Tumor immune microenvironment and genomic evolution in a patient with metastatic triple negative breast cancer and a complete response to atezolizumab

Luciana Molinero, Yijin Li, Ching-Wei Chang, Sophia Maund, Maureen Berg, Jeanne Harrison, Marcella Fassò, Carol O'Hear, Priti Hegde, Leisha A Emens, Luciana Molinero, Yijin Li, Ching-Wei Chang, Sophia Maund, Maureen Berg, Jeanne Harrison, Marcella Fassò, Carol O'Hear, Priti Hegde, Leisha A Emens

Abstract

Background: Metastatic TNBC (mTNBC) has a poor prognosis and few treatment options. The anti-PD-L1 antibody atezolizumab demonstrated clinical activity in mTNBC patients with PD-L1-positive tumor-infiltrating immune cells. The current study describes the tumor immune microenvironment (TiME) and genomic evolution across sequential therapies in a patient with a 31-year history of TNBC and a complete response (CR) to atezolizumab monotherapy.

Materials and methods: In 1986, the patient had surgery and radiotherapy (XRT) for newly diagnosed TNBC, followed by surgery and adjuvant chemotherapy for two locoregional recurrences. She developed mTNBC in 2009 and was sequentially treated with capecitabine, gemcitabine-carboplatin-iniparib (GCI), XRT and an experimental vaccine. She experienced disease progression (PD) to all these therapies. In 2013, she had a PD-L1 positive tumor and enrolled in a phase 1 atezolizumab monotherapy study (PCD4989g; NCT01375842). She received atezolizumab for 1 year with initial pseudo-progression followed by a partial response. After 1 year without treatment she experienced PD, reinitiated atezolizumab and subsequently achieved CR. Tumor specimens were collected at numerous times between 2008 and 2015 and assessed by immunohistochemistry, RNA-seq and DNA-seq.

Results: TiME biomarkers, including CD8, ICs and PD-L1 on IC, increased after capecitabine and remained high after GCI, XRT and through pseudo-progression on atezolizumab. At PD post-atezolizumab exposure, TiME biomarkers decreased but PD-L1 status remained positive. Immune-related RNA signatures confirmed these findings. TNBC subtyping revealed evolution from luminal androgen receptor (LAR) to basal-like immune activated (BLIA). Genomic profiling showed truncal alterations in RB1 and TP53, while the presence of other genomic alterations varied over time. Tumor mutational burden peaked after XRT and declined after atezolizumab exposure.

Conclusions: This case report describes the evolution of TiME and TNBC molecular subtypes/genomics over time with sequential therapies in a TNBC patient with a CR to atezolizumab monotherapy. These data suggest the TiME is pliable and may be manipulated to maximize response to immunotherapy (NCT01375842, https://ichgcp.net/clinical-trials-registry/NCT01375842?term=NCT01375842&rank=1 ).

Keywords: Atezolizumab; PD-L1; Triple negative breast cancer (TNBC).

Conflict of interest statement

LM, YL, C-WC, SM, MF, CO, and PH are employees of Genentech, Incorporated. LAE receives research funding from Genentech, Incorporated and Hoffman-La Roche, Incorporated.

Figures

Fig. 1
Fig. 1
Clinical course of disease and time-points of collected tumor biopsies. On the right: chronological schema of disease appearance and treatments, on years. On the left: time of sample collection (red dots) on days respect to initiation to atezolizumab first exposure. CMF: cyclophosphamide, methotrexate and fluorouracil; Biopsy Dx: diagnostic biopsy; LAD: lymphadenopathy
Fig. 2
Fig. 2
Change in tumor burden and circulating biomarkers after atezolizumab exposures. Changes in sum of lesion diameter (SLD) was assessed over time in initial (a) and second (b) exposure to atezolizumab. Plasma tumor antigens CEA and CA27–29 (c), circulating T, B and NK lymphocytes (d) and the cytokines IL18, CXCL10 and granzyme A (GZMA)) (e) were assessed over time during the first exposure to atezolizumab. Upper limit of normal levels for CEA (3 ng/ml) and CA27–29 (38 U/ml) are indicated by the dotted lines (blue: CA27-29; red: CEA). Changes in lymphocyte populations were plotted as ratio to baseline to pre-treatment values
Fig. 3
Fig. 3
Evolution of Tumor Microenvironment. Samples collected over time. Images at 100X (a) of tumor infiltrating immune cells (ICs), PDL1 in tumor cells and tumor infiltrating immune cells, CD8 T cells and CD163 macrophages were assessed by hematoxylin& eosin or immunostaining. b Quantification of the parameters in (A) are displayed as a percentage of tumor area and evaluated over time with respect to atezolizumab first exposure. c RNA-based signatures associated with T cells, regulatory T cells, CD8 effector T cells, NK cells, B cells, macrophages, immune checkpoints, cancer associated fibroblasts, cytolytic activity, antigen processing, angiogenesis, and proliferation were derived from RNA-Seq and plotted as PC1 scores and displayed over time. As reference, the aggregated value for the samples from the TNBC cohort in the PCD4989g study (PCD, all) is displayed as box plots representing median, 25th and 75th percentiles and the vertical bars represent range (maximum and minimum). d TNBC subtype classifiers were derived from RNA-Seq for each sample. Heatmap denotes relative RNA expression of genes involved in the subtype classifiers in the analyzed samples. BLIA and BLIS prob.: probability that the samples are BLIA or BLIS
Fig. 4
Fig. 4
Characterization of Genomic Landscape Over Time. Samples collected pre- and post-atezolizumab exposure were tested with the FoundationOne® targeted NGS assay. Upper panel: genes with detected single nucleotide variants (SNV). Mutant allele frequencies (MAF) are shown for each specimen. Asterisk (*) indicates that the variant was present at a frequency below the validated reporting threshold. Light gray: predicted somatic mutations, dark gray: predicted germline mutations; Bold: predicted subclonal somatic mutations. Middle panel: genes with detected copy number alterations (CNAs). Numbers indicate the number of copies detected. Asterisk (*) indicates that low-level amplifications were detected below the validated reporting threshold of > 5 copies. No homozygous deletions were observed. Lower panel: tumor mutational burden (TMB) indicated as mutations per megabase

References

    1. Gadi VK, Davidson NE. Practical approach to triple-negative breast Cancer. J Oncol Pract. 2017;13:293–300. doi: 10.1200/JOP.2017.022632.
    1. den Brok WD, Speers CH, Gondara L, Baxter E, Tyldesley SK, Lohrisch CA. Survival with metastatic breast cancer based on initial presentation, de novo versus relapsed. Breast Cancer Res Treat. 2017;161:549–556. doi: 10.1007/s10549-016-4080-9.
    1. Schmid P, Adams S, Rugo HS, Schneeweiss A, Barrios CH, Iwata H, 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. Savas P, Salgado R, Denkert C, Sotiriou C, Darcy PK, Smyth MJ, et al. Clinical relevance of host immunity in breast cancer: from TILs to the clinic. Nat Rev Clin Oncol. 2016;13:228–241. doi: 10.1038/nrclinonc.2015.215.
    1. Adams S, Gray RJ, Demaria S, Goldstein L, Perez EA, Shulman LN, et al. Prognostic value of tumor-infiltrating lymphocytes in triple-negative breast cancers from two phase III randomized adjuvant breast cancer trials: ECOG 2197 and ECOG 1199. J Clin Oncol. 2014;32:2959–2966. doi: 10.1200/JCO.2013.55.0491.
    1. Cimino-Mathews A, Thompson E, Taube JM, Ye X, Lu Y, Meeker A, et al. PD-L1 (B7-H1) expression and the immune tumor microenvironment in primary and metastatic breast carcinomas. Hum Pathol. 2016;47:52–63. doi: 10.1016/j.humpath.2015.09.003.
    1. Emens LA, Cruz C, Eder JP, Braiteh F, Chung C, Tolaney SM, et al. Long-term clinical outcomes and biomarker analyses of Atezolizumab therapy for patients with metastatic triple-negative breast Cancer: a phase 1 study. JAMA Oncol. 2019;5:74–82. doi: 10.1001/jamaoncol.2018.4224.
    1. Lipson EJ, Forde PM, Hammers HJ, Emens LA, Taube JM, Topalian SL. Antagonists of PD-1 and PD-L1 in Cancer treatment. Semin Oncol. 2015;42:587-600. 9. Herbst RS, Soria JC, Kowanetz M, fine GD, Hamid O, Gordon MS, et al. predictive correlates of response to the anti-PD-L1 antibody MPDL3280A in cancer patients. Nature. 2014;515:563–567. doi: 10.1038/nature14011.
    1. Vennapusa B, Baker B, Kowanetz M, Boone J, Menzl I, Bruey JM, et al. Development of a PD-L1 complementary diagnostic immunohistochemistry assay (SP142) for Atezolizumab. Appl Immunohistochem Mol Morphol. 2019;27:92–100. doi: 10.1097/PAI.0000000000000594.
    1. Wu TD, Nacu S. Fast and SNP-tolerant detection of complex variants and splicing in short reads. Bioinformatics. 2010;26:873–881. doi: 10.1093/bioinformatics/btq057.
    1. Law CW, Chen Y, Shi W, Smyth GK. voom: Precision weights unlock linear model analysis tools for RNA-seq read counts. Genome Biol. 2014;15:R29. doi: 10.1186/gb-2014-15-2-r29.
    1. Molinero L, Li Y, Chang Q-W, Maund S, Berg M, Harrison J, et al. Immunogenomic evolution and atypical response to atezolizumab in a patient with metastatic triple-negative breast cancer (mTNBC) abstract #p68. 2017; SITC; national harbor, Maryland.
    1. Burstein MD, Tsimelzon A, Poage GM, Covington KR, Contreras A, Fuqua SA, et al. Comprehensive genomic analysis identifies novel subtypes and targets of triple-negative breast cancer. Clin Cancer Res. 2015;21:1688–1698. doi: 10.1158/1078-0432.CCR-14-0432.
    1. Frampton GM, Fichtenholtz A, Otto GA, Wang K, Downing SR, He J, et al. Development and validation of a clinical cancer genomic profiling test based on massively parallel DNA sequencing. Nat Biotechnol. 2013;31:1023–1031. doi: 10.1038/nbt.2696.
    1. Chalmers ZR, Connelly CF, Fabrizio D, Gay L, Ali SM, Ennis R, et al. Analysis of 100,000 human cancer genomes reveals the landscape of tumor mutational burden. Genome Med. 2017;9:34. doi: 10.1186/s13073-017-0424-2.
    1. Sun JX, He Y, Sanford E, Montesion M, Frampton GM, Vignot S, et al. A computational approach to distinguish somatic vs germline origin of genomic alterations from deep sequencing of cancer specimens without a matched normal. PLoS Comput Biol. 2018;14:e1005965. doi: 10.1371/journal.pcbi.1005965.
    1. Cid S, Eiro N, Fernandez B, Sanchez R, Andicoechea A, Fernandez-Muniz PI, et al. Prognostic influence of tumor Stroma on breast Cancer subtypes. Clin Breast Cancer. 2018;18:e123–ee33. doi: 10.1016/j.clbc.2017.08.008.
    1. Karn T, Pusztai L, Holtrich U, Iwamoto T, Shiang CY, Schmidt M, et al. Homogeneous datasets of triple negative breast cancers enable the identification of novel prognostic and predictive signatures. PLoS One. 2011;6:e28403. doi: 10.1371/journal.pone.0028403.
    1. Rizvi H, Sanchez-Vega F, La K, Chatila W, Jonsson P, Halpenny D, et al. Molecular determinants of response to anti-programmed cell death (PD)-1 and anti-programmed death-ligand 1 (PD-L1) blockade in patients with non-small-cell lung Cancer profiled with targeted next-generation sequencing. J Clin Oncol. 2018;36:633–641. doi: 10.1200/JCO.2017.75.3384.
    1. Rosenberg JE, Hoffman-Censits J, Powles T, van der Heijden MS, Balar AV, Necchi A, et al. Atezolizumab in patients with locally advanced and metastatic urothelial carcinoma who have progressed following treatment with platinum-based chemotherapy: a single-arm, multicentre, phase 2 trial. Lancet. 2016;387:1909–1920. doi: 10.1016/S0140-6736(16)00561-4.
    1. Teschendorff AE, Miremadi A, Pinder SE, Ellis IO, Caldas C. An immune response gene expression module identifies a good prognosis subtype in estrogen receptor negative breast cancer. Genome Biol. 2007;8:R157. doi: 10.1186/gb-2007-8-8-r157.
    1. Emens LA, Middleton G. The interplay of immunotherapy and chemotherapy: harnessing potential synergies. Cancer Immunol Res. 2015;3:436–443. doi: 10.1158/2326-6066.CIR-15-0064.
    1. Voorwerk L, Slagter M, Horlings HM, Sikorska K, van de Vijver KK, de Maaker M, et al. Immune induction strategies in metastatic triple-negative breast cancer to enhance the sensitivity to PD-1 blockade: the TONIC trial. Nat Med. 2019;25:920–928. doi: 10.1038/s41591-019-0432-4.
    1. Riaz N, Havel JJ, Makarov V, Desrichard A, Urba WJ, Sims JS, et al. Tumor and microenvironment evolution during immunotherapy with Nivolumab. Cell. 2017;171:934–949. doi: 10.1016/j.cell.2017.09.028.
    1. Karn T, Jiang T, Hatzis C, Sanger N, El-Balat A, Rody A, et al. Association between genomic metrics and immune infiltration in triple-negative breast Cancer. JAMA Oncol. 2017;3:1707–1711. doi: 10.1001/jamaoncol.2017.2140.
    1. Zaretsky JM, Garcia-Diaz A, Shin DS, Escuin-Ordinas H, Hugo W, Hu-Lieskovan S, 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. Adams Sylvia, Diamond Jennifer R., Hamilton Erika, Pohlmann Paula R., Tolaney Sara M., Chang Ching-Wei, Zhang Wei, Iizuka Koho, Foster Paul G., Molinero Luciana, Funke Roel, Powderly John. Atezolizumab Plus nab-Paclitaxel in the Treatment of Metastatic Triple-Negative Breast Cancer With 2-Year Survival Follow-up. JAMA Oncology. 2019;5(3):334. doi: 10.1001/jamaoncol.2018.5152.
    1. Emens LA, Loi S, Rugo HS, Schneeweiss A, Diéras V, Iwata H, et al. IMpassion130: efficacy in immune biomarker subgroups from the global, randomized, double-blind, placebo-controlled, phase III study of atezolizumab + nab-paclitaxel in patients with treatment-naïve, locally advanced or metastatic triple-negative breast cancer abstract #GS1-04. 2018; SABCS; San Antonio, Texas.

Source: PubMed

3
Suscribir