Skip to main page content
U.S. flag

An official website of the United States government

Dot gov

The .gov means it’s official.
Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

Https

The site is secure.
The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
. 2022 Jun 27;12(11):4965-4979.
doi: 10.7150/thno.73152. eCollection 2022.

Docetaxel remodels prostate cancer immune microenvironment and enhances checkpoint inhibitor-based immunotherapy

Affiliations

Docetaxel remodels prostate cancer immune microenvironment and enhances checkpoint inhibitor-based immunotherapy

Zehua Ma et al. Theranostics. .

Abstract

Background: Prostate cancer is usually considered as immune "cold" tumor with poor immunogenic response and low density of tumor-infiltrating immune cells, highlighting the need to explore clinically actionable strategies to sensitize prostate cancer to immunotherapy. In this study, we investigated whether docetaxel-based chemohormonal therapy induces immunologic changes and potentiates checkpoint blockade immunotherapy in prostate cancer. Methods: We performed transcriptome and histopathology analysis to characterize the changes of prostate cancer immune microenvironment before and after docetaxel-based chemohormonal therapy. Furthermore, we investigated the therapeutic benefits and underlying mechanisms of chemohormonal therapy combined with anti-PD1 blockade using cellular experiments and xenograft prostate cancer models. Finally, we performed a retrospective cohort analysis to evaluate the antitumor efficacy of anti-PD1 blockade alone or in combination with docetaxel-based chemotherapy. Results: Histopathology assessments on patient samples confirmed the enrichment of tumor-infiltrating T cells after chemohormonal therapy. Moreover, we found that docetaxel activated the cGAS/STING pathway in prostate cancer, subsequently induced IFN signaling, resulting in lymphocytes infiltration. In a xenograft mouse model, docetaxel-based chemohormonal therapy prompted the intratumoral infiltration of T cells and upregulated the abundance of PD1 and PD-L1, thereby sensitizing mouse tumors to the anti-PD1 blockade. To determine the clinical significance of these results, we retrospectively analyzed a cohort of 30 metastatic castration-resistant prostate cancer patients and found that docetaxel combined with anti-PD1 blockade resulted in better prostate-specific antigen progression-free survival when compared with anti-PD1 blockade alone. Conclusions: Our study demonstrates that docetaxel activates the antitumoral immune response and facilitates T cell infiltration in a cGAS/STING-dependent manner, providing a combination immunotherapy strategy that would improve the clinical benefits of immunotherapy.

Keywords: Docetaxel; Immune microenvironment; Immunotherapy; Prostate Cancer; cGAS/STING.

PubMed Disclaimer

Conflict of interest statement

Competing Interests: The authors have declared that no competing interest exists.

Figures

Figure 1
Figure 1
Gene signatures of tumors before and after chemohormonal therapy in prostate cancer patients. A. Principal component analysis of transcriptomic data from all 22 patient samples. Each dot represents a patient sample that is colored on the basis of treatment (blue, pretreatment; red, posttreatment). B. The horizontal bar graph showing the top 20 of upregulated differentially enriched pathways and functions in post-chemohormonal therapy tumor samples compared to paired pretreatment samples. Red bars indicate the immune-related differentially expressed pathways and functions. C. Bubble plot illustrating relevant immune cell profiles of paired pre- and post-chemohormonal therapy tumor samples. Bubble size reflects the percentage of immune cell subtypes enriched in corresponding immune cell profile. Bubble color reflects the P value. D. Changes in fractions of plasma cells, CD8 T cells, M1 macrophages, and M2 Macrophages between paired pre- and post-chemohormonal therapy tumor samples. E. Changes in antigen presentation score, Batf3-DC signature score, and three T cell-phenotype signatures scores (CYT score, CD8+ effector T cell score, and T cell inflamed signature score) between paired pre- and post-chemohormonal therapy tumor samples. F. Changes in the numbers of TRA and TRB clones detected between paired pre- and post-chemohormonal therapy tumor samples. G. Changes in the numbers of individual TRA and TRB clonotypes between paired pre- and post-chemohormonal therapy tumor samples. Each point represents an independent sample. Data were presented as mean values ± SEM. Paired data were analyzed using the paired t-test or Wilcoxon paired rank test.
Figure 2
Figure 2
Multiplex immunofluorescence and immunohistochemistry assessment of immune cells in the tumor microenvironment of pre- and post-chemohormonal therapy tumor samples. A. Representative fluorescence images of immunolabeled CD4+ T cells (CD4+CD3+), CD8+ T cells (CD8+CD3+), and tumor-resident T cells (CD103+CD8+) from paired pre- and post-chemohormonal therapy tumor samples. B. Changes in the densities of immune cells in (A) between paired pre- and post-chemohormonal therapy tumor samples. FOV, field of view. C. Scatterplots comparing the densities of tumor-infiltrating lymphocytes, including CD3+ cells, CD4+ cells, and CD8+ cells between treatment naive and chemohormonal therapy treated tumor samples. Each point represents an independent sample. Data were presented as mean values ±SEM. Unpaired data were analyzed using the t-test or Wilcox rank sum test. Paired data were analyzed using the paired t-test or Wilcoxon paired rank test.
Figure 3
Figure 3
Combination of docetaxel and androgen-deprived treatment activates the cGAS/STING pathway in prostate cancer cells. A. QRT-PCR analysis of genomic DNA (RPL13 and RNA18S) and mitochondrial DNA (MT-ND1 and MT-ND2) in LNCaP cells (left panel) and PC3 cells (right panel) after treatment with DMSO, bicalutamide (BLM), docetaxel (DTX), or bicalutamide plus docetaxel (BLM+DTX) for the indicated time. B. Western blot analysis of cGAS/STING pathways components with indicated antibodies in LNCaP cells (left panel) and PC3 cells (right panel) after treatment with DMSO, bicalutamide (BLM), docetaxel (DTX), or bicalutamide plus docetaxel (BLM+DTX) for the indicated time. C. Representative flow plots and quantification of the positive percentage of p-STING and p-IRF3 expression in LNCaP cells (upper panel) and PC3 cells (lower panel) after DMSO, bicalutamide (BLM), docetaxel (DTX), or bicalutamide plus docetaxel (BLM+DTX) treatment for 24 hours. D. QRT-PCR analysis of cGAS/STING pathway downstream immune genes in LNCaP cells (left panel) and PC3 cells (right panel) after treatment with DMSO, bicalutamide (BLM), docetaxel (DTX), or bicalutamide plus docetaxel (BLM+DTX) for the indicated time. Each point represents an independent experiment. Data were presented as mean values ± SEM. Unpaired data were analyzed using the t-test or Wilcox rank sum test. *, P < 0.05; **, P < 0.01; ***, P < 0.001.
Figure 4
Figure 4
Chemohormonal therapy sensitizes RM1 tumor-bearing mice to anti-PD1 blockade therapy. A. QRT-PCR analysis of PD-L1 mRNA level in treatment naive and chemohormonal therapy tumor samples. Each point represents an independent sample. B. Western blot showing the protein level of PD-L1 in PC3 cells (left panel) and RM1 cells (right panel) after treatment with DMSO, bicalutamide (BLM), docetaxel (DTX), or bicalutamide plus docetaxel (BLM+DTX) for the indicated time. Each point represents an independent experiment. C-D. Tumor size (C) and tumor weight (D) of RM1 tumor xenografts in C57 mice, treated with castration plus either DMSO, anti-PD1 (RMP1-14, 6 mg/kg), bicalutamide (20 mg/kg) + docetaxel (10 mg/kg), or combination treatment. Tumor sizes were measured every 3 days. Each point represents an independent sample. E-F. Tumor size (E) and tumor weight (F) of RM1 (scramble or shSTING) tumor xenografts in C57 mice, treated with castration plus either DMSO or anti-PD1 (RMP1-14, 6 mg/kg) + bicalutamide (20 mg/kg) + docetaxel (10 mg/kg). Tumor sizes were measured every 3 days. Each point represents an independent sample. Data were presented as mean values ±SEM. Unpaired data were analyzed using the t-test or Wilcox rank sum test. *, P < 0.05; **, P < 0.01; ***, P < 0.001.
Figure 5
Figure 5
Chemohormonal therapy activates immune response to suppress xenografted tumors growth. A-B. Representative flow plots (A) and densities (B) of intratumoral CD45+ leukocytes, CD3+ T cells, and CD8+ T cells in xenografted tumors of indicated groups. C. The distribution of PD1 expression in xenografted tumors of indicated groups. The proportion of cells is indicated in the plot. D-E. Representative flow plots (D) and densities (E) of CD45+PD1+ cells and CD3+ PD1+ cells in xenografted tumors of indicated groups. F. Western blot showing the protein level of PD-L1 in xenografted tumors of indicated groups. G. QRT-PCR analysis of cGAS/STING pathway downstream immune genes in xenografted tumors of indicated groups. Each point represents an independent sample. Data were presented as mean values ± SEM. Unpaired data were analyzed using the t-test or Wilcox rank sum test. *, P < 0.05; **, P < 0.01; ***, P < 0.001.
Figure 6
Figure 6
Pilot clinical study to determine metastatic castration-resistant prostate cancer (mCRPC) response to docetaxel plus tislelizumab therapy. A. PSA responses among chemotherapy-naive mCRPC patients after the treatment of tislelizumab with or without docetaxel. Patients were labelled from A to T (left panel) or 1-10 (right panel). B. PSA progression-free survival in patients with ≥25% PSA reduction after the treatment of tislelizumab with or without docetaxel. C. The representative MRI images (upper panel) and 18F-FDG-PET-CT images (lower panel) showing tumor regression in two patients (patient #Q and #T) during the combination treatment of docetaxel and tislelizumab.

References

    1. Sung H, Ferlay J, Siegel R, Laversanne M, Soerjomataram I, Jemal A. et al. Global cancer statistics 2020: globocan estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA: a cancer journal for clinicians. 2021;71:209–49. - PubMed
    1. Cornford P, van den Bergh RCN, Briers E, Van den Broeck T, Cumberbatch MG, De Santis M. et al. Eau-eanm-estro-esur-siog guidelines on prostate cancer. part ii-2020 update: treatment of relapsing and metastatic prostate cancer. European Urology. 2021;79:263–82. - PubMed
    1. Petrylak DP, Tangen CM, Hussain MH, Lara PN Jr, Jones JA, Taplin ME. et al. Docetaxel and estramustine compared with mitoxantrone and prednisone for advanced refractory prostate cancer. N Engl J Med. 2004;351:1513–20. - PubMed
    1. Galon J, Bruni D. Approaches to treat immune hot, altered and cold tumours with combination immunotherapies. Nature reviews Drug discovery. 2019;18:197–218. - PubMed
    1. Sharma P, Allison J. The future of immune checkpoint therapy. Science (New York, NY) 2015;348:56–61. - PubMed

Publication types