Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Pharmacological disruption of the MTDH–SND1 complex enhances tumor antigen presentation and synergizes with anti-PD-1 therapy in metastatic breast cancer

Abstract

Despite increased overall survival rates, curative options for metastatic breast cancer remain limited. We have previously shown that metadherin (MTDH) is frequently overexpressed in poor prognosis breast cancer, where it promotes metastasis and therapy resistance through its interaction with staphylococcal nuclease domain-containing 1 (SND1). Through genetic and pharmacological targeting of the MTDH–SND1 interaction, we reveal a key role for this complex in suppressing antitumor T cell responses in breast cancer. The MTDH–SND1 complex reduces tumor antigen presentation and inhibits T cell infiltration and activation by binding to and destabilizing Tap1/2 messenger RNAs, which encode key components of the antigen-presentation machinery. Following small-molecule compound C26-A6 treatment to disrupt the MTDH–SND1 complex, we showed enhanced immune surveillance and sensitivity to anti-programmed cell death protein 1 therapy in preclinical models of metastatic breast cancer, in support of this combination therapy as a viable approach to increase immune-checkpoint blockade therapy responses in metastatic breast cancer.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: MTDH promotes metastatic breast cancer by enhancing immune evasion.
Fig. 2: MTDH inhibits T cell activation.
Fig. 3: MTDH destabilizes Tap1/2.
Fig. 4: MTDH forms a complex with SND1 to inhibit tumor antigen presentation and T cell activation.
Fig. 5: MTDH–SND1 blocking activates T cells by enhancing antigen presentation in tumors.
Fig. 6: MTDH–SND1 disruption and anti-PD-1 treatment synergistically inhibit metastatic breast cancer progression.
Fig. 7: C26-A6 treatment synergizes with anti-PD-1 therapy for metastatic breast cancer.
Fig. 8: MTDH expression negatively correlates with CD8+ T cell infiltration and PD-1 expression in patients with triple-negative breast cancer.

Similar content being viewed by others

Data availability

All RNA-sequencing data generated in this study have been deposited as a superseries at the NCBI Gene Expression Omnibus under accession code no. GSE174630. Further information and requests for resources and reagents should be directed to the corresponding author. All requests for raw and analyzed data and materials will be reviewed promptly by the corresponding author to verify whether the request is subject to any intellectual property or confidentiality obligations. Any data and materials that can be shared will be released via a material transfer agreement. Source data are provided with this paper.

References

  1. Criscitiello, C. & Curigliano, G. Immunotherapeutics for breast cancer. Curr. Opin. Oncol. 25, 602–608 (2013).

    Article  CAS  PubMed  Google Scholar 

  2. Ernst, B. & Anderson, K. S. Immunotherapy for the treatment of breast cancer. Curr. Oncol. Rep. 17, 5 (2015).

    Article  PubMed  Google Scholar 

  3. Ravelli, A. et al. Immune-related strategies driving immunotherapy in breast cancer treatment: a real clinical opportunity. Expert Rev. Anticancer Ther. 15, 689–702 (2015).

    Article  CAS  PubMed  Google Scholar 

  4. Page, D. B., Naidoo, J. & McArthur, H. L. Emerging immunotherapy strategies in breast cancer. Immunotherapy 6, 195–209 (2014).

    Article  CAS  PubMed  Google Scholar 

  5. Soliman, H. Immunotherapy strategies in the treatment of breast cancer. Cancer Control 20, 17–21 (2013).

    Article  PubMed  Google Scholar 

  6. Emens, L. A. Breast cancer immunobiology driving immunotherapy: vaccines and immune checkpoint blockade. Expert Rev. Anticancer Ther. 12, 1597–1611 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Hu, G. et al. MTDH activation by 8q22 genomic gain promotes chemoresistance and metastasis of poor-prognosis breast cancer. Cancer Cell 15, 9–20 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Blanco, M. A. et al. Identification of staphylococcal nuclease domain-containing 1 (SND1) as a metadherin-interacting protein with metastasis-promoting functions. J. Biol. Chem. 286, 19982–19992 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Qian, B. J. et al. MTDH/AEG-1-based DNA vaccine suppresses lung metastasis and enhances chemosensitivity to doxorubicin in breast cancer. Cancer Immunol. Immunother. 60, 883–893 (2011).

    Article  CAS  PubMed  Google Scholar 

  10. Wan, L. et al. MTDH-SND1 interaction is crucial for expansion and activity of tumor-initiating cells in diverse oncogene- and carcinogen-induced mammary tumors. Cancer Cell 26, 92–105 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Guo, F. et al. Structural insights into the tumor-promoting function of the MTDH–SND1 complex. Cell Rep. 8, 1704–1713 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Shen, M. et al. Inhibitors that disrupt the MTDH–SND1 complex suppress breast cancer progression and metastasis. Nat. Cancer https://doi.org/10.1038/s43018-021-00279-5 (2021).

  13. Ewens, A., Mihich, E. & Ehrke, M. J. Distant metastasis from subcutaneously grown E0771 medullary breast adenocarcinoma. Anticancer Res. 25, 3905–3915 (2005).

    PubMed  Google Scholar 

  14. Du, J. et al. PDK1 promotes tumor growth and metastasis in a spontaneous breast cancer model. Oncogene 35, 3314–3323 (2016).

    Article  CAS  PubMed  Google Scholar 

  15. Canovas, B. et al. Targeting p38α increases DNA damage, chromosome instability, and the anti-tumoral response to taxanes in breast cancer cells. Cancer Cell 33, 1094–1110 (2018).

    Article  CAS  PubMed  Google Scholar 

  16. Gibby, K. et al. Early vascular deficits are correlated with delayed mammary tumorigenesis in the MMTV-PyMT transgenic mouse following genetic ablation of the NG2 proteoglycan. Breast Cancer Res. 14, R67 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Clarke, S. R. et al. Characterization of the ovalbumin-specific TCR transgenic line OT-I: MHC elements for positive and negative selection. Immunol. Cell Biol. 78, 110–117 (2000).

    Article  CAS  PubMed  Google Scholar 

  18. Hogquist, K. A. et al. T cell receptor antagonist peptides induce positive selection. Cell 76, 17–27 (1994).

    Article  CAS  PubMed  Google Scholar 

  19. Leone, P. et al. MHC class I antigen processing and presenting machinery: organization, function, and defects in tumor cells. J. Natl Cancer Inst. 105, 1172–1187 (2013).

    Article  CAS  PubMed  Google Scholar 

  20. Das, K. et al. Generation of murine tumor cell lines deficient in MHC molecule surface expression using the CRISPR/Cas9 system. PLoS ONE 12, e0174077 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  21. Gejman, R. S. et al. Rejection of immunogenic tumor clones is limited by clonal fraction. eLife 7, e41090 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  22. Solinas, C. et al. Targeting immune checkpoints in breast cancer: an update of early results. ESMO Open 2, e000255 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  23. Nguyen, D. X., Bos, P. D. & Massague, J. Metastasis: from dissemination to organ-specific colonization. Nat. Rev. 9, 274–284 (2009).

    Article  CAS  Google Scholar 

  24. Mellman, I., Coukos, G. & Dranoff, G. Cancer immunotherapy comes of age. Nature 480, 480–489 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Park, T. S., Rosenberg, S. A. & Morgan, R. A. Treating cancer with genetically engineered T cells. Trends Biotechnol. 29, 550–557 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Wolchok, J. D. & Chan, T. A. Cancer: antitumour immunity gets a boost. Nature 515, 496–498 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Kroemer, G., Senovilla, L., Galluzzi, L., Andre, F. & Zitvogel, L. Natural and therapy-induced immunosurveillance in breast cancer. Nat. Med. 21, 1128–1138 (2015).

    Article  CAS  PubMed  Google Scholar 

  28. Marme, F. Immunotherapy in breast cancer. Oncol. Res. Treat. 39, 335–345 (2016).

    Article  CAS  PubMed  Google Scholar 

  29. Bates, J. P., Derakhshandeh, R., Jones, L. & Webb, T. J. Mechanisms of immune evasion in breast cancer. BMC Cancer 18, 556 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  30. Denkert, C. The immunogenicity of breast cancer–molecular subtypes matter. Ann. Oncol. 25, 1453–1455 (2014).

    Article  CAS  PubMed  Google Scholar 

  31. Lanitis, E., Dangaj, D., Irving, M. & Coukos, G. Mechanisms regulating T-cell infiltration and activity in solid tumors. Ann. Oncol. 28, xii18–xii32 (2017).

    Article  CAS  PubMed  Google Scholar 

  32. Wellenstein, M. D. & de Visser, K. E. Cancer-cell-intrinsic mechanisms shaping the tumor immune landscape. Immunity 48, 399–416 (2018).

    Article  CAS  PubMed  Google Scholar 

  33. Johnsen, A. K., Templeton, D. J., Sy, M. & Harding, C. V. Deficiency of transporter for antigen presentation (TAP) in tumor cells allows evasion of immune surveillance and increases tumorigenesis. J. Immunol. 163, 4224–4231 (1999).

    Article  CAS  PubMed  Google Scholar 

  34. Oancea, G. et al. Structural arrangement of the transmission interface in the antigen ABC transport complex TAP. Proc. Natl Acad. Sci. USA 106, 5551–5556 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Blum, J. S., Wearsch, P. A. & Cresswell, P. Pathways of antigen processing. Annu. Rev. Immunol. 31, 443–473 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Leonhardt, R. M., Keusekotten, K., Bekpen, C. & Knittler, M. R. Critical role for the tapasin-docking site of TAP2 in the functional integrity of the MHC class I-peptide-loading complex. J. Immunol. 175, 5104–5114 (2005).

    Article  CAS  PubMed  Google Scholar 

  37. Panter, M. S., Jain, A., Leonhardt, R. M., Ha, T. & Cresswell, P. Dynamics of major histocompatibility complex class I association with the human peptide-loading complex. J. Biol. Chem. 287, 31172–31184 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Sadasivan, B., Lehner, P. J., Ortmann, B., Spies, T. & Cresswell, P. Roles for calreticulin and a novel glycoprotein, tapasin, in the interaction of MHC class I molecules with TAP. Immunity 5, 103–114 (1996).

    Article  CAS  PubMed  Google Scholar 

  39. Gabathuler, R., Reid, G., Kolaitis, G., Driscoll, J. & Jefferies, W. A. Comparison of cell lines deficient in antigen presentation reveals a functional role for TAP-1 alone in antigen processing. J. Exp. Med. 180, 1415–1425 (1994).

    Article  CAS  PubMed  Google Scholar 

  40. Lampen, M. H. et al. CD8+ T cell responses against TAP-inhibited cells are readily detected in the human population. J. Immunol. 185, 6508–6517 (2010).

    Article  CAS  PubMed  Google Scholar 

  41. Henle, A. M., Nassar, A., Puglisi-Knutson, D., Youssef, B. & Knutson, K. L. Downregulation of TAP1 and TAP2 in early stage breast cancer. PLoS ONE 12, e0187323 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  42. Pedersen, M. H. et al. Downregulation of antigen presentation-associated pathway proteins is linked to poor outcome in triple-negative breast cancer patient tumors. Oncoimmunology 6, e1305531 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  43. Qin, Z. et al. Increased tumorigenicity, but unchanged immunogenicity, of transporter for antigen presentation 1-deficient tumors. Cancer Res. 62, 2856–2860 (2002).

    CAS  PubMed  Google Scholar 

  44. Meng, X. et al. Cytoplasmic metadherin (MTDH) provides survival advantage under conditions of stress by acting as RNA-binding protein. J. Biol. Chem. 287, 4485–4491 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  45. Liu, R., Ma, Y. & Chen, X. Quantitative assessment of the association between TAP2 rs241447 polymorphism and cancer risk. J. Cell. Biochem. 120, 15867–15873 (2019).

    Article  CAS  PubMed  Google Scholar 

  46. Chou, H. L., Tian, L., Kumamaru, T., Hamada, S. & Okita, T. W. Multifunctional RNA binding protein OsTudor-SN in storage protein mRNA transport and localization. Plant Physiol. 175, 1608–1623 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Hsu, J. C., Reid, D. W., Hoffman, A. M., Sarkar, D. & Nicchitta, C. V. Oncoprotein AEG-1 is an endoplasmic reticulum RNA-binding protein whose interactome is enriched in organelle resident protein-encoding mRNAs. RNA 24, 688–703 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Jariwala, N. et al. Role of the staphylococcal nuclease and tudor domain containing 1 in oncogenesis (review). Int. J. Oncol. 46, 465–473 (2015).

    Article  CAS  PubMed  Google Scholar 

  49. Tsuchiya, N. et al. SND1, a component of RNA-induced silencing complex, is up-regulated in human colon cancers and implicated in early stage colon carcinogenesis. Cancer Res. 67, 9568–9576 (2007).

    Article  CAS  PubMed  Google Scholar 

  50. Ohs, I. et al. Restoration of natural killer cell antimetastatic activity by IL12 and checkpoint blockade. Cancer Res. 77, 7059–7071 (2017).

    Article  CAS  PubMed  Google Scholar 

  51. Shen, M. et al. Tinagl1 suppresses triple-negative breast cancer progression and metastasis by simultaneously inhibiting integrin/FAK and EGFR signaling. Cancer Cell 35, 64–80 e67 (2019).

    Article  CAS  PubMed  Google Scholar 

  52. Jiang, Y. Z. et al. Preoperative measurement of breast cancer overestimates tumor size compared to pathological measurement. PLoS ONE 9, e86676 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  53. Xiao, Y. et al. Multi-omics profiling reveals distinct microenvironment characterization and suggests immune escape mechanisms of triple-negative breast cancer. Clin. Cancer Res. 25, 5002–5014 (2019).

    Article  CAS  PubMed  Google Scholar 

  54. Shackleton, M. et al. Generation of a functional mammary gland from a single stem cell. Nature 439, 84–88 (2006).

    Article  CAS  PubMed  Google Scholar 

  55. Twyman-Saint Victor, C. et al. Radiation and dual checkpoint blockade activate non-redundant immune mechanisms in cancer. Nature 520, 373–377 (2015).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank G. Ren, W. Li, Z. Li, W. Lu and other laboratory members for technical support and helpful discussions. We thank W. Wang at the Genomics Core Facility of Princeton University for RNA-seq, C. DeCoste and K. Rittenbach at the Molecular Biology Flow Cytometry Resource Facility of Princeton University for the flow cytometry assays. We thank W. Zhang (University of Florida) for providing the E0771 cells. This research was supported by the Brewster Foundation and Ludwig Cancer Research and grants from the Breast Cancer Research Foundation, the National Institutes of Health (no. R01CA134519), Department of Defense Breast Cancer Research Program (no. BC151403), American Cancer Society and Susan G. Komen Foundation to Y.K. and postdoctoral fellowships from Susan G. Komen (no. PDF17332118) and NJCCR (no. DFHS15PPCO21) to M.S. This research was also supported by the Preclinical Imaging and Flow Cytometry Shared Resources of the Rutgers Cancer Institute of New Jersey (no. P30CA072720).

Author information

Authors and Affiliations

Authors

Contributions

M.S. and H.A.S. designed and performed the experiments, analyzed the data and wrote the manuscript. Y.W. and L.W. generated the PyMT MTDH-WT/KO, Py8119-OVA cell lines and performed the RNA sequencing. N.W. and M.R. performed the in vitro tumor–immune cell coculture assays. Y.J., S.Z., S.W. and Z.S. collected the samples of patients with triple-negative breast cancer, performed the IHC staining and generated the survival plots. Y.T. performed the flow cytometry analysis. X.H. maintained the mouse strains and assisted with the animal experiments. Y.K. supervised the overall study, designed the experiments, analyzed the data and wrote the manuscript.

Corresponding author

Correspondence to Yibin Kang.

Ethics declarations

Competing interests

Princeton has filed a disclosure on the findings based on this study. Y.K. and M.S. are named as co-inventors on the disclosure. Y.K. is a cofounder and chair of the Scientific Advisory Board of Firebrand Therapeutics, which is currently developing new therapeutics based on the targeting of MTDH–SND1 and has licensed relevant technologies from Princeton University. Y.K. is also a cofounder of Kayothera and a member of the Scientific Advisory Board for Cytocare and Vibrant Pharma Limited. The other authors declare no competing interests.

Additional information

Peer review information Nature Cancer thanks Daniel Ajona, Taosheng Chen and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data

Extended Data Fig. 1 MTDH depletion reshapes immune cell populations in tumors.

a, KO/PyMT cells were rescued with vector or wild type MTDH. The expression of MTDH was validated by western blot. b, Western blot analysis of endogenous MTDH knockdown in WT/PyMT cells. c, Western blot analysis of endogenous MTDH knockdown in E0771 cells with stable luciferase expression. d, 1 × 106 of E0771 cells with (shMTDH-1, -2) or without (shCtrl) MTDH knockdown were injected into female C57/BL6 mice via tail-vein. 6 weeks after injection, lungs were collected (left) and metastatic nodules were counted (right). n = 6 mice per group. Size bar, 5 mm. Data represent mean ± SEM. Significance determined by one-way ANOVA analysis with Dunnett’s test for multiple comparisons. e, PyMT;UBC-CreERT+/-;Mtdhfl/fl mice with tumors established were treated with vehicle or Tamoxifen (Tmx) for five consecutive days. One week after the treatment, tumors were collected, and RNA was extracted for RNA sequencing. Gene sets that are significantly enriched in ranked gene list of Tmx treated versus control cells. n = 4 mice per group. f,g, Tumors from PyMT;UBC-CreERT+/-;Mtdhfl/fl mice treated with vehicle or Tmx were collected for immunohistochemistry (IHC) staining with indicated antibodies (f). The numbers of positive cells per field were quantified (g). Size bar, 50 μm. Data represent mean ± SEM. n = 8 fields from 4 mice in each group. Significance determined by two tailed Student’s t-test. h, PyMT;UBC-CreERT+/-;Mtdhfl/fl tumorspheres were treated with vehicle (Ctrl) or 4-Hydroxytamoxifen (4-OHT) (MTDH-KO). The spheres were then collected for RNA sequencing. Gene set enrichment analysis demonstrates the enrichment of the indicated gene sets. p and q values automatically determined by GSEA 3.0. The enrichment scores of the indicated signatures from tumorspheres in vitro or tumor samples in vivo (Fig. 1e) were presented (right). Numerical source data for d, g, and uncropped blots for a-c are provided.

Source data

Extended Data Fig. 2 CD8+ T cells depletion partially restores MTDH knockdown induced metastatic inhibition.

a, FVB females were treated with 125 μg/mouse of anti-CD8 antibody or isotype control for 3 days. Peripheral blood was collected for flow cytometry analysis at indicated days after treatment (top). % of CD8+ cells in CD45+ populations are shown. Anti-CD8 antibody treatment scheme that used for the in vivo experiments in this study (bottom). b, 1 × 106 E0771-shCtrl or shMTDH-1 (shMTDH hereafter) cells were injected into C57/BL6 females intravenously. The mice were subjected to isotype control or anti-CD4, anti-CD8 neutralizing antibodies treatment as in (a). 6 weeks after injection, the mice were euthanized, lungs were collected and fixed with Bouin’s solution. Representative lungs are shown and lung metastatic nodules were counted. n = 6 mice per group. Size bar, 5 mm. Data represent mean ± SEM. Significance determined by one-way ANOVA analysis with Sidak’s’s test for multiple comparisons. c, Kaplan-Meier survival curve of C57/BL6 female mice injected with 1 × 106 E0771-shMTDH that were treated with IgG or anti-CD8 neutralizing antibodies. n = 6 mice per group. Significance determined by Log-rank test. d, 5 × 105 -shCtrl or shMTDH E0771 cells were injected into the mammary fat pad of female C57BL6 mice, and the mice were treated with IgG or anti-CD8 neutralizing antibody as in (a). 6 weeks after injection, lungs were harvested and bioluminescent imaging (BLI) was performed to measure lung metastasis. Representative lungs (left), quantitative BLI signals (middle), and fold change of BLI signal between IgG and anti-CD8 groups (right) are shown. n = 16 mice per group. Data represent mean ± SEM. Significance determined by two tailed Student’s t-test. Numerical source data for b, c, d, are provided.

Source data

Extended Data Fig. 3 Characterization of in vitro tumor-immune cell co-culture system.

a, MHC-I (H-2Kd/H-2Dd) presentation in parental Py8119 cells were analyzed by flow cytometry. Isotype IgG served as negative control. b, Stable expression of Ovalbumin (OVA) in the resulted Py8119-OVA cells were confirmed with western blot. c, Surface presentation of OVA (H-2Kb-SIINFEKL) in parental Py8119 and Py8119-OVA cells with or without OT-1 splenocytes co-culture were analyzed with flow cytometry. d, Splenocytes isolated from OT-I mice were treated with PBS or 2 μg/ml of Ovalbumin peptide (OVAp257) for 2 hr. The cells were washed with PBS and plated in fresh media for another 24 hr followed by flow cytometry analysis. % of CD137+ or IFN-γ+ cells in live populations are shown. e, Splenocytes on the CD8 population were gated and the mean fluorescence intensity (MFI) of CD137 and IFN-γ were measured. n = 3 independent experiments. f, The indicated cells with endogenous Mtdh knockdown and vector (Vec) or wild type MTDH rescue were confirmed with western blot. g, The tumorigenesis ability of indicate cell lines were evaluated. Indicated cells were inoculated into the mammary fat pad of C57BL/6 female mice. Ten weeks after injection, lung metastatic nodules were counted. shCtrl, Py8119-shCtrl; KD, Py8119-shMTDH; shCtrl-OVA, Py8119-OVA-shCtrl; KD-OVA, Py8119-OVA-shMTDH; KD-OVA-Vec, Py8119-OVA-shMTDH rescued with vector; KD-OVA-MTDH, Py8119-OVA-shMTDH rescued with wild type MTDH. n = 5 mice per group. h, Py8119-OVA cells with/without endogenous Mtdh knockdown or with/without wild type MTDH rescued were employed for mammary fat pad injections. The injected OT-I female mice were treated with/without anti-CD8 neutralization antibody or IgG. Six weeks after treatment, lung metastasis was determined. n = 9 mice per group. i, Splenocytes co-cultured with indicated Py8119-OVA tumor cells (same as in Fig. 2b) for 24 hr were harvested for flow cytometry analysis. The expression of Granzyme B in CD8+ T cells were examined. MFI, Mean Fluorescence Intensity. n = 3 independent experiments. Data represent mean ± SEM. Significance determined by two tailed Student’s t-test (e,g), or one-way ANOVA analysis with Sidak’s’s test for multiple comparisons (h,i).Numerical source data for e, g, h, i, and uncropped blots for b, f are provided.

Source data

Extended Data Fig. 4 Mtdh acute loss enhances tumor antigen presentation.

a, PyMT;UBC-CreERT+/-;Mtdhfl/fl mice with tumors were treated with vehicle or 60 mg/kg of Tamoxifen (Tmx) for five consecutive days. One week after the treatment, tumors were collected, and RNA was extracted for RNA sequencing. Ingenuity pathway analysis shows the top five molecular and cellular functions of Mtdh acute loss up-regulated genes. p values automatically generated by QIAGEN Ingenuity Pathway Analysis (QIAGEN IPA). b, RNA samples from (a) were subjected to qRT-PCR test of indicated genes. Data represent mean ± SEM. n = 3 independent experiments. Significance determined by two tailed Student’s t-test. c, Py8119-OVA-shCtrl or shMTDH tumor cells were co-cultured with OT-I splenocytes for 0 or 24 hr. Tumor cells were then collected and subjected to flow cytometry to determine the surface presentation of MHC-I. MFI, mean fluorescence intensity. Data represent mean ± SEM. n = 3 independent experiments. Significance determined by two tailed Student’s t-test. Numerical source data for b, c are provided.

Source data

Extended Data Fig. 5 SND1 binds to Tap1/2 and promotes their degradation.

a, Endogenous SND1 knockdown in Py8119-OVA cells was confirmed with western blot. b, Tap1/2 mRNA levels in the indicated Py8119-OVA cells that were co-cultured with OT-I splenocytes for 0 or 24 hr was examined by qRT-PCR. n = 3 independent experiments. c, Indicated Py8119-OVA tumor cells co-cultured for 24 hr were treated with 10 μg/ml of actinomycin D for another 8 hr. RNA levels of Tap1/2 in tumor cells were determined by qRT-PCR. n = 3 independent experiments. d-g Py8119-OVA cells with SND1 (d) or MTDH (f) knockdown were subjected to RIP assay after 24 hr co-culture. The interaction between SND1 or MTDH and Tap1/2 was determined by PCR. Tap1/2 RNAs that bind to MTDH (e) or SND1 (g) were quantified and normalized to the pulled down MTDH and SND1 levels, respectively. n = 3 independent experiments. h, Electrophoretic mobility gel shift assay was performed with in vitro transcribed TAP1/2 mRNA incubated with PBS, recombinant MTDH and SND1 alone or in combination. Data represent mean ± SEM. Significance determined by one-way ANOVA analysis with Dunnett’s test for multiple comparisons (b,c,e), or two tailed Student’s t-test (g). Numerical source data for b, c, e, g,and uncropped images for a, d, f, h are provided.

Source data

Extended Data Fig. 6 SND1 inhibits antigen presentation and T-cell activation.

a-d, Py8119-OVA tumor cells (a,b) and OT-I splenocytes (c,d) after co-culture were collected to test OVA (H-2Kb-SIINFEKL) and MHC-I presentation, or CD137 and IFN-γ expression. MFI, mean fluorescence intensity; AU, arbitrary units. e, Media from (a) was employed for ELSA to test IFN-γ concentration and cytotoxicity assay. In panels from a-e, n = 3 independent experiments. f, PyMT tumor cells with MTDH KD or indicated rescues were injected into FVB females intravenously. Five weeks after injection, lung metastatic nodules were counted, and representative lungs were shown. n = 6 lungs per group. Data represent mean ± SEM. Significance determined by one-way ANOVA analysis with Dunnett’s test for multiple comparisons (a-e), or one-way ANOVA analysis with Sidak’s test for multiple comparisons (f). Numerical source data for a-f are provided.

Source data

Extended Data Fig. 7 C26-A6 treatment elevates immune responses in tumors.

a, The correlation between all the gene sets that alters by Mtdh acute loss and C26-A6 treatment. b,c, Py8119-OVA cells co-cultured with OT-I splenocytes were treated with 200 μM of C26-A6 or same amount of vehicle. The binding between SND1 and Tap1/2 in tumor cells were determined by RIP assay (b). Tap1/2 RNAs that bind to SND1 were quantified and normalized to the pulled down SND1 levels (c). d, MHC-I presentation in Py8119-OVA cells with/without 200 μM of C26-A6 treatment in co-culture were determined by flow cytometry. MFI, mean fluorescence intensity; AU, arbitrary units. e,f, The expression of CD137 and IFN-γ in splenocytes co-cultured with Py8119-OVA were determined by flow cytometry upon 200 μM of C26-A6 or vehicle treatment. MFI, Mean Fluorescence Intensity; AU, arbitrary units. In panels from b-f, data represent mean ± SEM. n = 3 independent experiments. Significance determined by two tailed Student’s t-test. Numerical source data for a, c-f and uncropped images for b are provided.

Source data

Extended Data Fig. 8 MTDH-SND1 complex promotes immune evasion through Tap1/2.

a, Western blot analyzing to confirm the knock down of Tap1/2 in E0771 cells (E0771-OVA) stably expressing luciferase and OVA after lentiviral transduction of respective shRNAs. b-d, E0771-OVA cells with (shTap1/2) or without (shCtrl) Tap1/2 knockdown were co-cultured with OT-I splenocytes with the ratio of 1:10 (tumor cells:splenocytes). 24 hr after co-culture, Tumor cells (b,c) and splenocytes (d) were collected for examining OVA (H-2Kb-SIINFEKL), MHC-I, and CD137 expression, respectively. MFI, mean fluorescence intensity; AU, arbitrary units. Data represent mean ± SEM. n = 3 independent experiments. Significance determined by one-way ANOVA analysis with Sidak’s test for multiple comparisons. e, The same co-culture experiment as in (b) were performed. The live tumor cells were indicated by luciferase signal. The percentage of live cells were determined as normalized to the non-co-culture control. Data represent mean ± SEM. n = 5 independent experiments. Significance determined by one-way ANOVA analysis with Sidak’s test for multiple comparisons. f, PresentER-Vector or PresentER-OVA (H-2Kb-SIINFEKL) system was stably expressed in Py8119 cells. The surface presentation of OVA (H-2Kb-SIINFEKL) was validated by flow cytometry analysis. g, Western blot analysis confirming the knock down of MTDH in Py8119 cells stably expressing luciferase and PresentER-OVA (Py8119-PresentER-OVA) after lentiviral transduction of respective shRNAs. h, Py8119-PresentER-OVA cells with (shMTDH) or without (shCtrl) MTDH knockdown were co-cultured with OT-I splenocytes with the ratio of 1:10 (tumor cells:splenocytes). 24 hr after co-culture, Tumor cells were collected for examining OVA (H-2Kb-SIINFEKL). Data represent mean ± SEM. n = 3 independent experiments. Significance determined by two tailed Student’s t-test. i, The same co-culture experiment as in (h) were performed. The live tumor cells were indicated by luciferase signal. The percentage of live cells were determined as normalized to the non-co-culture control. Data represent mean ± SEM. n = 3 independent experiments. Significance determined by two tailed Student’s t-test. Numerical source data for b-e, h, i and uncropped blots for a and g are provided.

Source data

Extended Data Fig. 9 MTDH-SND1 disruption and anti-PD-1 treatment synergistically enhance anti-tumor immune response.

a, OT-I splenocytes were co-cultured with Py8119-OVA cells with or without 200 μM of C26-A6 treatment for 24 hr. The expression of PD-1 in CD8+ T cells was examined by flow cytometry. % of CD8+PD-1+ in live populations are shown. Data represent mean ± SEM. n = 3 independent experiments. Significance determined by two tailed Student’s t-test. b, Cells in (a) were gated on the CD8+ T cell population and the mean fluorescence intensity (MFI) of PD-1 expression was measured. AU, arbitrary units. Data represent mean ± SEM. n = 3 independent experiments. Significance determined by two tailed Student’s t-test. c, Schematic diagram of treatment. PyMT;UBC-CreERT+/-;Mtdhfl/fl females with tumors established were treated with Tmx and anti-PD-1 alone or in combination. Tmx, Tamoxifen, 60 mg/kg i.p. for 5 consecutive days; anti-PD-1, 200 μg/mouse i.p. injection, twice per week for the first week and then once per week after that. d, Primary tumors and lungs from experiment in Fig. 6c were fixed for CD8 IHC staining. Scale bar, 100 μm. Numerical source data for a and b are provided.

Source data

Extended Data Fig. 10 C26-A6 combined with anti-PD-1 treatment reshapes the tumor immune microenvironment.

a,b, 100k PyMT tumor cells were orthotopically injected into the mammary glands of FVB females. The mice were randomized and divided into three groups when primary tumors were established, followed by vehicle, C26-A6, or C26-A6 + anti-PD-1 treatment. Six weeks after treatment, primary tumors and lung with metastatic lesions were collected for flow analysis with indicated antibodies. % of CD11b+F4/80+, Ly6GlowLy6Chigh, Ly6GhighLy6Clow, CD3NK1.1+ in CD45+ population are shown. % of CD4+FOXP3+ in CD3+ population are shown, and % of GITR+LAG-3+ in CD8+ population are shown. Anti-PD-1, 200 μg/mouse i.p. injection, twice per week for the first week and then once per week after that; C26-A6, 15 mg/kg i.v. injection, 5 days per week. n = 6 mice per group. Data represent mean ± SEM. Significance determined by one-way ANOVA analysis with Dunnett’s test for multiple comparisons. c, Negative correlation between MTDH expression and CD8+ T cell infiltration or PD-1 expression in TNBC patients. Representative IHC images are shown in Fig. 8a. p-value by two-sided chi square test tests. Numerical source data for a and b are provided.

Source data

Supplementary information

Supplementary Information

Supplementary Fig. 1

Reporting Summary

Supplementary Table

Supplementary Table 1

Source data

Source Data Fig. 1

Statistical source data for Fig. 1.

Source Data Fig. 2

Statistical source data for Fig. 2.

Source Data Fig. 2

Unprocessed western blots for Fig. 2.

Source Data Fig. 3

Statistical source data for Fig. 3.

Source Data Fig. 3

Unprocessed western blots and gels for Fig. 3.

Source Data Fig. 4

Statistical source data for Fig. 4.

Source Data Fig. 4

Unprocessed western blots and gels for Fig. 4.

Source Data Fig. 5

Statistical source data for Fig. 5.

Source Data Fig. 5

Unprocessed western blots and gels for Fig. 5.

Source Data Fig. 6

Statistical source data for Fig. 6.

Source Data Fig. 7

Statistical source data for Fig. 7.

Source Data Fig. 8

Statistical source data for Fig. 8.

Source Data Extended Data Fig. 1

Statistical source data for Extended Data Fig. 1.

Source Data Extended Data Fig. 1

Unprocessed western blots for Extended Data Fig. 1.

Source Data Extended Data Fig. 2

Statistical source data for Extended Data Fig. 2.

Source Data Extended Data Fig. 3

Statistical source data for Extended Data Fig. 3.

Source Data Extended Data Fig. 3

Unprocessed western blots for Extended Data Fig. 3.

Source Data Extended Data Fig. 4

Statistical source data for Extended Data Fig. 4.

Source Data Extended Data Fig. 5

Statistical source data for Extended Data Fig. 5.

Source Data Extended Data Fig. 5

Unprocessed western blots and gels for Extended Data Fig. 5.

Source Data Extended Data Fig. 6

Statistical source data for Extended Data Fig. 6.

Source Data Extended Data Fig. 7

Statistical source data for Extended Data Fig. 7.

Source Data Extended Data Fig. 7

Unprocessed western blots and gels for Extended Data Fig. 7.

Source Data Extended Data Fig. 8

Statistical source data for Extended Data Fig. 8.

Source Data Extended Data Fig. 8

Unprocessed western blots for Extended Data Fig. 8.

Source Data Extended Data Fig. 9

Statistical source data for Extended Data Fig. 9.

Source Data Extended Data Fig. 10

Statistical source data for Extended Data Fig. 10.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Shen, M., Smith, H.A., Wei, Y. et al. Pharmacological disruption of the MTDH–SND1 complex enhances tumor antigen presentation and synergizes with anti-PD-1 therapy in metastatic breast cancer. Nat Cancer 3, 60–74 (2022). https://doi.org/10.1038/s43018-021-00280-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s43018-021-00280-y

This article is cited by

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer