Abstract Background Despite the significant progress achieved with PD-1/PD-L1 inhibitors in the treatment of metastatic renal cell carcinoma (mRCC), most patients ultimately progress. Targeted radiotheranostics offer a new potential approach. Although a radiotheranostic platform targeting the prostate-specific membrane antigen (PSMA) has been tested in mRCC in patients preliminarily, thorough preclinical optimization has not been reported. Prostate-specific membrane antigen (PSMA) is overexpressed in tumor-associated neovascular endothelial cells of many solid tumors, including metastatic RCC. Furthermore, radiopharmaceutical therapy using β- and α-particle emitting agents causes immunomodulation by inducing immunogenic cell death, and release of tumor-associated antigens, potentially enhancing inflammatory phenotype. Here, we investigated whether PSMA-based radiotheranostics can be utilized to improve the efficacy of PD-1 therapy. Methods The PSMA+ RENCA model was developed by lentiviral transduction of RENCA (wt) cells and then characterized for basal and IFN-g induced PD-L1 expression. 68Ga-L1 and 225Ac-L1 were synthesized in high radiochemical yield and purity following our reported methods. Male and female BALB/c mice were used for tumor inoculation. 68Ga-L1 was evaluated in small animal PET/CT imaging in flank and PET/MR imaging in orthotopic implantation. A treatment study was conducted to assess the effect of combination therapy in seven groups in the flank tumors at 2 weeks post-inoculation: Control (saline); PD-1 (10 mg/kg); Anti-PD-1 (10 mg/kg)+axitinib (tyrosine kinase inhibitor, 5 mg/kg, oral gavage, for five days/wk); 225Ac-L1 (37 kBq); 225Ac-L1 (2×37 kBq, 1 wk apart); [225Ac-L1 (37 kBq)+PD-1 (10 mg/kg) and [225Ac-L1 2×37 kBq)+PD-1 (10 mg/kg)]. Therapeutic efficacy was assessed by tumor weight and time to progression of tumor volume doubling (TVD). To determine the mechanism of action of the 225Ac-L1/anti-PD-1 combination treatment, tumor-infiltrating lymphoid populations from tumor samples were collected at day 30, and Fluorescence-Activated Cell Sorting analysis was done with fluorochrome-labeled antibodies against CD45, CD3, CD8, IL10, and IL12. Results 68Ga-L1 and 225Ac-L1 confirmed 10-fold and 2-fold higher uptake, respectively, in the PSMA+ RENCA cells compared to RENCA (wt) cells. PET imaging in the flank model displayed ~7-fold higher accumulation of 68Ga-L1 in PSMA+ RENCA than the RENCA (wt). Two-fold higher accumulation of 68Ga-L1 was observed in orthotopic tumors than the normal kidneys during 1-3 h post-injection. Significant lung metastases were detected with 68Ga-L1 PET at 3 weeks in mice with orthotopic tumors. A combination therapy study was conducted using anti-PD-1 and anti-PD-1+axitinib and compared the efficacy with 225Ac-L1 as a single agent (37 kBq and 2×37 kBq, 1 wk apart) and in combination with PD-1. Median TVD increased from 12 d (control) to 16 d (anti-PD-1), 16 d (anti-PD-1+axitinib), 24 d [225Ac-L1 (37 kBq) P<0.001], 24 d [225Ac-L1 (2 × 37 kBq) P<0.0001}, 30 d [anti-PD-1+225Ac-L1 (1×37 kBq) P<0.0001], and undefined, [anti-PD-1+225Ac-L1 2×37 kBq), P<0.0001], respectively. Furthermore, treatment with 225Ac-L1 (2×37 kBq, 1 wk) resulted in a significant lowering of tumor growth (P< 0.01) compared to control, whereas anti-PD-1 (vs. control) alone moderately reduced tumor growth. The combination of the treatment of [PD-1+225Ac-L1 (2×37 kBq), P=0.0001] was the most effective in enhancing tumor growth inhibition compared with the PD-1+axitinib group. Flow cytometry of tumor-infiltrating immune cells of the treatment groups PD-1+225Ac-L1(37 kBq) and PD-1+225Ac-L1 (2×37 kBq) revealed a higher proportion of effector CD3+CD8+ T cells, accompanied by a significant decline in the proportion of immunosuppressive and pro-tumoral CD8+IL10+ T cells and increase in antitumor CD8+IL12+ T cells compared to untreated and treatment control groups (PD-1 or PD-1+axitinib groups). Conclusions Combining radiotheranostic platform, 68Ga-L1/225Ac-L1 with PD-1 therapy reduces tumor burden and improves TVD in a syngeic model of RCC. This is a promising option for metastatic RCC patients with low and heterogeneous PSMA expression. Translation of this method will be pursued actively. DOD CDMRP Funding: yes