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:

PAK4 inhibition improves PD-1 blockade immunotherapy

A Publisher Correction to this article was published on 22 January 2020

This article has been updated

Abstract

Lack of tumor infiltration by immune cells is the main mechanism of primary resistance to programmed cell death protein 1 (PD-1) blockade therapies for cancer. It has been postulated that cancer cell-intrinsic mechanisms may actively exclude T cells from tumors, suggesting that the finding of actionable molecules that could be inhibited to increase T cell infiltration may synergize with checkpoint inhibitor immunotherapy. Here, we show that p21-activated kinase 4 (PAK4) is enriched in non-responding tumor biopsies with low T cell and dendritic cell infiltration. In mouse models, genetic deletion of PAK4 increased T cell infiltration and reversed resistance to PD-1 blockade in a CD8 T cell-dependent manner. Furthermore, combination of anti-PD-1 with the PAK4 inhibitor KPT-9274 improved anti-tumor response compared with anti-PD-1 alone. Therefore, high PAK4 expression is correlated with low T cell and dendritic cell infiltration and a lack of response to PD-1 blockade, which could be reversed with PAK4 inhibition.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Fig. 1: Responding biopsies present features of an adaptive immune response, while non-responding biopsies lack sufficient immune cell infiltration.
Fig. 2: PAK4 expression is enriched in non-infiltrated tumor biopsies and negatively correlates with immune markers in melanoma.
Fig. 3: PAK4 expression correlates with WNT genes in tumor biopsies and regulates WNT signaling activation in vitro.
Fig. 4: PAK4 expression is enriched in non-responding tumor biopsies and negatively correlates with immune markers in multiple tumor types.
Fig. 5: Inhibition of PAK4 reverses tumor-specific T cell exclusion and sensitizes tumors to PD-1 blockade.
Fig. 6: Analysis of tumor-infiltrating immune cells by CyTOF.
Fig. 7: Pharmacological inhibition of PAK4 improves anti-PD-1 anti-tumor response.

Similar content being viewed by others

Data availability

RNA-Seq data supporting the findings of this study have been deposited in the National Center for Biotechnology Information database of Genotypes and Phenotypes (https://www.ncbi.nlm.nih.gov/gap/) with accession number phs001919. The data for the pan-cancer correlation analysis were derived from the TCGA Research Network (http://cancergenome.nih.gov/). Source data on unprocessed blots in Fig. 3 and Extended Data Figs. 35 and 7, as well as numerical raw data for Figs. 3, 5 and 7 and Extended Data Figs. 46 are provided with the paper. All other data supporting the findings of this study are available from the corresponding author on reasonable request.

Change history

References

  1. Ribas, A. & Wolchok, J. D. Cancer immunotherapy using checkpoint blockade. Science 359, 1350–1355 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Tumeh, P. C. et al. PD-1 blockade induces responses by inhibiting adaptive immune resistance. Nature 515, 568–571 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Chen, P. L. et al. Analysis of immune signatures in longitudinal tumor samples yields insight into biomarkers of response and mechanisms of resistance to immune checkpoint blockade. Cancer Discov. 6, 827–837 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  4. Ayers, M. et al. IFN-γ-related mRNA profile predicts clinical response to PD-1 blockade. J. Clin. Invest. 127, 2930–2940 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  5. Riaz, N. et al. Tumor and microenvironment evolution during immunotherapy with nivolumab. Cell 171, 934–949.e15 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Jerby-Arnon, L. et al. A cancer cell program promotes T cell exclusion and resistance to checkpoint blockade. Cell 175, 984–997.e24 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Sharma, P., Hu-Lieskovan, S., Wargo, J. A. & Ribas, A. Primary, adaptive, and acquired resistance to cancer immunotherapy. Cell 168, 707–723 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Liu, C. et al. BRAF inhibition increases tumor infiltration by T cells and enhances the antitumor activity of adoptive immunotherapy in mice. Clin. Cancer Res. 19, 393–403 (2013).

    Article  CAS  PubMed  Google Scholar 

  9. Peng, W. et al. Loss of PTEN promotes resistance to T cell-mediated immunotherapy. Cancer Discov. 6, 202–216 (2016).

    Article  CAS  PubMed  Google Scholar 

  10. Spranger, S. & Gajewski, T. F. Impact of oncogenic pathways on evasion of antitumour immune responses. Nat. Rev. Cancer 18, 139–147 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Spranger, S., Bao, R. & Gajewski, T. F. Melanoma-intrinsic β-catenin signalling prevents anti-tumour immunity. Nature 523, 231–235 (2015).

    Article  CAS  PubMed  Google Scholar 

  12. Nsengimana, J. et al. β-Catenin-mediated immune evasion pathway frequently operates in primary cutaneous melanomas. J. Clin. Invest. 128, 2048–2063 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  13. Grasso, C. S. et al. Genetic mechanisms of immune evasion in colorectal cancer. Cancer Discov. 8, 730–749 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Luke, J. J., Bao, R., Sweis, R. F., Spranger, S. & Gajewski, T. F. WNT/β-catenin pathway activation correlates with immune exclusion across human cancers. Clin. Cancer Res. 25, 3074–3083 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Li, Y. et al. Nucleo-cytoplasmic shuttling of PAK4 modulates β-catenin intracellular translocation and signaling. Biochim. Biophys. Acta 1823, 465–475 (2012).

    Article  CAS  PubMed  Google Scholar 

  16. Rane, C. K. & Minden, A. P21 activated kinase signaling in cancer. Semin. Cancer Biol. 54, 40–49 (2019).

    Article  CAS  PubMed  Google Scholar 

  17. Yun, C. Y. et al. p21-activated kinase 4 critically regulates melanogenesis via activation of the CREB/MITF and β-catenin/MITF pathways. J. Invest. Dermatol. 135, 1385–1394 (2015).

    Article  CAS  PubMed  Google Scholar 

  18. Vershinin, Z., Feldman, M., Chen, A. & Levy, D. PAK4 methylation by SETD6 promotes the activation of the Wnt/β-catenin pathway. J. Biol. Chem. 291, 6786–6795 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Moll, R., Divo, M. & Langbein, L. The human keratins: biology and pathology. Histochem. Cell Biol. 129, 705–733 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Becht, E. et al. Estimating the population abundance of tissue-infiltrating immune and stromal cell populations using gene expression. Genome Biol. 17, 218 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  21. Radu, M., Semenova, G., Kosoff, R. & Chernoff, J. PAK signalling during the development and progression of cancer. Nat. Rev. Cancer 14, 13–25 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Chen, S. et al. Wnt-1 signaling inhibits apoptosis by activating β-catenin/T cell factor-mediated transcription. J. Cell Biol. 152, 87–96 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Li, J. et al. LATS2 suppresses oncogenic Wnt signaling by disrupting β-catenin/BCL9 interaction. Cell Rep. 5, 1650–1663 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Aboukameel, A. et al. Novel p21-activated kinase 4 (PAK4) allosteric modulators overcome drug resistance and stemness in pancreatic ductal adenocarcinoma. Mol. Cancer Ther. 16, 76–87 (2017).

    Article  CAS  PubMed  Google Scholar 

  25. Takao, S. et al. Targeting the vulnerability to NAD+ depletion in B-cell acute lymphoblastic leukemia. Leukemia 32, 616–625 (2018).

    Article  CAS  PubMed  Google Scholar 

  26. Abu Aboud, O. et al. Dual and specific inhibition of NAMPT and PAK4 by KPT-9274 decreases kidney cancer growth. Mol. Cancer Ther. 15, 2119–2129 (2016).

    Article  CAS  PubMed  Google Scholar 

  27. Rane, C. et al. A novel orally bioavailable compound KPT-9274 inhibits PAK4, and blocks triple negative breast cancer tumor growth. Sci. Rep. 7, 42555 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Wang, K. et al. Inhibition of p21 activated kinase enhances tumour immune response and sensitizes pancreatic cancer to gemcitabine. Int. J. Oncol. 52, 261–269 (2018).

    CAS  PubMed  Google Scholar 

  29. Mosely, S. I. et al. Rational selection of syngeneic preclinical tumor models for immunotherapeutic drug discovery. Cancer Immunol. Res. 5, 29–41 (2017).

    Article  CAS  PubMed  Google Scholar 

  30. Ueha, S. et al. Robust antitumor effects of combined anti-CD4-depleting antibody and anti-PD-1/PD-L1 immune checkpoint antibody treatment in mice. Cancer Immunol. Res. 3, 631–640 (2015).

    Article  CAS  PubMed  Google Scholar 

  31. Homet Moreno, B. et al. Response to programmed cell death-1 blockade in a murine melanoma syngeneic model requires costimulation, CD4, and CD8 T cells. Cancer Immunol. Res. 4, 845–857 (2016).

    Article  CAS  PubMed  Google Scholar 

  32. Yadav, M. et al. Predicting immunogenic tumour mutations by combining mass spectrometry and exome sequencing. Nature 515, 572–576 (2014).

    Article  CAS  PubMed  Google Scholar 

  33. Li, X. & Minden, A. PAK4 functions in tumor necrosis factor (TNF) α-induced survival pathways by facilitating TRADD binding to the TNF receptor. J. Biol. Chem. 280, 41192–41200 (2005).

    Article  CAS  PubMed  Google Scholar 

  34. Li, Q. et al. p21-activated kinase 4 as a switch between caspase-8 apoptosis and NF-κB survival signals in response to TNF-α in hepatocarcinoma cells. Biochem. Biophys. Res. Commun. 503, 3003–3010 (2018).

    Article  CAS  PubMed  Google Scholar 

  35. Dunn, G. P., Old, L. J. & Schreiber, R. D. The three Es of cancer immunoediting. Annu. Rev. Immunol. 22, 329–360 (2004).

    Article  CAS  PubMed  Google Scholar 

  36. Nicholas, N. S. et al. PAK4 suppresses PDZ-RhoGEF activity to drive invadopodia maturation in melanoma cells. Oncotarget 7, 70881–70897 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  37. Chen, S. et al. Copy number alterations in pancreatic cancer identify recurrent PAK4 amplification. Cancer Biol. Ther. 7, 1793–1802 (2008).

    Article  CAS  PubMed  Google Scholar 

  38. Wells, C. M., Whale, A. D., Parsons, M., Masters, J. R. & Jones, G. E. PAK4: a pluripotent kinase that regulates prostate cancer cell adhesion. J. Cell Sci. 123, 1663–1673 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  39. Bi, Y. et al. Study on the expression of PAK4 and P54 protein in breast cancer. World J. Surg. Oncol. 14, 160 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  40. Rane, C. K. et al. Decrypting the PAK4 transcriptome profile in mammary tumor forming cells using next generation sequencing. Genomics 110, 248–256 (2018).

    Article  CAS  Google Scholar 

  41. Wong, L. E., Chen, N., Karantza, V. & Minden, A. The Pak4 protein kinase is required for oncogenic transformation of MDA-MB-231 breast cancer cells. Oncogenesis 2, e50 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Kim, H., Woo, D. J., Kim, S. Y. & Yang, E. G. p21-activated kinase 4 regulates HIF-1α translation in cancer cells. Biochem. Biophys. Res. Commun. 486, 270–276 (2017).

    Article  CAS  PubMed  Google Scholar 

  43. King, H. et al. PAK4 interacts with p85 alpha: implications for pancreatic cancer cell migration. Sci. Rep. 7, 42575 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Fu, X. et al. PAK4 confers cisplatin resistance in gastric cancer cells via PI3K/Akt- and MEK/ERK-dependent pathways. Biosci. Rep. 34, e00094 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  45. He, L. F. et al. Activated-PAK4 predicts worse prognosis in breast cancer and promotes tumorigenesis through activation of PI3K/AKT signaling. Oncotarget 8, 17573–17585 (2017).

    Article  PubMed  Google Scholar 

  46. Kim, D., Langmead, B. & Salzberg, S. L. HISAT: a fast spliced aligner with low memory requirements. Nat. Methods 12, 357–360 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Anders, S., Pyl, P. T. & Huber, W. HTSeq—a Python framework to work with high-throughput sequencing data. Bioinformatics 31, 166–169 (2015).

    Article  CAS  PubMed  Google Scholar 

  48. Love, M. I., Huber, W. & Anders, S. Moderated estimation of fold change and dispersion for RNA-Seq data with DESeq2. Genome Biol. 15, 550 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  49. Subramanian, A. et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc. Natl Acad. Sci. USA 102, 15545–15550 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Ran, F. A. et al. Genome engineering using the CRISPR–Cas9 system. Nat. Protoc. 8, 2281–2308 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Brinkman, E. K., Chen, T., Amendola, M. & van Steensel, B. Easy quantitative assessment of genome editing by sequence trace decomposition. Nucleic Acids Res. 42, e168 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  52. Escuin-Ordinas, H. et al. COX-2 inhibition prevents the appearance of cutaneous squamous cell carcinomas accelerated by BRAF inhibitors. Mol. Oncol. 8, 250–260 (2014).

    Article  CAS  PubMed  Google Scholar 

  53. Wei, S. C. et al. Distinct cellular mechanisms underlie anti-CTLA-4 and anti-PD-1 checkpoint blockade. Cell 170, 1120–1133 e1117 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Chen, H. et al. Cytofkit: a bioconductor package for an integrated mass cytometry data analysis pipeline. PLoS Comput. Biol. 12, e1005112 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

Download references

Acknowledgements

This study was funded in part by the Parker Institute for Cancer Immunotherapy, NIH grants R35 CA197633 and P01 CA168585, the Ressler Family Foundation, and support from K. Schultz and D. Schultz (to A.R.). G.A.-R. was supported by the Isabel and Harvey Kibel Fellowship award and Alan Ghitis Fellowship Award for Melanoma Research. D.Y.T. was supported by a Young Investigator Award from the American Society of Clinical Oncology, a grant from the Spanish Society of Medical Oncology for Translational Research in Reference Centers and the V Foundation–Gil Nickel Family Endowed Fellowship in Melanoma Research. J.M.Z. was part of the UCLA Medical Scientist Training Program supported by NIH training grant GM08042. T.S.N. was supported by NIH/NICHD grant K12-HD000850 (Pediatric Scientist Development Program). S.H.-L. was supported by a Young Investigator Award and a Career Development Award from the American Society of Clinical Oncology, a Tower Cancer Research Foundation Grant and a Dr. Charles A. Coltman Fellowship Award from the Hope Foundation. C.-Y.W. was supported by NIH/NIDCR grant R01DE15964. We acknowledge X. Li, L. Dong, J. Yoshizawa and J. Zhou from the UCLA Clinical Microarray Core for sequencing expertise, and J. Min Chen and J. Trent from the Parker Institute for Cancer Immunotherapy Center at UCLA for administrative support. Flow and mass cytometry were performed in the UCLA Jonsson Comprehensive Cancer Center and the Center for AIDS Research Flow Cytometry Core Facility (supported by NIH awards P30 CA016042 and 5P30 AI028697), as well as by the Jonsson Comprehensive Cancer Center, UCLA AIDS Institute and David Geffen School of Medicine at UCLA. The authors thank A. Minden from Rutgers, the State University of New Jersey, for helpful comments.

Author information

Authors and Affiliations

Authors

Contributions

G.A.-R., C.S.G. and A.R. conceived and designed the study. G.A.-R., D.Y.T., W.L., J.M.Z., C.P.-S., T.S.N., A.K., A.J.G., G.C.-L., B.C.-A., S.H.-L., C.-Y.W., C.S.G. and A.R. developed the methodology. B.B.-M., I.B.C., S.H.-L., C-Y.W. and A.R. acquired the data (provided animals, acquired and managed patients, provided facilities, and so on). G.A.-R., D.Y.T., W.L., J.T., E.M., M.J.Q., W.S., E.B., B.C.-A., C-Y.W., C.S.G. and A.R. analyzed and interpreted the data (including statistical analysis, biostatistics and computational analysis). G.A.-R. and A.R. wrote the manuscript. All authors reviewed the manuscript.

Corresponding author

Correspondence to Antoni Ribas.

Ethics declarations

Competing interests

G.A.-R. has received honoraria for consulting with Arcus Biosciences. W.S. and E.B. were employees of Karyopharm Therapeutics when this study was conducted. A.R. has received honoraria for consulting with Amgen, Bristol-Myers Squibb, Chugai, Genentech, Merck, Novartis, Roche and Sanofi, is or has been a member of the scientific advisory board, and holds stock in Advaxis, Arcus Biosciences, Bioncotech Therapeutics, Compugen, CytomX, Five Prime, FLX Bio, ImaginAb, IsoPlexis, Gilead Kite, Lutris Pharma, Merus, PACT Pharma, Rgenix and Tango Therapeutics. G.A.-R., D.Y.T., C.S.G. and A.R. are inventors in a patent application covering the use of PAK4 inhibitors for cancer immunotherapy.

Additional information

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

Extended data

Extended Data Fig. 1 Differential change in immune populations between non-responding and responding biopsies during anti-PD-1 therapy.

Comparison (two-sided, paired T-test) of each of the immune populations and immune markers between baseline and on-treatment tumour samples for responding (n= 5) and non-responding (n= 6) biopsies. From left to right: T cell score (R P= 0.007, NR P= 0.44), Dendritic cell score (R P= 0.009, NR P= 0.08), CD8 T cell score (R P= 0.006, NR P= 0.48), CTL score (R P= 0.01, NR P= 0.43), NK cell score (R P= 0.006, NR P= 0.32), Monocyte lineage score (R P= 0.004, NR P= 0.48), IFNg (R P= 0.01, NR P= 0.47), TNF (R P= 0.01, NR P= 0.9), GZMA (R P= 0.01, NR P= 0.73), PRF1 (R P= 0.004, NR P= 0.29) and CD8A (R P= 0.004, NR P= 0.52) expression. Increase in all immune populations and markers was significant (P < 0.05) only in responding biopsies. *P <0.05, **P < 0.01; ns, not significant

Extended Data Fig. 2 PAK4 expression analysis with immune infiltration and overlap with S100 and β-catenin staining.

a, Comparison of exclusion up Jerby-Arnon score expression (P = 3.28e-05) between tumour biopsies within the upper (n = 15) and lower (n = 15) quartile of PAK4 expression. b, PAK4 correlation with Jerby-Arnon score expression (n = 60) (R = 0.65, P = 1.78e-08). Exclusion up Jerby-Arnon was obtained based on the geometric mean of the 302 gene from Jerby-Arnon et al. c, CD8A (R = -0.39, P = 6.07e-05), TNF (R = -0.49, P = 1.89e-07), GZMA (R = -0.45, P = 2.47e-06), PRF1 (R = -0.28, P = 4e-03) and the different immune populations assessed using MCP-Counter: T cells (R = -0.39, P = 4.41e-05), CD8 T cells (R = -0.36, P = 1.71e-04), cytotoxic lymphocytes (R = -0.28, P = 4.9e-03) and dendritic cells (R = -0.57, P = 3.95e-10). n= 99 for all plots. d, Quantification of PAK4 positive cells out of S100 total positive cells. PT0158_tx2 and PT0112_tx are two biopsies with low T cell infiltration and high PAK4 expression while PT0294_tx2 and PT0349_tx have low PAK4 and high T cell infiltrate as determined by RNAseq. e, Quantification of PAK4 positive cells out of β-catenin total positive cells. From top to bottom box-plots define the maximum, 3rd quartile, median, 1st quartile and minimum values a. P values obtained using two-sided Welch’s t-test a. Correlations were calculated applying Pearson’s correlation coefficient test b, c

Extended Data Fig. 3 Validation of the generation of a PAK4 KO B16 cell line.

a, b, c TIDE analysis of the B16 PAK4 KO clones: 6.2, 8.1 and 8.2 respectively. d, e, Analysis of PAK4 protein expression in the three B16 PAK4 KO clones, B16 WT CRISPR control and rescue cell lines by Western blot. Results are representative from three independent experiments. Unprocessed blot images are provided as a Source Data file d, e.

Source Data

Extended Data Fig. 4 PAK4 depletion impact on nuclear protein β-catenin and WNT signalling activity.

a, Negative control for the Topflash experiment using the Fopflash luciferase vector which contains a mutated version of the TCF/LEF binding motifs. There are no changes in Fopflash activity upon stimulation with Wnt-3a ligand for 8 hours in any of the tested cell lines (n= 3 per group) (P > 0.05 for all comparisons). b, Baseline WNT activity levels assessed by Topflash assay (n= 3 per group). Values were normalized to B16 WT CC cell lines and no significant WNT activity changes were observed between PAK4 WT and KO cell. c, Immunoblots for nuclear β-catenin protein levels show no differences between B16 WT CRISPR control, PAK4 KO and PAK4 rescue cells. Results are representative from three independent experiments. Means +/- SEM two-tailed unpaired t-test a, b. Unprocessed blot images and raw data are provided as a Source Data file a-c.

Source Data

Extended Data Fig. 5 PAK4 inhibition disrupts WNT signalling and melanogenesis.

a, Cells were cultured with 2µM KPT-9274 for 72 hours before nuclear protein isolation. Showing immunoblots for nuclear β-catenin, nuclear phosphor-β-catenin (S675) and nuclear PAK4 protein levels. Results are representative from two independent experiments. b, Cells were cultured with 2µM KPT-9274 for 72 hours and Wnt-3a for 8 hours prior to Topflash assay (n= 3 per group). Pharmacological inhibition of PAK4 significantly decreases sensitivity to Wnt-3a stimulation (P= 0.005 for WT Wnt3a vs WT KPT-9274 + Wnt3a comparison). c, Baseline WNT activity levels assessed by Topflash assay of cell treated with 2µM KPT-9274 for 72 hours (n= 3 per group) (P > 0.05). Values were normalized to untreated B16 WT CC cells. d, RT-PCR for tyrosinase expression show that PAK4 depletion reduces the expression levels of this gene. Showing means +/- SEM. Results are normalized to B16 WT CRISPR control levels and then log2 transformed (n= 3). e, For image, cells were cultured and harvest upon reaching 80% confluency. B16 WT CRISPR Control cell line maintains melanin production over time while PAK4 KO clones lose their pigmentation. Results are representative from three independent experiments. Means +/- SEM two-tailed unpaired t-test b, c. Unprocessed blots and raw data are provided as a Source Data file a-c.

Source Data

Extended Data Fig. 6 In vivo experiments with additional B16 PAK4 KO and rescue clones and CD8 depletion validation.

a, Tumour growth curves for B16 PAK4 KO 8.1 tumours treated with isotype (blue, n = 10) or anti-PD-1 (red, n = 12) (P= 0.00024, day 15). b, Tumour growth curves for B16 PAK4 KO 8.2 tumours treated with isotype (blue, n = 10) or anti-PD-1 (red, n = 10) (P= 0.02, day 15). In both PAK4 KO cell lines anti-PD-1 treated tumours showed decreased tumour growth compared to untreated tumours. c, Tumour growth curves for B16 8.1 PAK4 rescue tumours treated with isotype (blue, n = 5) or anti-PD-1 (red, n = 5). Anti-PD-1 treatment did not result in any significant anti-tumour efficacy (P= 0.80, day 15). d, Flow cytometry analysis of CD8 positive splenocytes after CD8 depletion. Left panel show splenocytes pattern without anti-CD8 treatment (CD8 population = 18.9%) while middle and right panel show splenocytes derived from two independent mice treated with anti-CD8 antibody (CD8 population = 0.77% and 0.50% respectively). Plotting the mean +/- s.e.m a-c. Statistical significance and correction for multiple comparisons was calculated using Holm-Sidak method a-c. Raw data is provided as a Source Data file a-c. *P <0.05, **P < 0.01, ***P <0.001, ****P < 0.0001. ns, not significant.

Source Data

Extended Data Fig. 7 PAK4 KO validation and sensitivity to TNF in MC38 cells.

a, TIDE analysis of the MC38 PAK4 KO 6.9 clone. b, Analysis of PAK4 protein expression in MC38 PAK4 KO 6.9 clone and MC38 WT by Western blot. Results are representative from two independent experiments. c, Cells were plated by triplicate into 96 well plates and then treated with TNF at 100ng/mL. Cell proliferation was measured by cell confluence using the IncuCyte S3 Live Cell Analysis System. TNF treatment decreased proliferation of MC38 WT, MC38 PAK4 KO 6.9 and MC38 PAK4 KO 6.10 cells by 41%, 95% and 74% respectively compared to untreated cells (means +/- SEM). Results are representative from three biologically independent experiments. Unprocessed blots are provided as a Source Data file b.

Source Data

Supplementary information

Source data

Source Data Fig. 3

Unprocessed Western Blots for Fig. 3f

Source Data Fig. 3

Raw data for d, e.

Source Data Fig. 5

Raw data for a-c, f, g.

Source Data Fig. 7

Raw data for a-c

Source Data Extended Data Fig. 3

Unprocessed Western Blots for Extended Data Fig. 3d, e

Source Data Extended Data Fig. 4

Unprocessed Western Blots for Extended Data Fig. 4c

Source Data Extended Data Fig. 4

Raw data for 4a, b

Source Data Extended Data Fig. 5

Unprocessed Western Blots for Extended Data Fig. 5a

Source Data Extended Data Fig. 5

Raw data for b, c

Source Data Extended Data Fig. 6

Raw data for a-c

Source Data Extended Data Fig. 7

Unprocessed Western Blots for Extended Data Fig. 7b

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Abril-Rodriguez, G., Torrejon, D.Y., Liu, W. et al. PAK4 inhibition improves PD-1 blockade immunotherapy. Nat Cancer 1, 46–58 (2020). https://doi.org/10.1038/s43018-019-0003-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s43018-019-0003-0

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