Liver Metastasis and Treatment Outcome with Anti-PD-1 Monoclonal Antibody in Patients with Melanoma and NSCLC

The association between metastatic site and responses to anti-PD-1 immunotherapy was explored In both melanoma and lung cancer. Liver metastasis was associated with worse outcome and CD8+ T cell-poor tumors, suggesting a potential mechanism for the outcomes. We explored the association between liver metastases, tumor CD8+ T-cell count, and response in patients with melanoma or lung cancer treated with the anti-PD-1 antibody, pembrolizumab. The melanoma discovery cohort was drawn from the phase I Keynote 001 trial, whereas the melanoma validation cohort was drawn from Keynote 002, 006, and EAP trials and the non–small cell lung cancer (NSCLC) cohort from Keynote 001. Liver metastasis was associated with reduced response and shortened progression-free survival [PFS; objective response rate (ORR), 30.6%; median PFS, 5.1 months] compared with patients without liver metastasis (ORR, 56.3%; median PFS, 20.1 months) P ≤ 0.0001, and confirmed in the validation cohort (P = 0.0006). The presence of liver metastasis significantly increased the likelihood of progression (OR, 1.852; P < 0.0001). In a subset of biopsied patients (n = 62), liver metastasis was associated with reduced CD8+ T-cell density at the invasive tumor margin (liver metastasis+ group, n = 547 ± 164.8; liver metastasis− group, n = 1,441 ± 250.7; P < 0.016). A reduced response rate and shortened PFS was also observed in NSCLC patients with liver metastasis [median PFS, 1.8 months; 95% confidence interval (CI), 1.4–2.0], compared with those without liver metastasis (n = 119, median PFS, 4.0 months; 95% CI, 2.1–5.1), P = 0.0094. Thus, liver metastatic patients with melanoma or NSCLC that had been treated with pembrolizumab were associated with reduced responses and PFS, and liver metastases were associated with reduced marginal CD8+ T-cell infiltration, providing a potential mechanism for this outcome. Cancer Immunol Res; 5(5); 417–24. ©2017 AACR.

[1]  S. Elledge,et al.  Tumor aneuploidy correlates with markers of immune evasion and with reduced response to immunotherapy , 2017, Science.

[2]  J. Wolchok,et al.  Targeting T Cell Co-receptors for Cancer Therapy. , 2016, Immunity.

[3]  Antoni Ribas,et al.  The “cancer immunogram” , 2016, Science.

[4]  J. Wolchok,et al.  Association of Pembrolizumab With Tumor Response and Survival Among Patients With Advanced Melanoma. , 2016, JAMA.

[5]  Y. Shentu,et al.  Pembrolizumab versus docetaxel for previously treated, PD-L1-positive, advanced non-small-cell lung cancer (KEYNOTE-010): a randomised controlled trial , 2016, The Lancet.

[6]  K. Steele,et al.  Safety and antitumour activity of durvalumab plus tremelimumab in non-small cell lung cancer: a multicentre, phase 1b study. , 2016, The Lancet. Oncology.

[7]  Jason B. Williams,et al.  Commensal Bifidobacterium promotes antitumor immunity and facilitates anti–PD-L1 efficacy , 2015, Science.

[8]  C. Rudin,et al.  Nivolumab versus Docetaxel in Advanced Nonsquamous Non-Small-Cell Lung Cancer. , 2015, The New England journal of medicine.

[9]  Wei Zhou,et al.  Pembrolizumab versus investigator-choice chemotherapy for ipilimumab-refractory melanoma (KEYNOTE-002): a randomised, controlled, phase 2 trial. , 2015, The Lancet. Oncology.

[10]  J. Larkin,et al.  Pembrolizumab versus Ipilimumab in Advanced Melanoma. , 2015, The New England journal of medicine.

[11]  J. Wolchok,et al.  Long-term efficacy of pembrolizumab (pembro; MK-3475) in a pooled analysis of 655 patients (pts) with advanced melanoma (MEL) enrolled in KEYNOTE-001. , 2015 .

[12]  A. Daud,et al.  Clinical characteristics predictive of response to pembrolizumab in advanced melanoma. , 2015 .

[13]  J. Lunceford,et al.  Pembrolizumab for the treatment of non-small-cell lung cancer. , 2015, The New England journal of medicine.

[14]  T. Gajewski,et al.  Melanoma-intrinsic β-catenin signalling prevents anti-tumour immunity , 2015, Nature.

[15]  Martin L. Miller,et al.  Mutational landscape determines sensitivity to PD-1 blockade in non–small cell lung cancer , 2015, Science.

[16]  H. Kohrt,et al.  Predictive correlates of response to the anti-PD-L1 antibody MPDL3280A in cancer patients , 2014, Nature.

[17]  R. Emerson,et al.  PD-1 blockade induces responses by inhibiting adaptive immune resistance , 2014, Nature.

[18]  J. Lunceford,et al.  Abstract CT104: Antitumor activity of the anti-PD-1 monoclonal antibody MK-3475 in melanoma(MEL): Correlation of tumor PD-L1 expression with outcome , 2014 .

[19]  Antoni Ribas,et al.  Anti-programmed-death-receptor-1 treatment with pembrolizumab in ipilimumab-refractory advanced melanoma: a randomised dose-comparison cohort of a phase 1 trial , 2014, The Lancet.

[20]  J. Taube,et al.  Association of PD-1, PD-1 Ligands, and Other Features of the Tumor Immune Microenvironment with Response to Anti–PD-1 Therapy , 2014, Clinical Cancer Research.

[21]  P. Kubes,et al.  Immune surveillance by the liver , 2013, Nature Immunology.

[22]  C. Horak,et al.  Nivolumab plus ipilimumab in advanced melanoma. , 2013, The New England journal of medicine.

[23]  Antoni Ribas,et al.  Safety and tumor responses with lambrolizumab (anti-PD-1) in melanoma. , 2013, The New England journal of medicine.

[24]  C. Drake,et al.  Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. , 2012, The New England journal of medicine.

[25]  David C. Smith,et al.  Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. , 2012, The New England journal of medicine.

[26]  Alison P. Klein,et al.  Colocalization of Inflammatory Response with B7-H1 Expression in Human Melanocytic Lesions Supports an Adaptive Resistance Mechanism of Immune Escape , 2012, Science Translational Medicine.

[27]  Drew M. Pardoll,et al.  The blockade of immune checkpoints in cancer immunotherapy , 2012, Nature Reviews Cancer.

[28]  Axel Hoos,et al.  Ipilimumab plus dacarbazine for previously untreated metastatic melanoma. , 2011, The New England journal of medicine.

[29]  Israel Lowy,et al.  Phase I study of single-agent anti-programmed death-1 (MDX-1106) in refractory solid tumors: safety, clinical activity, pharmacodynamics, and immunologic correlates. , 2010, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[30]  R. Pierce,et al.  Hepatitis C virus core protein subverts the antiviral activities of human Kupffer cells. , 2010, Gastroenterology.

[31]  L. Schwartz,et al.  New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1). , 2009, European journal of cancer.

[32]  F. Momburg,et al.  Cross‐presentation of oral antigens by liver sinusoidal endothelial cells leads to CD8 T cell tolerance , 2005, European journal of immunology.

[33]  B. John,et al.  Passive and Active Mechanisms Trap Activated CD8+ T Cells in the Liver1 , 2004, The Journal of Immunology.

[34]  A. Livingstone,et al.  Cutting Edge: CD4+ T Cell Help Can Be Essential for Primary CD8+ T Cell Responses In Vivo 1 , 2003, The Journal of Immunology.

[35]  I. N. Crispe,et al.  Hepatic T cells and liver tolerance , 2003, Nature Reviews Immunology.

[36]  R. Calne Immunological tolerance: the liver effect , 2002, Immunological reviews.

[37]  H. Ljunggren,et al.  Efficient presentation of exogenous antigen by liver endothelial cells to CD8+ T cells results in antigen-specific T-cell tolerance , 2000, Nature Medicine.

[38]  Wei Li,et al.  Apoptosis within spontaneously accepted mouse liver allografts: evidence for deletion of cytotoxic T cells and implications for tolerance induction. , 1997, Journal of immunology.

[39]  R. Calne Mechanisms in the acceptance of organ grafts. , 1976, British medical bulletin.

[40]  R. Binns,et al.  Induction of Immunological Tolerance by Porcine Liver Allografts , 1969, Nature.

[41]  HARVEY M. CANTOR,et al.  Hepatic Suppression of Sensitization to Antigen absorbed into the Portal System , 1967, Nature.