at least three times unless otherwise noted.

In vivo studies were performed to evaluate the effects of 211At-AITM in xenograft mice bearing 7 subtypes from 4 solid tumors with different tissue origins and distinct mGluR1 expression levels, as well as its pharmacokinetics, radiation dosimetry, biological effective dose, safety, and phenotypic changes of cancer tissues after intravenous injection of 211At-AITM. Xenotransplantation studies were carried out using more than 4 animals per group unless otherwise specified. Animals were transplanted by injecting a single-cell suspension of human cancer cells (5×106) into the left flanks of mice and randomly assigned to experimental groups as defined in the Figure legends. All groups were dosed with a single intravenous administration of 211At-AITM, FITM, or vehicle (saline). The end point of these in vivo therapeutic experiments was set at 30 days after injection and body weight loss of more than 20%. Exclusion criteria included unspecified death. All experiments were carried out as unblinded studies.

GRM1 expression data in human cancer tissues

To analyze GRM1 expression in human cancer samples, data were downloaded from cBioPortal (www.cbioportal.org/), and raw sequencing data from TCGA were imported (https://cancergenome.nih.gov/). All available GRM1 mRNA data as of March 2022 were downloaded and transformed into a plot using GraphPad Prism 8 (GraphPad Software, San Diego, CA, USA). Patient samples covering 10,967 across 32 tumor types were analyzed and grouped into two subpopulations according to the expression status of GRM1 (GRM1+ cancers and GRM1- cancers).

Molecular docking study

The three-dimension FITM-mGluR1 complex structure (PDB ID: 4OR2) was used for molecular modeling. The initial structure of AITM complex was generated using MOE (Molecular Operating Environment; Chemical Computing Group, Montreal, QC, Canada). The FITM and AITM complexes were added by lipid bilayer and water molecules around the complexes using VMD (Visual Molecular Dynamics). For the FITM complex, the energy minimized (MM) calculations and molecular dynamics (MD) simulations (5 ns) were performed using AMBER18/SANDER. The conditions of MD simulations were constant temperature (300 K) and pressure (1 atm). On the other hand, the MM calculations were performed for the AITM complex using MOE. The final structures of FITM or AITM complexes were visualized using PyMol.

Radiosynthesis of 211At-AITM

211At-AITM was synthesized by reacting a stannyl precursor (100 μg) with 211At in the presence of an oxidizing agent. 211At-AITM was prepared as an injectable solution according to our previously reported method.33 211At-AITM was obtained with 36–118 MBq in 45.7 ± 6.5% radiochemical yield (n = 20, no decay-corrected, based on the total radionuclides used) with > 99% radiochemical purity, which was used for all experiments.

Cell binding assay

Human cancer cells (2 × 105 cells) were seeded into 24-well plates and allowed to adhere though incubation overnight for all further experiments. Cells were incubated in triplicate with 18.5 kBq/mL 211At-AITM for 1, 3, and 21 h at 37°C. Excess unlabeled mGluR1-competitive antagonist FITM (10 μmol/L; MedChemExpress, Monmouth Junction, NJ, USA) was used to determine nonspecific binding.14 The percentage of internalization and membrane-bound 211At-AITM was determined using a previously published method.38 Briefly, MAD-MB231 and MIA PaCa2 cells, after incubation with 211At-AITM, were washed with medium and treated with a mild acid buffer (50 mmol/L glycine +150 mmol/L NaCl, pH 2.8) at 4°C for 5 min. The buffer was collected, and cells were further washed with the same amount of buffer. The pooled washes were considered to represent membrane-bound radioactivity, while the unreleased radioactivity was considered internalized and was collected by treating the cells with 0.2 mol/L NaOH. Protein quantification was performed using a protein assay kit (Bio-Rad, Hercules, CA, USA), while radioactivity uptake was assessed using a γ-counter (PerkinElmer, Waltham, MA, USA). Cellular radioactivity uptake was calculated as a percentage of the incubated radioactivity dose normalized per mg of protein (% ICD/mg protein). The ratio between internalized and membrane-bound radioactivity was expressed as a percentage of cell-associated radioactivity dose (% CD).

Immunohistochemistry

To investigate 211At-AITM-induced damage events, MDA-MB231 (2 × 105 cells) and MIA PaCa2 cells (2 × 105 cells) were exposed to 18.5 kBq/mL 211At-AITM or vehicle (PBS) for 21 h in triplicate in 24-well plates. Alterations in cell morphology, viability, and cell cycle distribution were subsequently analyzed until 7 days or otherwise indicated. Animals were anesthetized with isoflurane and then sacrificed. Tissue samples of xenograft tumors were harvested and frozen at 2, 7, and 30 days after 211At-AITM injection. Tumor sections (5 μm) were prepared using a Microm HM560 microtome (Carl Zeiss Jena, Germany).

mGluR1, Ki67 and CD31 staining

For cell immunohistochemistry, cancer cells after the indicated treatment were fixed with 4% paraformaldehyde (PFA) solution, incubated overnight at 4°C with primary rabbit anti-human mGluR1 antibody (1:200; LS-A882, LSBio, Seattle, WA, USA), incubated with secondary Alexa Fluor® 488 goat anti-rabbit IgG antibodies (1:500; A-11008, Invitrogen, Carlsbad, CA, USA) for 1 h at room temperature (RT), and mounted using mounting medium with DAPI (Vector Laboratories, Burlingame, CA, USA). For tissue immunohistochemistry, the tumor sections treated as indicated were subjected to immunofluorescence staining with rabbit anti-human mGluR1 antibody (1:200), mouse anti-human Ki67 antibody (1:50; 550609, BD Pharmingen, San Diego, CA, USA), or rat anti-mouse CD31 antibody (1:100; 550274, BD Pharmingen), after which they were incubated with Alexa Fluor® 488 goat anti-rabbit IgG (1:500; Invitrogen), Alexa Fluor® 546 goat anti-mouse IgG (1:500; A-11003, Invitrogen), or Alexa Fluor® 488 goat anti-rat IgG (1:500; A-11006, Invitrogen) and mounted using mounting medium with DAPI.

γH2AX staining

At the indicated time points after 211At-AITM treatment, cells were fixed with 4% PFA and used as a substrate for a γH2AX DNA Damage Detection Kit (G266, Dojindo, Kumamoto, Japan) according to the manufacturer’s instructions. Briefly, cells were washed twice with PBS and permeabilized for 30 min in 1% Triton X-100 at RT. Next, cells were incubated overnight at 4°C with primary mouse anti-γH2AX (1:50), incubated with goat anti-mouse-Red (1:50) secondary antibody for 1 h at RT, and mounted using mounting medium with DAPI.

Senescence-associated β-galactosidase assay

Commercially available senescence-associated β-galactosidase (SA-β-gal) staining kits (SG02, Dojindo) were used to detect senescence in cells and tissues as instructed by the manufacturer. Briefly, tumor cells and tissue sections were fixed with 4% PFA, washed with PBS and stained for 30 min with fresh SA-β-gal solution (10 μmol/L, pH 6.0) at 37°C in an incubator without CO2. Samples were mounted using mounting medium with DAPI.

Immunohistochemistry images were acquired using a Keyence BZ-X710 microscope (Keyence, Osaka, Japan). The fluorescence intensity, frequency and area of positively stained cells were measured automatically by using specialized Hybrid Cell Count software (Keyence). The results were expressed as the fluorescence intensity per cell and the percentage of total cells or tissue areas that exhibited positive staining. Negative control slides were processed in the absence of primary antibody, secondary antibody, or isotype control IgG to ensure specificity. For comparisons between images from the same experiment, all parameters were adjusted equally, and the ratio between the parameters was not altered.

Cell viability assay

A Muse® Cell Analyzer (Luminex, Austin, TX, USA) was used to determine the viability of tumor cells after 211At-AITM treatment using the Muse® Count & Viability Kit (MCH100102, Luminex) as instructed by the manufacturer. Briefly, at the indicated time points after 211At-AITM or vehicle (PBS) treatment, MDA-MB231 and MIA PaCa2 cells were harvested in triplicate as single-cell suspensions through digestion with Gibco trypsin-EDTA (0.05%) at 37°C for 5 min. After the incubation, cells were washed and mixed in fresh PBS. Next, 20 μL of this cell suspension was mixed with 380 μL of Count & Viability reagent. The suspension was incubated for 5 min at RT and thereafter examined for cell count and viability using the Muse® Cell Analyzer.

Cell cycle assay

The effect of 211At-AITM on the human tumor cell cycle was determined using a Muse® Cell Analyzer and a Muse® Cell Cycle kit (MCH100106, Luminex) according to the manufacturer’s instruction. Briefly, at the indicated time points after 211At-AITM or vehicle (PBS) treatment, MDA-MB231 and MIA PaCa2 cells were harvested in triplicate as single-cell suspensions and washed with fresh PBS, followed by spinning at 3000 rpm for 5 min. The cell pellet was fixed in 1 mL of 70% ice-cold ethanol and stored at −20°C until used. Next, 200 μL of fixed cells was aliquoted, centrifuged at 3000 rpm for 5 min, and washed with PBS solution. To the fixed cells, 200 μL of Muse® Cell Cycle reagent was mixed and incubated for 30 min in the dark at RT. Samples were examined using the Muse® Cell Analyzer.

In vivo performance, radiation dosimetry and biological effective dose

The in vivo effect of 211At-AITM was calculated using biodistribution data in corresponding tumor-bearing mice. A therapeutic amount of 211At-AITM (2.96 MBq/100 μL) was administered intravenously to mice, which were subsequently sacrificed by cervical dislocation at 1, 3, and 21 h post-injection. The tumor and major organs were promptly excised, harvested, as well as weighed. The radioactivity was measured using a 2470 Wizard automatic γ-counter (PerkinElmer). A time profile of the radioactivity in each tumor and tissue was generated by normalizing the decay-corrected radioactivity time to a percentage of the injected dose per gram of wet tissue (%ID/g). The dosimetry of 211At-AITM absorbed by each organ and tumor was estimated in accordance with the standard method using the medical internal radiation dose formula,40 and the results are expressed as the mean dose per unit of injected activity (gray per MBq; Gy/MBq). The biological effective dose was calculated by using a quality factor of 5, expressed as sievert per MBq (Sv/MBq).41,42

Therapeutic study of 211At-AITM in xenograft-bearing mice

Radiotherapy experiments were performed on 6−8-week-old male and female Balb/c-nu/nu mice (body weight, 21.18 ± 0.25 g) bearing 7 different human tumor cell types (MDA-MB231, MIA PaCa2, PANC1, A375, Bowes, A2058, and DLD1). Tumor-bearing mice with a tumor volume of ∼50 mm3 (approximately 1 week after tumor inoculation) were randomly assigned to treatment and vehicle groups. The therapeutic efficacy was evaluated with a single intravenous administration of 2.96 MBq of 211At-AITM, 1 mg/kg of FITM or vehicle (saline) in 100 μL to each tumor-bearing mouse. Tumor dimensions were measured twice weekly with digital calipers in a blinded fashion, and tumor volumes were calculated using the following formula: (width2 × length)/2. The tumor xenografts were examined at 2, 7, and 30 days after 211At-AITM injection by immunohistochemistry and gene analysis.

Safety assessment

Changes in the body weight of mice were evaluated as an indicator of the radiation-related side effects of 211At-AITM. The hematology and liver and kidney chemistry were evaluated in human cancer-bearing mice administered 2.96 MBq of 211At-AITM or vehicle (saline). Hematological analyses included leukocyte, erythrocyte and platelet counts were performed using a Celltack F Automated Hematology Analyzer (Nihon Kohden, Tokyo, Japan). Liver and kidney analyses included determinations of glutamic oxaloacetic transaminase (GOT), glutamic pyruvic transaminase (GPT) values, creatinine and blood urea nitrogen (BUN) values, which were conducted using Japan Society of Clinical Chemistry reference methods according to the manufacturer’s instructions. Histological analysis of the major organs (liver, kidney, lung, spleen, and stomach) was performed. The samples were collected from the xenograft mice treated with 211At-AITM or vehicle (saline) at 30 days post-injection, fixed with 10% PFA and embedded in paraffin blocks. Tissue sections (5 μm) were stained with hematoxylin-eosin staining, investigated using a Keyence BZ-X710 microscope.

RNA preparation and qRT-PCR analysis

Total RNA was extracted from tumor cells using the RNeasy Mini Kit (Qiagen, Valencia, CA, USA) and from frozen tumor tissues using Sepasol-RNA I Super (Nacalai Tesque, Kyoto, Japan) according to the manufacturer’s protocol and subsequently purified with the DNA-free™ DNA Removal kit (Ambion, Austin, TX, USA). The quality of the total RNA was verified using the 260/280 nm ratio and a NanoDrop 2000 (Thermo Fisher Scientific, Wilmington, DE, USA). qRT-PCR was performed using a TaqMan system on an Applied Biosystems StepOne™ machine (Carlsbad, CA, USA) according to the manufacturer’s instruction. Target-specific primers and probes for human GRM1 (Hs00168250_m1), 53BP1 (Hs00996827_m1), CDKN1A (Hs00355782_m1), IL-6 (Hs00174131_m1), CXCL8 (Hs00174103_m1), TNF-α (Hs00174128_m1), VEGFA (Hs00900055_m1), MET (Hs01565584_m1) and 18S ribosomal RNA (18S rRNA, Hs99999901_s1) were purchased from Applied Biosystems. The normalized Ct value of each gene was obtained by subtracting the Ct value for 18S rRNA.

Quantification and statistical analysis

Quantitative data are presented as mean ± standard error of the mean (SEM). Intergroup comparisons were performed using an unpaired two-tailed Student’s t-test or two-way analysis of variance (ANOVA) followed by Tukey’s multiple comparison test in GraphPad Prism8. Pearson’s correlation analysis was used to estimate the relationship between tumor mGluR1 expression and cytotoxicity, 211At-AITM uptake, or therapeutic efficacy in vitro and in vivo. The threshold for statistical significance was set at P < 0.05.

Acknowledgments

We would like to thank the staff of National Institute for Quantum Science and Technology for their technical support in the radiosynthesis and animal experiments. We would like to thank Tomomi Kokufuta for her excellent technical support. We would like to thank Dr. Atsushi B. Tsuji and Dr. Zhao-Hui Jin for helpful scientific and technical discussion. This work was supported in part by the AMED Moonshot Research and Development Program Grant no. 21zf0127003h001 to M.-R.Z., JSPS KAKENHI Grant no. 20H03635 to M.-R.Z. and 21K07659 to L.X, JSPS International Joint Research Program Grant no. JPJSBP120207203 to M.-R.Z., the initiative for realizing diversity in the research environment to L.X., and the Directorate’s Fund Project given by Dr. Takashi Nakano to L.X.

Author contributions

L.X., L.Z. K.H., and M.-R.Z. conceptualized the study. L.X., L.Z., and K.H. contributed to the methodology. L.X., L.Z., K.H., Y.Z., T.O., K.N., C.J., T.S., K.A., F.W. M.H., M.F., K.M., and M.-R.Z. carried out experimental investigation. L.X. and M.-R.Z. contributed to project administration. L.X., L.Z., K.H., N.O., S.Y., R.W., and M.-R.Z. performed data analysis and wrote the original draft. All authors were involved with reviewing and editing the final draft.

Declaration of interests

The authors declare no competing interests.

Published: March 31, 2023

Footnotes

Supplemental information can be found online at https://doi.org/10.1016/j.xcrm.2023.100960.

Contributor Information

Lin Xie, Email: xie.lin@qst.go.jp.

Ming-Rong Zhang, Email: zhang.ming-rong@qst.go.jp.

Supplemental Information

Document S1. Figures S1–S7
mmc1.pdf (3.2MB, pdf)
Document S2. Article plus supplemental information
mmc2.pdf (11MB, pdf)

Data and code availability

All data generated from this study are included in the paper and the supplementary information. The RNA-seq data downloaded from TCGA have been deposited to Mendeley Data (https://data.mendeley.com/datasets/xfh84npb2h/2) with the database https://doi.org/10.17632/xfh84npb2h.2. This paper does not report original code. Any additional information required to reanalyze the data reported in this paper is available from the lead contact upon request.

References

  • 1.Hanahan D. Hallmarks of cancer: new dimensions. Cancer Discov. 2022;12:31–46. doi: 10.1158/2159-8290.Cd-21-1059. [DOI] [PubMed] [Google Scholar]
  • 2.DeBerardinis R.J., Lum J.J., Hatzivassiliou G., Thompson C.B. The biology of cancer: metabolic reprogramming fuels cell growth and proliferation. Cell Metabol. 2008;7:11–20. doi: 10.1016/j.cmet.2007.10.002. [DOI] [PubMed] [Google Scholar]
  • 3.Jones R.G., Thompson C.B. Tumor suppressors and cell metabolism: a recipe for cancer growth. Genes Dev. 2009;23:537–548. doi: 10.1101/gad.1756509. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Ye T., Lin L., Cao L., Huang W., Wei S., Shan Y., Zhang Z. Novel prognostic signatures of hepatocellular carcinoma based on metabolic pathway phenotypes. Front. Oncol. 2022;12:863266. doi: 10.3389/fonc.2022.863266. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Xu H., Liu Z., Gao D., Li P., Shen Y., Sun Y., Xu L., Song N., Wang Y., Zhan M., et al. Reprogramming hormone-sensitive prostate cancer to a lethal neuroendocrine cancer lineage by mitochondrial pyruvate carrier (MPC) Mol. Metabol. 2022;59:101466. doi: 10.1016/j.molmet.2022.101466. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Pantel A.R., Ackerman D., Lee S.C., Mankoff D.A., Gade T.P. Imaging cancer metabolism: underlying biology and emerging strategies. J. Nucl. Med. 2018;59:1340–1349. doi: 10.2967/jnumed.117.199869. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Yonekura Y., Benua R.S., Brill A.B., Som P., Yeh S.D., Kemeny N.E., Fowler J.S., MacGregor R.R., Stamm R., Christman D.R., Wolf A.P. Increased accumulation of 2-deoxy-2-[18F]Fluoro-D-glucose in liver metastases from colon carcinoma. J. Nucl. Med. 1982;23:1133–1137. [PubMed] [Google Scholar]
  • 8.Som P., Atkins H.L., Bandoypadhyay D., Fowler J.S., MacGregor R.R., Matsui K., Oster Z.H., Sacker D.F., Shiue C.Y., Turner H., et al. A fluorinated glucose analog, 2-fluoro-2-deoxy-D-glucose (F-18): nontoxic tracer for rapid tumor detection. J. Nucl. Med. 1980;21:670–675. [PubMed] [Google Scholar]
  • 9.Hensley C.T., Wasti A.T., DeBerardinis R.J. Glutamine and cancer: cell biology, physiology, and clinical opportunities. J. Clin. Invest. 2013;123:3678–3684. doi: 10.1172/jci69600. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Newsholme P., Procopio J., Lima M.M.R., Pithon-Curi T.C., Curi R. Glutamine and glutamate--their central role in cell metabolism and function. Cell Biochem. Funct. 2003;21:1–9. doi: 10.1002/cbf.1003. [DOI] [PubMed] [Google Scholar]
  • 11.Pollock P.M., Cohen-Solal K., Sood R., Namkoong J., Martino J.J., Koganti A., Zhu H., Robbins C., Makalowska I., Shin S.S., et al. Melanoma mouse model implicates metabotropic glutamate signaling in melanocytic neoplasia. Nat. Genet. 2003;34:108–112. doi: 10.1038/ng1148. [DOI] [PubMed] [Google Scholar]
  • 12.Prickett T.D., Samuels Y. Molecular pathways: dysregulated glutamatergic signaling pathways in cancer. Clin. Cancer Res. 2012;18:4240–4246. doi: 10.1158/1078-0432.Ccr-11-1217. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Choi K.Y., Chang K., Pickel J.M., Badger J.D., Roche K.W., Roche K.W. Expression of the metabotropic glutamate receptor 5 (mGluR5) induces melanoma in transgenic mice. Proc. Natl. Acad. Sci. USA. 2011;108:15219–15224. doi: 10.1073/pnas.1107304108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Wu H., Wang C., Gregory K.J., Han G.W., Cho H.P., Xia Y., Niswender C.M., Katritch V., Meiler J., Cherezov V., et al. Structure of a class C GPCR metabotropic glutamate receptor 1 bound to an allosteric modulator. Science. 2014;344:58–64. doi: 10.1126/science.1249489. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Speyer C.L., Smith J.S., Banda M., DeVries J.A., Mekani T., Gorski D.H. Metabotropic glutamate receptor-1: a potential therapeutic target for the treatment of breast cancer. Breast Cancer Res. Treat. 2012;132:565–573. doi: 10.1007/s10549-011-1624-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Gómez V., Galazi M., Weitsman G., Monypenny J., Al-Salemee F., Barber P.R., Ng K., Beatson R., Szokol B., Őrfi L., et al. HER2 mediates PSMA/mGluR1-driven resistance to the DS-7423 dual PI3K/mTOR inhibitor in PTEN wild-type prostate cancer models. Mol. Cancer Therapeut. 2022;21:667–676. doi: 10.1158/1535-7163.Mct-21-0320. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Nord K.H., Lilljebjörn H., Vezzi F., Nilsson J., Magnusson L., Tayebwa J., de Jong D., Bovée J.V.M.G., Hogendoorn P.C.W., Szuhai K. GRM1 is upregulated through gene fusion and promoter swapping in chondromyxoid fibroma. Nat. Genet. 2014;46:474–477. doi: 10.1038/ng.2927. [DOI] [PubMed] [Google Scholar]
  • 18.Xia H., Zhao Y.N., Yu C.H., Zhao Y.L., Liu Y. Inhibition of metabotropic glutamate receptor 1 suppresses tumor growth and angiogenesis in experimental non-small cell lung cancer. Eur. J. Pharmacol. 2016;783:103–111. doi: 10.1016/j.ejphar.2016.04.053. [DOI] [PubMed] [Google Scholar]
  • 19.Niswender C.M., Conn P.J. Metabotropic glutamate receptors: physiology, pharmacology, and disease. Annu. Rev. Pharmacol. Toxicol. 2010;50:295–322. doi: 10.1146/annurev.pharmtox.011008.145533. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Shah R., Singh S.J., Eddy K., Filipp F.V., Chen S. Concurrent targeting of glutaminolysis and metabotropic glutamate receptor 1 (GRM1) reduces glutamate bioavailability in GRM1(+) melanoma. Cancer Res. 2019;79:1799–1809. doi: 10.1158/0008-5472.Can-18-1500. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Wen Y., Li J., Koo J., Shin S.S., Lin Y., Jeong B.S., Mehnert J.M., Chen S., Cohen-Sola K.A., Goydos J.S. Activation of the glutamate receptor GRM1 enhances angiogenic signaling to drive melanoma progression. Cancer Res. 2014;74:2499–2509. doi: 10.1158/0008-5472.Can-13-1531. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Mehnert J.M., Silk A.W., Lee J.H., Dudek L., Jeong B.S., Li J., Schenkel J.M., Sadimin E., Kane M., Lin H., et al. A phase II trial of riluzole, an antagonist of metabotropic glutamate receptor 1 (GRM1) signaling, in patients with advanced melanoma. Pigment Cell Melanoma Res. 2018;31:534–540. doi: 10.1111/pcmr.12694. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Teh J.L.F., Shah R., La Cava S., Dolfi S.C., Mehta M.S., Kongara S., Price S., Ganesan S., Reuhl K.R., Hirshfield K.M., et al. Metabotropic glutamate receptor 1 disrupts mammary acinar architecture and initiates malignant transformation of mammary epithelial cells. Breast Cancer Res. Treat. 2015;151:57–73. doi: 10.1007/s10549-015-3365-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Yip D., Le M.N., Chan J.L.K., Lee J.H., Mehnert J.A., Yudd A., Kempf J., Shih W.J., Chen S., Goydos J.S. A phase 0 trial of riluzole in patients with resectable stage III and IV melanoma. Clin. Cancer Res. 2009;15:3896–3902. doi: 10.1158/1078-0432.Ccr-08-3303. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Silk A.W., Saraiya B., Groisberg R., Chan N., Spencer K., Girda E., Shih W., Palmeri M., Saunders T., Berman R.M., et al. A phase Ib dose-escalation study of troriluzole (BHV-4157), an oral glutamatergic signaling modulator, in combination with nivolumab in patients with advanced solid tumors. Eur. J. Med. Res. 2022;27:107. doi: 10.1186/s40001-022-00732-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Le M.N., Chan J.L.K., Rosenberg S.A., Nabatian A.S., Merrigan K.T., Cohen-Solal K.A., Goydos J.S. The glutamate release inhibitor Riluzole decreases migration, invasion, and proliferation of melanoma cells. J. Invest. Dermatol. 2010;130:2240–2249. doi: 10.1038/jid.2010.126. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Marín Y.E., Namkoong J., Cohen-Solal K., Shin S.S., Martino J.J., Oka M., Chen S. Stimulation of oncogenic metabotropic glutamate receptor 1 in melanoma cells activates ERK1/2 via PKCepsilon. Cell. Signal. 2006;18:1279–1286. doi: 10.1016/j.cellsig.2005.10.012. [DOI] [PubMed] [Google Scholar]
  • 28.Wall B.A., Yu L.J., Khan A., Haffty B., Goydos J.S., Chen S. Riluzole is a radio-sensitizing agent in an in vivo model of brain metastasis derived from GRM1 expressing human melanoma cells. Pigment Cell Melanoma Res. 2015;28:105–109. doi: 10.1111/pcmr.12327. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Prokopi A., Tripp C.H., Tummers B., Hornsteiner F., Spoeck S., Crawford J.C., Clements D.R., Efremova M., Hutter K., Bellmann L., et al. Skin dendritic cells in melanoma are key for successful checkpoint blockade therapy. J. Immunother. Cancer. 2021;9:e000832. doi: 10.1136/jitc-2020-000832. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Zacherl M.J., Gildehaus F.J., Mittlmeier L., Böning G., Gosewisch A., Wenter V., Unterrainer M., Schmidt-Hegemann N., Belka C., Kretschmer A., et al. First clinical results for PSMA-targeted α-therapy using (225)Ac-PSMA-I&T in advanced-mCRPC patients. J. Nucl. Med. 2021;62:669–674. doi: 10.2967/jnumed.120.251017. [DOI] [PubMed] [Google Scholar]
  • 31.Batra V., Samanta M., Makvandi M., Groff D., Martorano P., Elias J., Ranieri P., Tsang M., Hou C., Li Y., et al. Preclinical development of [211At]meta-astatobenzylguanidine ([211At]MABG) as an alpha particle radiopharmaceutical therapy for neuroblastoma. Clin. Cancer Res. 2022;28:4146–4157. doi: 10.1158/1078-0432.Ccr-22-0400. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Parker C., Nilsson S., Heinrich D., Helle S.I., O'Sullivan J.M., Fosså S.D., Chodacki A., Wiechno P., Logue J., Seke M., et al. Alpha emitter radium-223 and survival in metastatic prostate cancer. N. Engl. J. Med. 2013;369:213–223. doi: 10.1056/NEJMoa1213755. [DOI] [PubMed] [Google Scholar]
  • 33.Xie L., Hanyu M., Fujinaga M., Zhang Y., Hu K., Minegishi K., Jiang C., Kurosawa F., Morokoshi Y., Li H.K., et al. (131)I-IITM and (211)At-AITM: two novel small-molecule radiopharmaceuticals targeting oncoprotein metabotropic glutamate receptor 1. J. Nucl. Med. 2020;61:242–248. doi: 10.2967/jnumed.119.230946. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Satoh A., Nagatomi Y., Hirata Y., Ito S., Suzuki G., Kimura T., Maehara S., Hikichi H., Satow A., Hata M., et al. Discovery and in vitro and in vivo profiles of 4-fluoro-N-[4-[6-(isopropylamino)pyrimidin-4-yl]-1,3-thiazol-2-yl]-N-methylbenzamide as novel class of an orally active metabotropic glutamate receptor 1 (mGluR1) antagonist. Bioorg. Med. Chem. Lett. 2009;19:5464–5468. doi: 10.1016/j.bmcl.2009.07.097. [DOI] [PubMed] [Google Scholar]
  • 35.Pike V.W. PET radiotracers: crossing the blood-brain barrier and surviving metabolism. Trends Pharmacol. Sci. 2009;30:431–440. doi: 10.1016/j.tips.2009.05.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Waterhouse R.N. Determination of lipophilicity and its use as a predictor of blood-brain barrier penetration of molecular imaging agents. Mol. Imag. Biol. 2003;5:376–389. doi: 10.1016/j.mibio.2003.09.014. [DOI] [PubMed] [Google Scholar]
  • 37.Fujinaga M., Xie L., Yamasaki T., Yui J., Shimoda Y., Hatori A., Kumata K., Zhang Y., Nengaki N., Kawamura K., Zhang M.R. Synthesis and evaluation of 4-halogeno-N-[4-[6-(isopropylamino)pyrimidin-4-yl]-1,3-thiazol-2-yl]-N-[11C]methylbenzamide for imaging of metabotropic glutamate 1 receptor in melanoma. J. Med. Chem. 2015;58:1513–1523. doi: 10.1021/jm501845n. [DOI] [PubMed] [Google Scholar]
  • 38.Joyal J.L., Barrett J.A., Marquis J.C., Chen J., Hillier S.M., Maresca K.P., Boyd M., Gage K., Nimmagadda S., Kronauge J.F., et al. Preclinical evaluation of an 131I-labeled benzamide for targeted radiotherapy of metastatic melanoma. Cancer Res. 2010;70:4045–4053. doi: 10.1158/0008-5472.can-09-4414. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Vakifahmetoglu H., Olsson M., Zhivotovsky B. Death through a tragedy: mitotic catastrophe. Cell Death Differ. 2008;15:1153–1162. doi: 10.1038/cdd.2008.47. [DOI] [PubMed] [Google Scholar]
  • 40.Stabin M.G. MIRDOSE: personal computer software for internal dose assessment in nuclear medicine. J. Nucl. Med. 1996;37:538–546. [PubMed] [Google Scholar]
  • 41.Sudo H., Tsuji A.B., Sugyo A., Harada Y., Nagayama S., Katagiri T., Nakamura Y., Higashi T. FZD10-targeted α-radioimmunotherapy with (225) Ac-labeled OTSA101 achieves complete remission in a synovial sarcoma model. Cancer Sci. 2022;113:721–732. doi: 10.1111/cas.15235. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Sgouros G., Roeske J.C., McDevitt M.R., Palm S., Allen B.J., Fisher D.R., Brill A.B., Song H., Howell R.W., Akabani G., et al. MIRD Pamphlet No. 22 (abridged): radiobiology and dosimetry of alpha-particle emitters for targeted radionuclide therapy. J. Nucl. Med. 2010;51:311–328. doi: 10.2967/jnumed.108.058651. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Spetz J., Rudqvist N., Forssell-Aronsson E. Biodistribution and dosimetry of free 211At, 125I- and 131I- in rats. Cancer Biother. Radiopharm. 2013;28:657–664. doi: 10.1089/cbr.2013.1483. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Yu L.J., Wall B.A., Wangari-Talbot J., Chen S. Metabotropic glutamate receptors in cancer. Neuropharmacology. 2017;115:193–202. doi: 10.1016/j.neuropharm.2016.02.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Cuddihy A.R., Bristow R.G. The p53 protein family and radiation sensitivity: yes or no? Cancer Metastasis Rev. 2004;23:237–257. doi: 10.1023/B:CANC.0000031764.81141.e4. [DOI] [PubMed] [Google Scholar]
  • 46.Emami B. Tolerance of normal tissue to therapeutic irradiation. Rep. Radiother. Oncol. 2013;1:123–127. doi: 10.1016/0360-3016(91)90171-y. [DOI] [PubMed] [Google Scholar]
  • 47.Herranz N., Gallage S., Mellone M., Wuestefeld T., Klotz S., Hanley C.J., Raguz S., Acosta J.C., Innes A.J., Banito A., et al. mTOR regulates MAPKAPK2 translation to control the senescence-associated secretory phenotype. Nat. Cell Biol. 2015;17:1205–1217. doi: 10.1038/ncb3225. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Jin Z.-H., Tsuji A.B., Degardin M., Sugyo A., Obara S., Wakizaka H., Nagatsu K., Hu K., Zhang M.-R., Dumy P., et al. Radiotheranostic agent 64Cu-cyclam-RAFT-c(-RGDfK-)4 for management of peritoneal metastasis in ovarian cancer. Clin. Cancer Res. 2020;26:6230–6241. doi: 10.1158/1078-0432.Ccr-20-1205. [DOI] [PubMed] [Google Scholar]
  • 49.Sgouros G. α-Particle-Emitter radiopharmaceutical therapy: resistance is futile. Cancer Res. 2019;79:5479–5481. doi: 10.1158/0008-5472.can-19-2806. [DOI] [PubMed] [Google Scholar]
  • 50.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. doi: 10.1038/s41573-020-0073-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Xie L., Yui J., Fujinaga M., Hatori A., Yamasaki T., Kumata K., Wakizaka H., Furutsuka K., Takei M., Jin Z.H., et al. Molecular imaging of ectopic metabotropic glutamate 1 receptor in melanoma with a positron emission tomography radioprobe (18) F-FITM. Int. J. Cancer. 2014;135:1852–1859. doi: 10.1002/ijc.28842. [DOI] [PubMed] [Google Scholar]
  • 52.Zalutsky M.R., Pozzi O.R. Radioimmunotherapy with alpha-particle emitting radionuclides. Q. J. Nucl. Med. Mol. Imaging. 2004;48:289–296. [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Document S1. Figures S1–S7
mmc1.pdf (3.2MB, pdf)
Document S2. Article plus supplemental information
mmc2.pdf (11MB, pdf)

Data Availability Statement

All data generated from this study are included in the paper and the supplementary information. The RNA-seq data downloaded from TCGA have been deposited to Mendeley Data (https://data.mendeley.com/datasets/xfh84npb2h/2) with the database https://doi.org/10.17632/xfh84npb2h.2. This paper does not report original code. Any additional information required to reanalyze the data reported in this paper is available from the lead contact upon request.


Articles from Cell Reports Medicine are provided here courtesy of Elsevier

RESOURCES