Molecular correlates of response to eribulin and pembrolizumab in hormone receptor-positive metastatic breast cancer

Tanya E Keenan, Jennifer L Guerriero, Romualdo Barroso-Sousa, Tianyu Li, Tess O'Meara, Anita Giobbie-Hurder, Nabihah Tayob, Jiani Hu, Mariano Severgnini, Judith Agudo, Ines Vaz-Luis, Leilani Anderson, Victoria Attaya, Jihye Park, Jake Conway, Meng Xiao He, Brendan Reardon, Erin Shannon, Gerburg Wulf, Laura M Spring, Rinath Jeselsohn, Ian Krop, Nancy U Lin, Ann Partridge, Eric P Winer, Elizabeth A Mittendorf, David Liu, Eliezer M Van Allen, Sara M Tolaney, Tanya E Keenan, Jennifer L Guerriero, Romualdo Barroso-Sousa, Tianyu Li, Tess O'Meara, Anita Giobbie-Hurder, Nabihah Tayob, Jiani Hu, Mariano Severgnini, Judith Agudo, Ines Vaz-Luis, Leilani Anderson, Victoria Attaya, Jihye Park, Jake Conway, Meng Xiao He, Brendan Reardon, Erin Shannon, Gerburg Wulf, Laura M Spring, Rinath Jeselsohn, Ian Krop, Nancy U Lin, Ann Partridge, Eric P Winer, Elizabeth A Mittendorf, David Liu, Eliezer M Van Allen, Sara M Tolaney

Abstract

Immune checkpoint inhibitors (ICIs) have minimal therapeutic effect in hormone receptor-positive (HR+ ) breast cancer. We present final overall survival (OS) results (n = 88) from a randomized phase 2 trial of eribulin ± pembrolizumab for patients with metastatic HR+ breast cancer, computationally dissect genomic and/or transcriptomic data from pre-treatment tumors (n = 52) for molecular associations with efficacy, and identify cytokine changes differentiating response and ICI-related toxicity (n = 58). Despite no improvement in OS with combination therapy (hazard ratio 0.95, 95% CI 0.59-1.55, p = 0.84), immune infiltration and antigen presentation distinguished responding tumors, while tumor heterogeneity and estrogen signaling independently associated with resistance. Moreover, patients with ICI-related toxicity had lower levels of immunoregulatory cytokines. Broadly, we establish a framework for ICI response in HR+ breast cancer that warrants diagnostic and therapeutic validation. ClinicalTrials.gov Registration: NCT03051659.

Conflict of interest statement

R.B.-S. has served as an advisor/consultant to Eli Lilly and has received honoraria from Roche for participation in Speakers Bureau. J.L.G. is a consultant for GlaxoSmithKline (GSK), Array BioPharma, Codagenix, Verseau, and Kymera and receives sponsored research support from GSK, Eli Lilly, and Array BioPharma. I.V.L. has received institutional honoraria from Pfizer, AstraZeneca, and Amgen. M.X.H. has been a consultant to Amplify Medicines and Ikena Oncology. L.M.S. has been a consultant/advisor for Novartis and Avrobio. N.U.L. has received institutional research funding from Genentech, Cascadian Therapeutics, Array Biopharma, Seattle Genetics, Novartis, Merck, and Pfizer and has been a consultant/advisor to Seattle Genetics, Puma, and Daichii Sankyo. E.P.W. receives consulting fees from InfiniteMD and Leap Therapeutics, honoraria from Genentech, Roche, Tesaro, Lilly, and institutional research funding from Genentech. E.A.M. reports personal financial interests: research support for lab from GlaxoSmithKline; honoraria from Physician Education Resource; compensated service on Scientific Advisory Boards for AstraZeneca, Exact Sciences (formerly Genomic Health), Merck, Peregrine Pharmaceuticals, Roche/Genentech, Sellas Lifesciences, TapImmune Inc; uncompensated service on Steering Committees for BMS, Lilly, Roche/Genentech. E.A.M. reports institutional financial interests from MD Anderson: clinical trial funding from AstraZeneca, EMD Serono, Galena Biopharma, Roche/Genentech; and institutional financial interests from DFCI: clinical trial funding from Roche/Genentech (via SU2C grant). E.M.V.A. serves as a consultant/advisor to Tango Therapeutics, Invitae, Genome Medical, Dynamo, Foresite Capital, and Illumina; holds research support from Novartis and Bristol-Myers Squibb; and holds equity in Synapse, Genome Medical, Tango, and Microsoft Corp. S.M.T. receives institutional research funding from AstraZeneca, Lilly, Merck, Nektar, Novartis, Pfizer, Genentech/Roche, Immunomedics, Exelixis, Bristol-Myers Squibb, Eisai, Nanostring, Cyclacel, Odonate, and Seattle Genetics; has served as an advisor/consultant to AstraZeneca, Lilly, Merck, Nektar, Novartis, Pfizer, Genentech/Roche, Immunomedics, Bristol-Myers Squibb, Eisai, Nanostring, Puma, Sanofi, Celldex, Paxman, Puma, Silverback Therapeutics, G1 Therapeutics, AbbVie, Anthenex, OncoPep, Outcomes4Me, Kyowa Kirin Pharmaceuticals, Daiichi-Sankyo, and Samsung Bioepsis Inc. The remaining authors declare no competing interests.

© 2021. The Author(s).

Figures

Fig. 1. Cohort clinical and genomic characteristics…
Fig. 1. Cohort clinical and genomic characteristics and tumor mutation burden (TMB).
a, b Updated overall survival curves for eribulin ± pembrolizumab in metastatic TNBC show no difference in all patients (a) and in patients with PD-L1 + tumors (b). c Comutation plot shows no association between TMB and response. Each column represents a tumor. Tumors are ordered by RECIST response, and within each response subgroup by decreasing nonsynonymous (Nonsyn) mutational load (top row). Nonsynonymous mutational burden is further subdivided into clonal (dark blue) and subclonal (light blue) mutational load. Tumor purity is the inferred proportion of the tumor sample that is from cancer cells compared to other cell types (Methods). The protocol therapy and biopsy timing (archival primary or metastatic tumor vs. baseline metastatic tumor collected during trial screening) are indicated. Mutational signatures (sig) indicate the dominant signature present at the highest relative proportion and the presence or absence of APOBEC signatures consisting of COSMIC signatures 2 or 13,. Mutations and copy number alterations in genes commonly mutated in breast cancer are shown for each tumor. d, e In tumors treated with eribulin and pembrolizumab, nonsynonymous mutational burden was not different in patients with clinical benefit (green) vs. those without clinical benefit (yellow) (d) and did not differ by RECIST response (e). Unadjusted two-sided Mann–Whitney–Wilcoxon p-values are shown. Boxplot limits indicate the interquartile range (IQR; 25th to 75th percentile), with a center line indicating the median. Whiskers show the value ranges up to 1.5 × IQR above the 75th or below the 25th percentile with outliers beyond those ranges shown as individual points. Mb, megabase; NE, not evaluable; PD, progressive disease; PR, partial response; SD, stable disease; TNBC, triple-negative breast cancer.
Fig. 2. Tumor heterogeneity and purity correlate…
Fig. 2. Tumor heterogeneity and purity correlate with resistance to eribulin and pembrolizumab.
ac Focused analysis of eribulin pembrolizumab WES cohort (n = 27): a Tumor ploidy, defined as the overall genomic copy number (a normal diploid cell has a copy number of 2; Methods) was not different in patients with clinical benefit (green) versus those without clinical benefit (yellow) or by RECIST response. b Tumor heterogeneity, defined as the proportion of subclonal mutations in each tumor (Methods), was lower in patients with clinical benefit (green) versus those without clinical benefit (yellow) and was lower in patients with partial response (PR, green) vs. progressive disease (PD, yellow). c Tumor purity, defined as the proportion of DNA from tumor versus other cells in the sample (Methods), was lower in patients with clinical benefit (green) versus those without clinical benefit (yellow) and was lower in patients with partial response (PR, green) vs. progressive disease (PD, yellow). Unadjusted two-sided Mann-Whitney-Wilcoxon p values are shown. Boxplot limits indicate the interquartile range (IQR; 25th to 75th percentile), with a center line indicating the median. Whiskers show the value ranges up to 1.5 × IQR above the 75th or below the 25th percentile with outliers beyond those ranges shown as individual points. d, e In the overall WES cohort treated with eribulin ± pembrolizumab (n = 50), patients with tumors that had top quartile heterogeneity (d) and purity (e) had longer progression-free survival. Unadjusted two-sided log rank p-values are shown. NE, not evaluable; SD, stable disease; WES, whole exome sequencing.
Fig. 3. Immune gene set enrichment correlates…
Fig. 3. Immune gene set enrichment correlates with response to eribulin and pembrolizumab.
a For tumors treated with eribulin and pembrolizumab (pembro), the top 5 cancer hallmark gene sets (GSEA) enriched in patients with clinical benefit (n = 6) versus those without clinical benefit (n = 8) consisted of immune-related pathways with normalized enrichment scores (NES) > 2.25 and false discovery rate (FDR) q values < 0.001. b For tumors treated with eribulin ± pembrolizumab, the top 5 cancer hallmark gene sets (GSEA) enriched in patients with clinical benefit (n = 11) versus those without clinical benefit (n = 19) also consisted of immune-related pathways with NES > 2.25 and FDR q values < 0.001. c For tumors treated with eribulin alone, the same immune-related gene sets (GSEA) were less enriched in patients with clinical benefit (n = 5) versus those without clinical benefit (n = 11), as these gene sets were not all ranked in the top 5 enriched gene sets, had lower NES down to 1.52, and larger FDR q values up to 0.023. d A heatmap of single-sample gene set enrichment analysis (ssGSEA) score, where each column is a tumor arranged first by clinical benefit (green) versus no clinical benefit (yellow) and then by eribulin pembrolizumab treatment arm (purple) versus eribulin treatment arm (gray), shows that tumors with clinical benefit had enrichment of these immune gene sets. Color indicates the ssGSEA score from least enriched (blue) to most enriched (red). e, f In the overall RNAseq cohort treated with eribulin ± pembrolizumab (n = 30), patients with tumors that had high versus low antigen presentation (AP) gene set enrichment scores (ssGSEA; divided by the median) had longer progression-free (e) and overall survival (f) in unadjusted analyses. All Hallmark pathways and their GSEA enrichment scores are included in Supplementary Data File 1: Supplementary Table 3, and the antigen presentation gene set is shown in Supplementary Data File 1: Supplementary Table 4. Unadjusted two-sided log rank p-values are shown.
Fig. 4. Tumor immune infiltration correlates with…
Fig. 4. Tumor immune infiltration correlates with response to eribulin ± pembrolizumab.
a Absolute immune cell infiltrate inferred by CIBERSORTx was higher in patients with clinical benefit (green) versus those without clinical benefit (yellow) to eribulin + /- pembrolizumab. b Tumor infiltrating lymphocytes, calculated as the sum of all lymphocytes inferred by CIBERSORTx (Methods), were higher in patients with clinical benefit (green) versus those without clinical benefit (yellow) to eribulin ± pembrolizumab. c, d Resting memory CD4 + T cells (c) and follicular helper T cells (d) were higher in patients with clinical benefit (green) versus those without clinical benefit (yellow) to eribulin ± pembrolizumab. e, f M2 macrophages (e) were higher in patients with clinical benefit (green) versus those without clinical benefit (yellow) to eribulin ± pembrolizumab, while M1 macrophages (f) trended towards being higher in patients with clinical benefit (green) with borderline significance (p = 0.05). Unadjusted two-sided Mann–Whitney–Wilcoxon p-values are shown. Boxplot limits indicate the interquartile range (IQR; 25th to 75th percentile), with a center line indicating the median. Whiskers show the value ranges up to 1.5 × IQR above the 75th or below the 25th percentile with outliers beyond those ranges shown as individual points.
Fig. 5. Estrogen signaling correlates with resistance…
Fig. 5. Estrogen signaling correlates with resistance to eribulin and pembrolizumab.
a For tumors treated with eribulin and pembrolizumab (pembro), the top 2 cancer hallmark gene sets (GSEA) enriched in patients without clinical benefit (n = 8) versus those with clinical benefit (n = 6) consisted of estrogen response pathways with normalized enrichment scores (NES) < −1.75 and false discovery rate (FDR) q values < 0.01. b For tumors treated with eribulin ± pembrolizumab, the top 2 cancer hallmark gene sets (GSEA) enriched in patients without clinical benefit (n = 19) versus those with clinical benefit (n = 11) consisted of protein secretion and early estrogen response pathways with NES < −1.75 and FDR q values < 0.001. c For tumors treated with eribulin alone, the top cancer hallmark gene sets (GSEA) enriched in patients without clinical benefit (n = 11) versus those with clinical benefit (n = 5) was the protein secretion pathway. d A heatmap of single-sample GSEA (ssGSEA) score, where each column is a tumor arranged first by clinical benefit (green) versus no clinical benefit (yellow) and then by eribulin pembrolizumab treatment arm (purple) versus eribulin treatment arm (gray), shows that tumors with no clinical benefit had enrichment of these gene sets, particularly estrogen response signaling. Color indicates the ssGSEA score from least enriched (blue) to most enriched (red). e, f In the overall RNAseq cohort treated with eribulin ± pembrolizumab (n = 30), patients with tumors that had top quartile early estrogen response (ER) gene set enrichment scores (ssGSEA) had shorter progression-free (e) and overall survival (f) in unadjusted analyses. Unadjusted two-sided log rank p values are shown. All Hallmark pathways and their GSEA enrichment scores are shown in Supplementary Data File 1: Supplementary Table 3.
Fig. 6. Response correlate interplay and cytokine…
Fig. 6. Response correlate interplay and cytokine analyses.
a Spearman’s correlation coefficients between genomic and transcriptomic features associated with response and b hierarchical clustering of these correlation coefficients. Color indicates the Spearman’s correlation between features, from perfect negative correlation (Spearman’s r = −1, blue) to perfect positive correlation (Spearman’s r = 1, red). Tumor infiltrating lymphocytes (TILs), immune infiltrate, and antigen presentation single-sample gene set enrichment analysis (ssGSEA) score all clustered together, while tumor purity negatively correlated with this cluster. Tumor heterogeneity and estrogen response ssGSEA score were independent from the immune-infiltrated cluster and tumor purity. The sample size for each correlation depended on the number of available data points: correlations involving exclusively genomic or transcriptomic data had n = 50 or 30 tumor samples, respectively, whereas correlations involving genomic and transcriptomic features had n = 28 tumor samples with data available. c Schematic representation of potential interplay of genomic and transcriptomic features. Antigen presentation may increase responses in non-immune infiltrated tumors, while heterogeneity and estrogen signaling may reduce responses in immune-infiltrated tumors. d Heatmap of median pre- to on-treatment fold changes in cytokines across three immune-related toxicity groups: patients treated with (1) eribulin or (2) eribulin and pembrolizumab with no immune-related toxicity versus patients treated with (3) eribulin and pembrolizumab with immune-related toxicity. Shown are unadjusted two-sided Mann–Whitney–Wilcoxon p-values for significant fold-change (FC) differences between adjacent groups. The sample size for each group depended on the number of available data points and ranged from 15–19 patients as indicated in Supplementary Data File 1: Supplementary Tables 10–11. AP, antigen presentation; ER, estrogen response; TMB, tumor mutational burden; tox, toxicity.

References

    1. Fan W, Chang J, Fu P. Endocrine therapy resistance in breast cancer: current status, possible mechanisms and overcoming strategies. Future Med. Chem. 2015;7:1511–1519. doi: 10.4155/fmc.15.93.
    1. Osborne CK, Schiff R. Mechanisms of endocrine resistance in breast cancer. Annu. Rev. Med. 2011;62:233–247. doi: 10.1146/annurev-med-070909-182917.
    1. Twelves C, et al. Subgroup analyses from a phase 3, open-label, randomized study of eribulin mesylate versus capecitabine in pretreated patients with advanced or metastatic breast cancer. Breast Cancer. 2016;10:77–84.
    1. Dirix LY, et al. Avelumab, an anti-PD-L1 antibody, in patients with locally advanced or metastatic breast cancer: a phase 1b JAVELIN Solid Tumor study. Breast Cancer Res. Treat. 2018;167:671–686. doi: 10.1007/s10549-017-4537-5.
    1. Rugo HS, et al. Safety and antitumor activity of pembrolizumab in patients with estrogen receptor-positive/human epidermal growth factor receptor 2-negative advanced breast cancer. Clin. Cancer Res. 2018;24:2804–2811. doi: 10.1158/1078-0432.CCR-17-3452.
    1. Tolaney SM, et al. Effect of eribulin with or without pembrolizumab on progression-free survival for patients with hormone receptor-positive, ERBB2-negative metastatic breast cancer: a randomized clinical trial. JAMA Oncol. 2020;6:1598–1605. doi: 10.1001/jamaoncol.2020.3524.
    1. Keenan TE, Burke KP, Van Allen EM. Genomic correlates of response to immune checkpoint blockade. Nat. Med. 2019;25:389–402. doi: 10.1038/s41591-019-0382-x.
    1. Schmid P, et al. Atezolizumab and nab-paclitaxel in advanced triple-negative breast cancer. N. Engl. J. Med. 2018;379:2108–2121. doi: 10.1056/NEJMoa1809615.
    1. Sobral-Leite M, et al. Assessment of PD-L1 expression across breast cancer molecular subtypes, in relation to mutation rate, BRCA1-like status, tumor-infiltrating immune cells and survival. Oncoimmunology. 2018;7:e1509820. doi: 10.1080/2162402X.2018.1509820.
    1. Le DT, et al. Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade. Science. 2017;357:409–413. doi: 10.1126/science.aan6733.
    1. Barroso-Sousa R, et al. Prevalence and mutational determinants of high tumor mutation burden in breast cancer. Ann. Oncol. 2020;31:387–394. doi: 10.1016/j.annonc.2019.11.010.
    1. Barroso-Sousa R, et al. Tumor mutational burden and PTEN alterations as molecular correlates of response to PD-1/L1 blockade in metastatic triple-negative breast cancer. Clin. Cancer Res. 2020;26:2565–2572. doi: 10.1158/1078-0432.CCR-19-3507.
    1. Denkert C, et al. Tumour-infiltrating lymphocytes and prognosis in different subtypes of breast cancer: a pooled analysis of 3771 patients treated with neoadjuvant therapy. Lancet Oncol. 2018;19:40–50. doi: 10.1016/S1470-2045(17)30904-X.
    1. Ayers M, et al. IFN-gamma-related mRNA profile predicts clinical response to PD-1 blockade. J. Clin. Invest. 2017;127:2930–2940. doi: 10.1172/JCI91190.
    1. Terranova-Barberio M, et al. Exhausted T cell signature predicts immunotherapy response in ER-positive breast cancer. Nat. Commun. 2020;11:3584. doi: 10.1038/s41467-020-17414-y.
    1. Nanda R, et al. Effect of pembrolizumab plus neoadjuvant chemotherapy on pathologic complete response in women with early-stage breast cancer: an analysis of the ongoing phase 2 adaptively randomized I-SPY2 trial. JAMA Oncol. 2020;6:676–684. doi: 10.1001/jamaoncol.2019.6650.
    1. Liu D, et al. Integrative molecular and clinical modeling of clinical outcomes to PD1 blockade in patients with metastatic melanoma. Nat. Med. 2019;25:1916–1927. doi: 10.1038/s41591-019-0654-5.
    1. Althouse AD. Adjust for multiple comparisons? It’s not that simple. Ann. Thorac. Surg. 2016;101:1644–1645. doi: 10.1016/j.athoracsur.2015.11.024.
    1. Ranganathan P, Pramesh CS, Buyse M. Common pitfalls in statistical analysis: the perils of multiple testing. Perspect. Clin. Res. 2016;7:106–107. doi: 10.4103/2229-3485.179436.
    1. Cancer Genome Atlas, N. Comprehensive molecular portraits of human breast tumours. Nature. 2012;490:61–70. doi: 10.1038/nature11412.
    1. Bertucci F, et al. Genomic characterization of metastatic breast cancers. Nature. 2019;569:560–564. doi: 10.1038/s41586-019-1056-z.
    1. Alexandrov LB, et al. Signatures of mutational processes in human cancer. Nature. 2013;500:415–421. doi: 10.1038/nature12477.
    1. Parker JS, et al. Supervised risk predictor of breast cancer based on intrinsic subtypes. J. Clin. Oncol. 2009;27:1160–1167. doi: 10.1200/JCO.2008.18.1370.
    1. Eisenhauer EA, et al. New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1) Eur. J. Cancer. 2009;45:228–247. doi: 10.1016/j.ejca.2008.10.026.
    1. Samstein RM, et al. Tumor mutational load predicts survival after immunotherapy across multiple cancer types. Nat. Genet. 2019;51:202–206. doi: 10.1038/s41588-018-0312-8.
    1. Miao D, et al. Genomic correlates of response to immune checkpoint blockade in microsatellite-stable solid tumors. Nat. Genet. 2018;50:1271–1281. doi: 10.1038/s41588-018-0200-2.
    1. Subramanian A, et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc. Natl Acad. Sci. USA. 2005;102:15545–15550.
    1. Yoshida T, et al. Eribulin mesilate suppresses experimental metastasis of breast cancer cells by reversing phenotype from epithelial-mesenchymal transition (EMT) to mesenchymal-epithelial transition (MET) states. Br. J. Cancer. 2014;110:1497–1505. doi: 10.1038/bjc.2014.80.
    1. Fermaintt CS, Hastings S, Mooberry SL, Risinger AL. Abstract P5-05-03: Eribulin treatment activates type 1 IFNs to promote a gene expression signature associated with antitumor immunity. Cancer Res. 2020;80:P5-05-03.
    1. Hilligan KL, Ronchese F. Antigen presentation by dendritic cells and their instruction of CD4 + T helper cell responses. Cell. Mol. Immunol. 2020;17:587–599. doi: 10.1038/s41423-020-0465-0.
    1. Mostafa AA, et al. Activation of ERalpha signaling differentially modulates IFN-gamma induced HLA-class II expression in breast cancer cells. PLoS ONE. 2014;9:e87377. doi: 10.1371/journal.pone.0087377.
    1. Tarhini AA, et al. Baseline circulating IL-17 predicts toxicity while TGF-beta1 and IL-10 are prognostic of relapse in ipilimumab neoadjuvant therapy of melanoma. J. Immunother. Cancer. 2015;3:39. doi: 10.1186/s40425-015-0081-1.
    1. Bridge JA, Lee JC, Daud A, Wells JW, Bluestone JA. Cytokines, chemokines, and other biomarkers of response for checkpoint inhibitor therapy in skin cancer. Front. Med. 2018;5:351. doi: 10.3389/fmed.2018.00351.
    1. Groh V, Wu J, Yee C, Spies T. Tumour-derived soluble MIC ligands impair expression of NKG2D and T-cell activation. Nature. 2002;419:734–738. doi: 10.1038/nature01112.
    1. Roda-Navarro P, Vales-Gomez M, Chisholm SE, Reyburn HT. Transfer of NKG2D and MICB at the cytotoxic NK cell immune synapse correlates with a reduction in NK cell cytotoxic function. Proc. Natl Acad. Sci. USA. 2006;103:11258–11263. doi: 10.1073/pnas.0600721103.
    1. Lim SY, et al. Circulating cytokines predict immune-related toxicity in melanoma patients receiving anti-PD-1-based immunotherapy. Clin. Cancer Res. 2019;25:1557–1563. doi: 10.1158/1078-0432.CCR-18-2795.
    1. Khan S, et al. Immune dysregulation in cancer patients developing immune-related adverse events. Br. J. Cancer. 2019;120:63–68. doi: 10.1038/s41416-018-0155-1.
    1. Tolaney SM, et al. Eribulin plus pembrolizumab in patients with metastatic triple-negative breast cancer (ENHANCE 1): a phase 1b/2 study. Clin. Cancer Res. 2021;27:3061–3068. doi: 10.1158/1078-0432.CCR-20-4726.
    1. Perez-Garcia JM, et al. Pembrolizumab plus eribulin in hormone-receptor-positive, HER2-negative, locally recurrent or metastatic breast cancer (KELLY): An open-label, multicentre, single-arm, phase trial. Eur. J. Cancer. 2021;148:382–394. doi: 10.1016/j.ejca.2021.02.028.
    1. Cortes J, et al. Pembrolizumab plus chemotherapy versus placebo plus chemotherapy for previously untreated locally recurrent inoperable or metastatic triple-negative breast cancer (KEYNOTE-355): a randomised, placebo-controlled, double-blind, phase 3 clinical trial. Lancet. 2020;396:1817–1828. doi: 10.1016/S0140-6736(20)32531-9.
    1. Karimian Z, Mavoungou S, Salem JE, Tubach F, Dechartres A. The quality of reporting general safety parameters and immune-related adverse events in clinical trials of FDA-approved immune checkpoint inhibitors. BMC Cancer. 2020;20:1128. doi: 10.1186/s12885-020-07518-5.
    1. Hsiehchen D, Watters MK, Lu R, Xie Y, Gerber DE. Variation in the assessment of immune-related adverse event occurrence, grade, and timing in patients receiving immune checkpoint inhibitors. JAMA Netw. Open. 2019;2:e1911519. doi: 10.1001/jamanetworkopen.2019.11519.
    1. Maher VE, et al. Analysis of the association between adverse events and outcome in patients receiving a programmed death protein 1 or programmed death ligand 1 antibody. J. Clin. Oncol. 2019;37:2730–2737. doi: 10.1200/JCO.19.00318.
    1. Riudavets M, et al. Correlation between immune-related adverse events (irAEs) and efficacy in patients with solid tumors treated with immune-checkpoints inhibitors (ICIs) J. Clin. Oncol. 2018;36:3064–3064. doi: 10.1200/JCO.2018.36.15_suppl.3064.
    1. Giobbie-Hurder A, Gelber RD, Regan MM. Challenges of guarantee-time bias. J. Clin. Oncol. 2013;31:2963–2969. doi: 10.1200/JCO.2013.49.5283.
    1. Friedlander P, et al. A whole-blood RNA transcript-based gene signature is associated with the development of CTLA-4 blockade-related diarrhea in patients with advanced melanoma treated with the checkpoint inhibitor tremelimumab. J. Immunother. Cancer. 2018;6:90. doi: 10.1186/s40425-018-0408-9.
    1. Liu D, et al. Mutational patterns in chemotherapy resistant muscle-invasive bladder cancer. Nat. Commun. 2017;8:2193. doi: 10.1038/s41467-017-02320-7.
    1. McGranahan N, et al. Clonal neoantigens elicit T cell immunoreactivity and sensitivity to immune checkpoint blockade. Science. 2016;351:1463–1469. doi: 10.1126/science.aaf1490.
    1. Morris LG, et al. Pan-cancer analysis of intratumor heterogeneity as a prognostic determinant of survival. Oncotarget. 2016;7:10051–10063. doi: 10.18632/oncotarget.7067.
    1. Spranger S, Bao R, Gajewski TF. Melanoma-intrinsic beta-catenin signalling prevents anti-tumour immunity. Nature. 2015;523:231–235. doi: 10.1038/nature14404.
    1. Gainor JF, et al. EGFR mutations and ALK rearrangements are associated with low response rates to PD-1 pathway blockade in non-small cell lung cancer: a retrospective analysis. Clin. Cancer Res. 2016;22:4585–4593. doi: 10.1158/1078-0432.CCR-15-3101.
    1. Lee CK, et al. Clinical and molecular characteristics associated with survival among patients treated with checkpoint inhibitors for advanced non-small cell lung carcinoma: a systematic review and meta-analysis. JAMA Oncol. 2018;4:210–216. doi: 10.1001/jamaoncol.2017.4427.
    1. Peng W, et al. Loss of PTEN promotes resistance to T cell-mediated immunotherapy. Cancer Disco. 2016;6:202–216. doi: 10.1158/-15-0283.
    1. George S, et al. Loss of PTEN is associated with resistance to anti-PD-1 checkpoint blockade therapy in metastatic uterine leiomyosarcoma. Immunity. 2017;46:197–204. doi: 10.1016/j.immuni.2017.02.001.
    1. Skoulidis F, et al. STK11/LKB1 mutations and PD-1 inhibitor resistance in KRAS-mutant lung adenocarcinoma. Cancer Discov. 2018;8:822–835. doi: 10.1158/-18-0099.
    1. Prat A, et al. Immune-related gene expression profiling after PD-1 blockade in non-small cell lung carcinoma, head and neck squamous cell carcinoma, and melanoma. Cancer Res. 2017;77:3540–3550. doi: 10.1158/0008-5472.CAN-16-3556.
    1. Danaher P, et al. Pan-cancer adaptive immune resistance as defined by the tumor inflammation signature (TIS): results from The Cancer Genome Atlas (TCGA) J. Immunother. Cancer. 2018;6:63. doi: 10.1186/s40425-018-0367-1.
    1. Zaretsky JM, et al. Mutations associated with acquired resistance to PD-1 blockade in melanoma. N. Engl. J. Med. 2016;375:819–829. doi: 10.1056/NEJMoa1604958.
    1. Sade-Feldman M, et al. Resistance to checkpoint blockade therapy through inactivation of antigen presentation. Nat. Commun. 2017;8:1136. doi: 10.1038/s41467-017-01062-w.
    1. Gettinger S, et al. Impaired HLA class I antigen processing and presentation as a mechanism of acquired resistance to immune checkpoint inhibitors in lung cancer. Cancer Discov. 2017;7:1420–1435. doi: 10.1158/-17-0593.
    1. Cristescu R, et al. Pan-tumor genomic biomarkers for PD-1 checkpoint blockade-based immunotherapy. Science. 2018;362:eaar3593. doi: 10.1126/science.aar3593.
    1. Rodig SJ, et al. MHC proteins confer differential sensitivity to CTLA-4 and PD-1 blockade in untreated metastatic melanoma. Sci. Transl. Med. 2018;10:eaar3342. doi: 10.1126/scitranslmed.aar3342.
    1. Sobral-Leite M, et al. Cancer-immune interactions in ER-positive breast cancers: PI3K pathway alterations and tumor-infiltrating lymphocytes. Breast Cancer Res. 2019;21:90. doi: 10.1186/s13058-019-1176-2.
    1. Chung YR, Kim HJ, Jang MH, Park SY. Prognostic value of tumor infiltrating lymphocyte subsets in breast cancer depends on hormone receptor status. Breast Cancer Res. Treat. 2017;161:409–420. doi: 10.1007/s10549-016-4072-9.
    1. Stanton SE, Disis ML. Clinical significance of tumor-infiltrating lymphocytes in breast cancer. J. Immunother. Cancer. 2016;4:59. doi: 10.1186/s40425-016-0165-6.
    1. Wagner J, et al. A single-cell atlas of the tumor and immune ecosystem of human breast cancer. Cell. 2019;177:1330–1345 e1318. doi: 10.1016/j.cell.2019.03.005.
    1. Guerriero JL. Macrophages: the road less traveled, changing anticancer therapy. Trends Mol. Med. 2018;24:472–489. doi: 10.1016/j.molmed.2018.03.006.
    1. Sinn BV, et al. Human leucocyte antigen class I in hormone receptor-positive, HER2-negative breast cancer: association with response and survival after neoadjuvant chemotherapy. Breast Cancer Res. 2019;21:142. doi: 10.1186/s13058-019-1231-z.
    1. Johnson DB, et al. Melanoma-specific MHC-II expression represents a tumour-autonomous phenotype and predicts response to anti-PD-1/PD-L1 therapy. Nat. Commun. 2016;7:10582. doi: 10.1038/ncomms10582.
    1. Yang ZZ, et al. Soluble IL-2Ralpha facilitates IL-2-mediated immune responses and predicts reduced survival in follicular B-cell non-Hodgkin lymphoma. Blood. 2011;118:2809–2820. doi: 10.1182/blood-2011-03-340885.
    1. Shih VF, et al. Homeostatic IL-23 receptor signaling limits Th17 response through IL-22-mediated containment of commensal microbiota. Proc. Natl Acad. Sci. USA. 2014;111:13942–13947. doi: 10.1073/pnas.1323852111.
    1. Luoma AM, et al. Molecular pathways of colon inflammation induced by cancer immunotherapy. Cell. 2020;182:655–671 e622. doi: 10.1016/j.cell.2020.06.001.
    1. Schoenfeld JD, et al. Pneumonitis resulting from radiation and immune checkpoint blockade illustrates characteristic clinical, radiologic and circulating biomarker features. J. Immunother. Cancer. 2019;7:112. doi: 10.1186/s40425-019-0583-3.
    1. Miao D, et al. Genomic correlates of response to immune checkpoint therapies in clear cell renal cell carcinoma. Science. 2018;359:801–806. doi: 10.1126/science.aan5951.
    1. Van der Auwera GA, et al. From FastQ data to high confidence variant calls: the Genome Analysis Toolkit best practices pipeline. Curr. Protoc. Bioinformatics. 2013;43:11.10.11–11.10.33. doi: 10.1002/0471250953.bi1110s43.
    1. Cibulskis K, et al. ContEst: estimating cross-contamination of human samples in next-generation sequencing data. Bioinformatics. 2011;27:2601–2602. doi: 10.1093/bioinformatics/btr446.
    1. Carter SL, et al. Absolute quantification of somatic DNA alterations in human cancer. Nat. Biotechnol. 2012;30:413–421. doi: 10.1038/nbt.2203.
    1. Shen R, Seshan VE. FACETS: allele-specific copy number and clonal heterogeneity analysis tool for high-throughput DNA sequencing. Nucleic Acids Res. 2016;44:e131. doi: 10.1093/nar/gkw520.
    1. Taylor-Weiner A, et al. DeTiN: overcoming tumor-in-normal contamination. Nat. Methods. 2018;15:531–534. doi: 10.1038/s41592-018-0036-9.
    1. Cibulskis K, et al. Sensitive detection of somatic point mutations in impure and heterogeneous cancer samples. Nat. Biotechnol. 2013;31:213–219. doi: 10.1038/nbt.2514.
    1. Costello M, et al. Discovery and characterization of artifactual mutations in deep coverage targeted capture sequencing data due to oxidative DNA damage during sample preparation. Nucleic Acids Res. 2013;41:e67. doi: 10.1093/nar/gks1443.
    1. Saunders CT, et al. Strelka: accurate somatic small-variant calling from sequenced tumor-normal sample pairs. Bioinformatics. 2012;28:1811–1817. doi: 10.1093/bioinformatics/bts271.
    1. Ramos AH, et al. Oncotator: cancer variant annotation tool. Hum. Mutat. 2015;36:E2423–2429. doi: 10.1002/humu.22771.
    1. Rosenthal R, McGranahan N, Herrero J, Taylor BS, Swanton C. DeconstructSigs: delineating mutational processes in single tumors distinguishes DNA repair deficiencies and patterns of carcinoma evolution. Genome Biol. 2016;17:31. doi: 10.1186/s13059-016-0893-4.
    1. Forbes SA, et al. COSMIC: somatic cancer genetics at high-resolution. Nucleic Acids Res. 2017;45:D777–D783. doi: 10.1093/nar/gkw1121.
    1. Olshen AB, Venkatraman ES, Lucito R, Wigler M. Circular binary segmentation for the analysis of array-based DNA copy number data. Biostatistics. 2004;5:557–572. doi: 10.1093/biostatistics/kxh008.
    1. Li B, Dewey CN. RSEM: accurate transcript quantification from RNA-Seq data with or without a reference genome. BMC Bioinformatics. 2011;12:323. doi: 10.1186/1471-2105-12-323.
    1. Dobin A, et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics. 2013;29:15–21. doi: 10.1093/bioinformatics/bts635.
    1. Gendoo DM, et al. Genefu: an R/Bioconductor package for computation of gene expression-based signatures in breast cancer. Bioinformatics. 2016;32:1097–1099. doi: 10.1093/bioinformatics/btv693.
    1. Liberzon A, et al. The Molecular Signatures Database (MSigDB) hallmark gene set collection. Cell Syst. 2015;1:417–425. doi: 10.1016/j.cels.2015.12.004.
    1. Barbie DA, et al. Systematic RNA interference reveals that oncogenic KRAS-driven cancers require TBK1. Nature. 2009;462:108–112. doi: 10.1038/nature08460.
    1. Newman AM, et al. Robust enumeration of cell subsets from tissue expression profiles. Nat. Methods. 2015;12:453–457. doi: 10.1038/nmeth.3337.
    1. Newman AM, et al. Determining cell type abundance and expression from bulk tissues with digital cytometry. Nat. Biotechnol. 2019;37:773–782. doi: 10.1038/s41587-019-0114-2.
    1. Feng F, et al. A computational solution to improve biomarker reproducibility during long-term projects. PLoS ONE. 2019;14:e0209060. doi: 10.1371/journal.pone.0209060.

Source: PubMed

3
Subscribe