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:

Neoadjuvant cabozantinib and nivolumab convert locally advanced hepatocellular carcinoma into resectable disease with enhanced antitumor immunity

Abstract

A potentially curative hepatic resection is the optimal treatment for hepatocellular carcinoma (HCC) but most patients are not candidates for resection and most resected HCCs eventually recur. Until recently, neoadjuvant systemic therapy for HCC has been limited by a lack of effective systemic agents. Here, in a single-arm phase 1b study, we evaluated the feasibility of neoadjuvant cabozantinib and nivolumab in patients with HCC, including patients outside of traditional resection criteria (ClinicalTrials.gov ID NCT03299946). Of 15 patients enrolled, 12 (80%) underwent successful margin-negative resection and 5 out of 12 (42%) had major pathological responses. In-depth biospecimen profiling demonstrated an enrichment in effector T cells, as well as tertiary lymphoid structures, CD138+ plasma cells, and a distinct spatial arrangement of B cells in responders compared to nonresponders, indicating an orchestrated B cell contribution to antitumor immunity in HCC.

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: Clinical responses to neoadjuvant cabozantinib and nivolumab.
Fig. 2: Cabozantinib enhances systemic and local antitumor T cell responses.
Fig. 3: Response to cabozantinib and nivolumab is characterized by an immune-rich TME.
Fig. 4: Spatial relationships among cell types are distinct with respect to response.
Fig. 5: Proximity between lymphoid and macrophage subtypes is key determinant of response to cabozantinib and nivolumab.

Similar content being viewed by others

Data availability

The CyTOF datasets analyzed in this study have been deposited in FlowRepository (FR-FCM-Z34P). The imaging mass cytometry dataset has been deposited as part of the published Code Ocean capsule (https://doi.org/10.24433/CO.3769407.v2). Source data are provided with this paper. All other data supporting the findings of this study are available from the corresponding author upon reasonable request.

Code availability

The code developed for all the spatial analyses performed in this study has been published as an executable version on Code Ocean (https://doi.org/10.24433/CO.3769407.v2). The code is distributed under the Massachusetts Institute of Technology license (https://opensource.org/licenses/MIT).

References

  1. Park, J.-W. et al. Global patterns of hepatocellular carcinoma management from diagnosis to death: the BRIDGE Study. Liver Int. 35, 2155–2166 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  2. Yang, J. D. et al. A global view of hepatocellular carcinoma: trends, risk, prevention and management. Nat. Rev. Gastroenterol. Hepatol. 16, 589–604 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  3. Siegel, R. L., Miller, K. D. & Jemal, A. Cancer statistics, 2020. CA Cancer J. Clin. 70, 7–30 (2020).

    Article  PubMed  Google Scholar 

  4. Liver Cancer Study Group of Japan Primary liver cancer in Japan. Clinicopathologic features and results of surgical treatment. Ann. Surg. 211, 277–287 (1990).

    Google Scholar 

  5. Parker, G. A. et al. Intraoperative ultrasound of the liver affects operative decision making. Ann. Surg. 209, 569–576 (1989).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Fong, Y., Sun, R. L., Jarnagin, W. & Blumgart, L. H. An analysis of 412 cases of hepatocellular carcinoma at a Western center. Ann. Surg. 229, 790–799 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Minagawa, M., Ikai, I., Matsuyama, Y., Yamaoka, Y. & Makuuchi, M. Staging of hepatocellular carcinoma: assessment of the Japanese TNM and AJCC/UICC TNM systems in a cohort of 13,772 patients in Japan. Ann. Surg. 245, 909–922 (2007).

    Article  PubMed  PubMed Central  Google Scholar 

  8. Belghiti, J., Panis, Y., Farges, O., Benhamou, J. P. & Fekete, F. Intrahepatic recurrence after resection of hepatocellular carcinoma complicating cirrhosis. Ann. Surg. 214, 114–117 (1991).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Chen, M. F. et al. Postoperative recurrence of hepatocellular carcinoma. Two hundred five consecutive patients who underwent hepatic resection in 15 years. Arch. Surg. 129, 738–742 (1994).

    Article  CAS  PubMed  Google Scholar 

  10. Tabrizian, P., Jibara, G., Shrager, B., Schwartz, M. & Roayaie, S. Recurrence of hepatocellular cancer after resection: patterns, treatments, and prognosis. Ann. Surg. 261, 947–955 (2015).

    Article  PubMed  Google Scholar 

  11. Llovet, J. M. et al. Sorafenib in advanced hepatocellular carcinoma. N. Engl. J. Med. 359, 378–390 (2008).

    Article  CAS  PubMed  Google Scholar 

  12. Bruix, J. et al. Adjuvant sorafenib for hepatocellular carcinoma after resection or ablation (STORM): a phase 3, randomised, double-blind, placebo-controlled trial. Lancet Oncol. 16, 1344–1354 (2015).

    Article  CAS  PubMed  Google Scholar 

  13. Adachi, E. et al. Effects of preoperative transcatheter hepatic arterial chemoembolization for hepatocellular carcinoma. The relationship between postoperative course and tumor necrosis. Cancer 72, 3593–3598 (1993).

    Article  CAS  PubMed  Google Scholar 

  14. Uchida, M. et al. Role of preoperative transcatheter arterial oily chemoembolization for resectable hepatocellular carcinoma. World J. Surg. 20, 326–331 (1996).

    Article  CAS  PubMed  Google Scholar 

  15. Lee, K. T. et al. The effect of preoperative transarterial chemoembolization of resectable hepatocellular carcinoma on clinical and economic outcomes. J. Surg. Oncol. 99, 343–350 (2009).

    Article  PubMed  Google Scholar 

  16. Wu, C. C. et al. Preoperative transcatheter arterial chemoembolization for resectable large hepatocellular carcinoma: a reappraisal. Br. J. Surg. 82, 122–126 (1995).

    Article  CAS  PubMed  Google Scholar 

  17. Sasaki, A. et al. Preoperative transcatheter arterial chemoembolization reduces long-term survival rate after hepatic resection for resectable hepatocellular carcinoma. Eur. J. Surg. Oncol. 32, 773–779 (2006).

    Article  CAS  PubMed  Google Scholar 

  18. Yarchoan, M. et al. Recent developments and therapeutic strategies against hepatocellular carcinoma. Cancer Res. 79, 4326–4330 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Abou-Alfa, G. K. et al. Cabozantinib in patients with advanced and progressing hepatocellular carcinoma. N. Engl. J. Med. 379, 54–63 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Pinato, D. J. et al. Immune-based therapies for hepatocellular carcinoma. Oncogene 39, 3620–3637 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Kudo, M. Scientific rationale for combined immunotherapy with PD-1/PD-L1 antibodies and VEGF inhibitors in advanced hepatocellular carcinoma. Cancers (Basel) 12, 1089 (2020).

    Article  CAS  Google Scholar 

  22. Yi, M. et al. Synergistic effect of immune checkpoint blockade and anti-angiogenesis in cancer treatment. Mol. Cancer 18, 60 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  23. Finn, R. S. et al. Atezolizumab plus bevacizumab in unresectable hepatocellular carcinoma. N. Engl. J. Med. 382, 1894–1905 (2020).

    Article  CAS  PubMed  Google Scholar 

  24. Yau, T. et al. Nivolumab (NIVO) + ipilimumab (IPI) + cabozantinib (CABO) combination therapy in patients (pts) with advanced hepatocellular carcinoma (aHCC): results from CheckMate 040. J. Clin. Oncol. 38, 478 (2020).

    Article  Google Scholar 

  25. Ho, W. J. et al. Viral status, immune microenvironment and immunological response to checkpoint inhibitors in hepatocellular carcinoma. J. Immunother. Cancer 8, e000394 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  26. Goyal, L. et al. A phase 2 and biomarker study of cabozantinib in patients with advanced cholangiocarcinoma. Cancer 123, 1979–1988 (2017).

    Article  CAS  PubMed  Google Scholar 

  27. Leibowitz-Amit, R. et al. Changes in plasma biomarkers following treatment with cabozantinib in metastatic castration-resistant prostate cancer: a post hoc analysis of an extension cohort of a phase II trial. J. Transl. Med. 14, 12 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  28. Van Gassen, S. et al. FlowSOM: using self-organizing maps for visualization and interpretation of cytometry data. Cytometry A 87, 636–645 (2015).

    Article  PubMed  Google Scholar 

  29. Tokunaga, R. et al. CXCL9, CXCL10, CXCL11/CXCR3 axis for immune activation—a target for novel cancer therapy. Cancer Treat. Rev. 63, 40–47 (2018).

    Article  CAS  PubMed  Google Scholar 

  30. Gregory, J. L. et al. Macrophage migration inhibitory factor induces macrophage recruitment via CC chemokine ligand 2. J. Immunol. 177, 8072–8079 (2006).

    Article  CAS  PubMed  Google Scholar 

  31. Zhao, L. et al. Recruitment of a myeloid cell subset (CD11b/Gr1mid) via CCL2/CCR2 promotes the development of colorectal cancer liver metastasis. Hepatology 57, 829–839 (2013).

    Article  CAS  PubMed  Google Scholar 

  32. Lan, Q. et al. CCL26 participates in the PRL-3-induced promotion of colorectal cancer invasion by stimulating tumor-associated macrophage infiltration. Mol. Cancer Ther. 17, 276–289 (2018).

    Article  CAS  PubMed  Google Scholar 

  33. Shalapour, S. et al. Inflammation-induced IgA+ cells dismantle anti-liver cancer immunity. Nature 551, 340–345 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Förster, R. et al. CCR7 coordinates the primary immune response by establishing functional microenvironments in secondary lymphoid organs. Cell 99, 23–33 (1999).

    Article  PubMed  Google Scholar 

  35. Tumanov, A. V. et al. Cellular source and molecular form of TNF specify its distinct functions in organization of secondary lymphoid organs. Blood 116, 3456–3464 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Boussiotis, V. A., Nadler, L. M., Strominger, J. L. & Goldfeld, A. E. Tumor necrosis factor alpha is an autocrine growth factor for normal human B cells. Proc. Natl Acad. Sci. USA 91, 7007–7011 (1994).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Li, J. et al. Tumor cell-intrinsic factors underlie heterogeneity of immune cell infiltration and response to immunotherapy. Immunity 49, 178–193.e7 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Theivanthiran, B. et al. A tumor-intrinsic PD-L1/NLRP3 inflammasome signaling pathway drives resistance to anti-PD-1 immunotherapy. J. Clin. Invest. 130, 2570–2586 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Wei, Z.-W. et al. CXCL1 promotes tumor growth through VEGF pathway activation and is associated with inferior survival in gastric cancer. Cancer Lett. 359, 335–343 (2015).

    Article  CAS  PubMed  Google Scholar 

  40. Scapini, P. et al. CXCL1/macrophage inflammatory protein-2-induced angiogenesis in vivo is mediated by neutrophil-derived vascular endothelial growth factor-A. J. Immunol. 172, 5034–5040 (2004).

    Article  CAS  PubMed  Google Scholar 

  41. Wang, D., Sun, H., Wei, J., Cen, B. & DuBois, R. N. CXCL1 is critical for premetastatic niche formation and metastasis in colorectal cancer. Cancer Res. 77, 3655–3665 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Kaseb, A. et al. Randomized, open-label, perioperative phase II study evaluating nivolumab alone versus nivolumab plus ipilimumab in patients with resectable HCC. Ann. Oncol. 30, IV112 (2019).

    Article  Google Scholar 

  43. Topalian, S. L., Taube, J. M. & Pardoll, D. M. Neoadjuvant checkpoint blockade for cancer immunotherapy. Science 367, eaax0182 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Helmink, B. A. et al. B cells and tertiary lymphoid structures promote immunotherapy response. Nature 577, 549–555 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Cabrita, R. et al. Tertiary lymphoid structures improve immunotherapy and survival in melanoma. Nature 577, 561–565 (2020).

    Article  CAS  PubMed  Google Scholar 

  46. Petitprez, F. et al. B cells are associated with survival and immunotherapy response in sarcoma. Nature 577, 556–560 (2020).

    Article  CAS  PubMed  Google Scholar 

  47. Lund, F. E. Cytokine-producing B lymphocytes—key regulators of immunity. Curr. Opin. Immunol. 20, 332–338 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Yan, J., Harvey, B. P., Gee, R. J., Shlomchik, M. J. & Mamula, M. J. B cells drive early T cell autoimmunity in vivo prior to dendritic cell-mediated autoantigen presentation. J. Immunol. 177, 4481–4487 (2006).

    Article  CAS  PubMed  Google Scholar 

  49. Hong, S. et al. B cells are the dominant antigen-presenting cells that activate naive CD4+ T cells upon immunization with a virus-derived nanoparticle antigen. Immunity 49, 695–708 (2018).

    Article  CAS  PubMed  Google Scholar 

  50. Liu, Y. et al. Immune cell PD-L1 colocalizes with macrophages and is associated with outcome in PD-1 pathway blockade therapy. Clin. Cancer Res. 26, 970–977 (2020).

    Article  CAS  PubMed  Google Scholar 

  51. Giurisato, E. et al. Extracellular-regulated protein kinase 5-mediated control of p21 expression promotes macrophage proliferation associated with tumor growth and metastasis. Cancer Res. 80, 3319–3330 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Lane, R. S. et al. IFNγ-activated dermal lymphatic vessels inhibit cytotoxic T cells in melanoma and inflamed skin. J. Exp. Med. 215, 3057–3074 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Tang, H. et al. PD-L1 on host cells is essential for PD-L1 blockade-mediated tumor regression. J. Clin. Invest. 128, 580–588 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  54. Rodriguez, P. C. et al. Arginase I production in the tumor microenvironment by mature myeloid cells inhibits T-cell receptor expression and antigen-specific T-cell responses. Cancer Res. 64, 5839–5849 (2004).

    Article  CAS  PubMed  Google Scholar 

  55. Cesano, A. nCounter® PanCancer Immune Profiling Panel (NanoString Technologies, Inc., Seattle, WA). J. Immunother. Cancer 3, 42 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  56. Weber, L. M., Nowicka, M., Soneson, C. & Robinson, M. D. diffcyt: differential discovery in high-dimensional cytometry via high-resolution clustering. Commun. Biol. 2, 183 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  57. Mahnke, Y. D., Brodie, T. M., Sallusto, F., Roederer, M. & Lugli, E. The who’s who of T-cell differentiation: human memory T-cell subsets. Eur. J. Immunol. 43, 2797–2809 (2013).

    Article  CAS  PubMed  Google Scholar 

  58. Hartmann, F. J. et al. Comprehensive immune monitoring of clinical trials to advance human immunotherapy. Cell Rep. 28, 819–831.e4 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Becht, E. et al. Dimensionality reduction for visualizing single-cell data using UMAP. Nat. Biotechnol. 37, 38–44 (2019).

    Article  CAS  Google Scholar 

  60. Berg, S. et al. ilastik: interactive machine learning for (bio)image analysis. Nat. Methods 16, 1226–1232 (2019).

    Article  CAS  PubMed  Google Scholar 

  61. Carpenter, A. E. CellProfiler: image analysis software for identifying and quantifying cell phenotypes. Genome Biol. 7, R100 (2006).

    Article  PubMed  PubMed Central  Google Scholar 

  62. Nembrini, S., König, I. R. & Wright, M. N. The revival of the Gini importance? Bioinformatics 34, 3711–3718 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Taube, J. M. et al. Association of PD-1, PD-1 ligands, and other features of the tumor immune microenvironment with response to anti-PD-1 therapy. Clin. Cancer Res. 20, 5064–5074 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. 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 

  65. Robinson, M. D., McCarthy, D. J. & Smyth, G. K. edgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics 26, 139–140 (2010).

    Article  CAS  PubMed  Google Scholar 

  66. Ritchie, M. E. et al. limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 43, e47 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank the patients and their families who participated in this research and the clinical and laboratory research teams. We also thank E. Wang, A. Cooper and S. Isibor for helpful discussions and technical assistance. We thank the University of Maryland School of Medicine Center for Innovative Biomedical Resources Flow Cytometry and Mass Cytometry Core Facility for their help with the CyTOF data collection and acknowledge the support of Therapeutics Insights Services at Fluidigm for the IMC data collection. Finally, we acknowledge the support from the Emerson Collective Cancer Research Fund (grant no. 640183 to E.M.J.), the National Cancer Institute (NCI) Specialized Program of Research Excellence in Gastrointestinal Cancers (grant no. P50CA062924 to E.M.J.), the NCI Informatics Technologies for Cancer Research (grant no. U01CA212007 to E.J.F.), the National Institutes of Health (NIH) Multiscale Modeling Consortium (grant no. U01CA212007 to E.J.F.), Exelixis (M.Y.), Bristol Myers Squibb (M.Y.), the NIH Center Core Grant P30CA006973 and Johns Hopkins Bloomberg–Kimmel Institute for Cancer Immunotherapy.

Author information

Authors and Affiliations

Authors

Contributions

W.J.H., Q.Z. and M.Y. designed and led all correlative analyses. M.Y. conceived and designed the clinical trial. J.D., A.P., B.W., I.R.K., M.W., B.P., R.B., W.R.B., C.S., A.E., J.H., D.A.L. and M.Y. consented patients and/or conducted the clinical trial. Q.Z. and R.A.A. performed the histopathological review and immunohistochemical analyses. W.J.H. led the development of the mass cytometry workflows. S.X., J.L., A.M., G.M., N.G. and S.C. provided sample and data processing support. S.Z., D.L., D.Q., A.D., L.D., G.S. and E.J.F. provided bioinformatics support. E.A.S. and E.M.J. were involved in study design and data interpretation. W.J.H. and M.Y. wrote the manuscript. All authors reviewed and approved the manuscript.

Corresponding author

Correspondence to Mark Yarchoan.

Ethics declarations

Competing interests

W.J.H. is a coinventor of patents with potential for receiving royalties from Rodeo Therapeutics, is a consultant for Exelixis and receives research funding from Sanofi. R.A.A. reports receiving a commercial research support from Bristol Myers Squibb and is a consultant/advisory board member for Bristol Myers Squibb, Merck, AstraZeneca, Incyte and RAPT Therapeutics. E.M.J. reports receiving a commercial research grant from Bristol Myers Squibb and Aduro Biotech and is a consultant/advisory board member for the Lustgarten Foundation, Parker Institute for Cancer Immunotherapy, CStone Pharmaceuticals, Dragonfly, Genocea, Achilles Therapeutics and Adaptive Biotechnologies; she is cofounder of Abmeta Biotech. M.Y. reports receiving research grants from Incyte, Bristol Myers Squibb and Exelixis and is a consultant for AstraZeneca, Eisai, Exelixis and Genentech. The other authors declare no competing interests.

Additional information

Peer review information Nature Cancer thanks Tim Greten, Song Liuand 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 Cabozantinib induces changes in plasma correlates.

Concentrations (pg/ml) of VEGF-R2, VEGF-C, VEGF-A, c-MET, Angiopoietin-1, Angiopoietin-2, Tie-2, and AXL in longitudinally obtained plasma samples are shown as line graphs. AXL and c-MET are measured by ELISA assays. All other correlates were measured by Luminex multiplex assays. Data representative of two technical replicates. Each line represents an individual patient. Red and blue lines reflect pathologic non-responders and responders, respectively. Indicated are significant FDR-adjusted P values (<0.05, paired two-tailed t-tests).

Source data

Extended Data Fig. 2 T cell activation is observed in both Nanostring and CyTOF analysis.

T cell activation is observed in both Nanostring and CyTOF analysis. Pre- vs. post-cabozantinib changes in lymphoid cells, granzyme expression, and expression of IL2, IFNγ, PD1 in T cells are shown for two patients for whom the data was available. Y-axis represents a non-universal unit scale (expression levels for Nanostring and abundance levels or metal intensities for CyTOF) log2-transformed for ease of visualization. Abbreviation: N, naïve; NK, natural killer cells; ns, Nanostring; PB, peripheral blood; TIL, tumor infiltrating leukocytes.

Source data

Extended Data Fig. 3 Immunohistochemistry (IHC) analysis of immune cells.

Immunohistochemistry (IHC) analysis of immune cells. a, Each tissue section was manually annotated in image analysis software HALO™ into non-tumor and tumor regions (live, necrosis). b, Density of CD3, CD8, and CD20 in post-treatment surgical samples in the non-tumor regions by IHC (nonresponder, n = 4 patients; responder, n = 3 patients) (mean ± s.d., all P > 0.05, unpaired two-tailed t-test). c, Representative staining results of baseline core biopsies (PRE) and post-treatment surgical samples (POST) for CD3, CD8, and CD20 IHC (one patient selected from each group, representative of n = 3 nonresponder, n = 2 responder paired patient samples; quantitative data shown in panel b).

Source data

Extended Data Fig. 4 Workflow for imaging mass cytometry (IMC).

A tissue microarray of 37 cores was constructed from 12 total patients who underwent surgery after neoadjuvant cabozantinib and nivolumab. 5 patients were pathologic responders and 7 patients were nonresponders. This tissue microarray was then stained with a cocktail of metal-conjugated antibodies against 27 markers. Metal intensities from the stained slide are acquired by Hyperion™. The resulting images were evaluated using MCD Viewer™ and then segmented into single cell data using Ilastik and CellProfiler.

Extended Data Fig. 5 Representative IMC data.

a, Representative multicolored images for every core in the tissue microarray constructed from post-treatment surgical samples for aSMA, CD4, CD20, CD8a, and DNA, stratified by response in one set, and aSMA, Vimentin, CD16, E-Cadherin, CD68, and Ki-67, stratified by response in another. b, Abundances of major immune cell types assessed by IMC across the three representative cores from non-responder (NR) and responder (R) patient samples are shown.

Source data

Extended Data Fig. 6 Combination of cabozantinib and nivolumab promotes local T cell responses.

a, Six post-treatment surgical samples were enzymatically dissociated into single-cell tumor infiltrating leukocytes and were assayed by a 28-marker CyTOF panel dedicated to phenotyping T cells. FlowSOM algorithm was employed to generate 40 metaclusters which were annotated into 21 final clusters. Scaled expression profile for each of the clusters are shown in the heatmap and UMAP. b, Stacked bar plots show immune cell subtype distribution at the single cell level for non-responder (NR, n = 79939 cells) and responder (R, n = 25086 cells) samples. ***P < 2.2e-16 (Chi-squared). c, Abundance of each subtype as a percentage of CD45 cells for each patient sample. Abbreviations: DNT, double-negative T; DPT, double-positive T; EFF, effector; EM, effector memory; EX, exhaustion marker positive; N, naïve; NK, natural killer; UA, unassigned. d, Comparison of CD8 T cell quantification by CyTOF, IMC, and IHC.

Source data

Extended Data Fig. 7 Combination of cabozantinib and nivolumab promotes systemic T cell responses.

a, PBMCs from eight paired pre- and post-treatment samples were assayed by a 28-marker CyTOF panel dedicated to phenotyping T cells. FlowSOM algorithm was employed to generate 30 metaclusters which were then annotated into 23 final clusters. Scaled expression profile for each of the clusters are shown in the heatmap. b, Radar plot showing average fold changes for the abundance of every T cell subtype. Color legends apply to both panels A and B. P-values (edgeR) are annotated. Abbreviations: CM, central memory, DNT, double-negative T; DPT, double-positive T; Eff, effector; EM, effector memory; EX, exhaustion marker positive; N, naïve; NK, natural killer; UA, unassigned.

Source data

Extended Data Fig. 8 Treatment alters levels of immunomodulatory chemokines in plasma.

Concentrations (pg/ml) of MIG (CXCL9), IP-10 (CXCL10), I-TAC (CXCL11), MCP-1 (CCL2), Eotaxin-3 (CCL26), Rantes (CCL5), MCP-2 (CCL8), and MIP-1a (CCL3) in longitudinally obtained plasma samples are shown as line graphs. All correlates were measured by Luminex multiplex assays. Data representative of two technical replicates. Each line represents an individual patient. Red and blue lines reflect pathologic non-responders and responders, respectively. All comparisons not statistically significant by FDR-adjusted P values (<0.05 considered significant, paired two-tailed t-tests).

Source data

Extended Data Fig. 9 B cells indirectly contribute to antitumor immune response.

a, Results from immunohistochemistry of CD138 in nonresponders (NR) and responders (R) for tumor regions (left; NR, n = 7; R, n = 5 patients) and non-tumor regions (right; NR, n = 4; R, n = 5). Data represented as mean±s.d.; P values based on unpaired two-tailed t-test. b, Representative dual CD20-IgA staining result of a responder patient (left) and a positive control tonsil tissue (right). Image selected from one of five responder patient. c, Violin plots of per-cell metal intensities for CCR7, TNFα, IL2, and IFNγ in NR vs. R samples measured by CyTOF (NR, n = 2789 cells; R, n = 1593 cells). Indicated are FDR-adjusted P values (linear modeling). d, Violin plot (left) of per-cell granzyme B (GZMB) metal intensity in NR (n = 2789 cells) vs. R (n = 1593 cells). FDR-adjusted P value (linear modeling). Representative multicolored image of IMC in a responder core exhibiting a tertiary lymphoid aggregate with a prominent focus of B cells along with CD3, HLADR, and granzyme B expression. Image selected from one of 15 responder cores.

Source data

Supplementary information

Source data

Source Data Fig. 1

Statistical source data.

Source Data Fig. 2

Statistical source data.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 4

Statistical source data.

Source Data Fig. 5

Statistical source data.

Source Data Extended Data Fig. 1

Statistical source data.

Source Data Extended Data Fig. 2

Statistical source data.

Source Data Extended Data Fig. 3

Statistical source data.

Source Data Extended Data Fig. 5

Statistical source data.

Source Data Extended Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 7

Statistical source data.

Source Data Extended Data Fig. 8

Statistical source data.

Source Data Extended Data Fig. 9

Statistical source data.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Ho, W.J., Zhu, Q., Durham, J. et al. Neoadjuvant cabozantinib and nivolumab convert locally advanced hepatocellular carcinoma into resectable disease with enhanced antitumor immunity. Nat Cancer 2, 891–903 (2021). https://doi.org/10.1038/s43018-021-00234-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s43018-021-00234-4

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