Neoadjuvant anti-PD-1 immunotherapy promotes a survival benefit with intratumoral and systemic immune responses in recurrent glioblastoma

Timothy F Cloughesy, Aaron Y Mochizuki, Joey R Orpilla, Willy Hugo, Alexander H Lee, Tom B Davidson, Anthony C Wang, Benjamin M Ellingson, Julie A Rytlewski, Catherine M Sanders, Eric S Kawaguchi, Lin Du, Gang Li, William H Yong, Sarah C Gaffey, Adam L Cohen, Ingo K Mellinghoff, Eudocia Q Lee, David A Reardon, Barbara J O'Brien, Nicholas A Butowski, Phioanh L Nghiemphu, Jennifer L Clarke, Isabel C Arrillaga-Romany, Howard Colman, Thomas J Kaley, John F de Groot, Linda M Liau, Patrick Y Wen, Robert M Prins, Timothy F Cloughesy, Aaron Y Mochizuki, Joey R Orpilla, Willy Hugo, Alexander H Lee, Tom B Davidson, Anthony C Wang, Benjamin M Ellingson, Julie A Rytlewski, Catherine M Sanders, Eric S Kawaguchi, Lin Du, Gang Li, William H Yong, Sarah C Gaffey, Adam L Cohen, Ingo K Mellinghoff, Eudocia Q Lee, David A Reardon, Barbara J O'Brien, Nicholas A Butowski, Phioanh L Nghiemphu, Jennifer L Clarke, Isabel C Arrillaga-Romany, Howard Colman, Thomas J Kaley, John F de Groot, Linda M Liau, Patrick Y Wen, Robert M Prins

Abstract

Glioblastoma is the most common primary malignant brain tumor in adults and is associated with poor survival. The Ivy Foundation Early Phase Clinical Trials Consortium conducted a randomized, multi-institution clinical trial to evaluate immune responses and survival following neoadjuvant and/or adjuvant therapy with pembrolizumab in 35 patients with recurrent, surgically resectable glioblastoma. Patients who were randomized to receive neoadjuvant pembrolizumab, with continued adjuvant therapy following surgery, had significantly extended overall survival compared to patients that were randomized to receive adjuvant, post-surgical programmed cell death protein 1 (PD-1) blockade alone. Neoadjuvant PD-1 blockade was associated with upregulation of T cell- and interferon-γ-related gene expression, but downregulation of cell-cycle-related gene expression within the tumor, which was not seen in patients that received adjuvant therapy alone. Focal induction of programmed death-ligand 1 in the tumor microenvironment, enhanced clonal expansion of T cells, decreased PD-1 expression on peripheral blood T cells and a decreasing monocytic population was observed more frequently in the neoadjuvant group than in patients treated only in the adjuvant setting. These findings suggest that the neoadjuvant administration of PD-1 blockade enhances both the local and systemic antitumor immune response and may represent a more efficacious approach to the treatment of this uniformly lethal brain tumor.

Figures

Fig. 1. CONSORT diagram
Fig. 1. CONSORT diagram
Flow diagram of disposition of patients enrolled in the study.
Fig. 2. Kaplan-Meier plot of progression-free survival
Fig. 2. Kaplan-Meier plot of progression-free survival
Median progression-free survival (PFS) for patients who received pembrolizumab only in the adjuvant setting was 72.5 days; patients who received neoadjuvant and adjuvant pembrolizumab had a median PFS of 99.5 days (HR = 0.43, 95% CI 0.20 to 0.90; two-sided P = 0.03 by log-rank test).
Fig. 3. Eighteen gene interferon-γ-related signature scores…
Fig. 3. Eighteen gene interferon-γ-related signature scores in neoadjuvant versus adjuvant-only groups
Line at middle of box represents the median; box extends from the 25th to 75th percentiles; whiskers represent minimum and maximum values; n = 28 independent biological samples; P = 0.025, U = 49 by two-sided Mann-Whitney U test. *: P < 0.05.
Fig. 4. RNAseq comparison to other recurrent…
Fig. 4. RNAseq comparison to other recurrent glioblastoma samples
We combined our RNAseq dataset with that of GSE79671 (an RNAseq dataset of recurrent glioblastoma pre- and post- bevacizumab treatment; only pre-treatment (Pre-Tx) samples were used) and The Cancer Genome Atlas (TCGA) glioblastoma samples. We applied appropriate batch correction on log transformed, normalized mRNA expression values using the removeBatchEffect function in the R package limma to estimate the fraction of glioblastoma patients with positive enrichment of the cell cycle/cancer proliferation signatures (GSVA score ≤ 0.2). The proportion of positive enrichment of the cell cycle/cancer proliferation signatures in our dataset as a whole is similar to GSE79671 (14 out of 29 (48%) vs. 11 out of 20 (55%)). The number of samples with positive enrichment in the TCGA GBM is lower at 41%. We observed that neoadjuvant PD-1 monoclonal antibody therapy group is associated with lower fraction of tumors with the cell cycle signatures. There were only 3 out of 14 tumors in the neoadjuvant group demonstrating positive enrichment while there were 11 of 15 tumors in the adjuvant group and 11 of 20 tumors in the GSE79671 set (one-sided Fisher exact test P = 0.01 and P = 0.05, respectively). GSVA: gene set variation analysis.
Fig. 5. RNAseq comparison to TCGA
Fig. 5. RNAseq comparison to TCGA
We combined our RNAseq dataset with that of TCGA glioblastoma dataset with appropriate batch correction to estimate the fraction of glioblastoma patients with positive enrichment of the cell cycle/cancer proliferation signatures (GSVA score ≥ 0.2). Three out of 14 tumors in the neoadjuvant group demonstrated positive enrichment while there were 11 of 15 tumors in the adjuvant group and 73 of 166 tumors in The Cancer Genome Atlas set. TCGA: The Cancer Genome Atlas. GSVA: gene set variation analysis.
Fig. 6. Mass cytometry dimension reduction
Fig. 6. Mass cytometry dimension reduction
(a) Diffusion map of peripheral blood mononuclear cells (PBMCs) sampled from n = 28 patients at baseline, the time of surgery and on-treatment. Phenotypically similar cells are depicted in an unsupervised manner along the same continuous axes in a pseudotemporal progression. (b) t-distributed stochastic neighbor embedding (tSNE) plot of PBMCs from n = 28 patients at all three time points. Phenotypically similar cells are clustered in an unsupervised manner. All represented cells in both panels are colored by algorithmically assigned cluster numbers using the FlowSOM package. CD3+CD4+, CD3+CD8+, CD3−CD19+ and CD3−CD14+CD16+CD11b+CD11c+ cells are labelled to demonstrate how clustered cells are plotted in close proximity to one another.
Figure 1.. Neoadjuvant pembrolizumab confers significant improvement…
Figure 1.. Neoadjuvant pembrolizumab confers significant improvement in overall and progression-free survival in patients with recurrent glioblastoma.
Patients in the neoadjuvant arm (red) received 200 mg pembrolizumab 14±5 days prior to surgical resection; patients in the adjuvant-only arm (blue) did not; both groups received 200 mg adjuvant pembrolizumab every 3 weeks. (a) Kaplan-Meier plot of overall survival. Median overall survival for the patients receiving adjuvant treatment only was 228.5 days, whereas median survival in the neoadjuvant group was 417 days (hazard ratio (HR) 0.39 neoadjuvant/adjuvant; P = 0.04 by log-rank test, n = 35 patients). (b) Swimmer plot of individual patients depicting start and stop times of dexamethasone or equivalent (as recorded at study MRI visit) and/or bevacizumab, if applicable, as well as time to progression, in days after enrollment. Bars represent overall survival in days. Rightward arrow indicates that the patient was still alive at the time of final data collection. (c) Serial magnetic resonance imaging (MRI) scans of a representative patient from the neoadjuvant group. The patient, a 66-year old female in first recurrence, is IDH negative and MGMT unmethylated. IDH: isocitrate dehydrogenase. MGMT: O6-methylguanine DNA methyltransferase. PD: progression of disease. Pembro: pembrolizumab. Tx: treatment.
Figure 2.. Tumor gene expression profile altered…
Figure 2.. Tumor gene expression profile altered by neoadjuvant PD-1 blockade.
(a) Heat map of tumor mRNA expression of interferon-γ related gene panel for individual patients. Within this panel, “A” denotes patient in neoadjuvant group; “B” denotes patient in adjuvant-only group. Dendrograms represent unsupervised hierarchical clustering by Ward’s minimum distance. Green coloration represents decreased expression; red coloration represents increased expression. (b) (Top) Heatmap showing the gene set variation analysis (GSVA) enrichment scores of gene sets with interquartile range (IQR) ≥ 1. The gene sets can be grouped into the three categories: 1) interferon pathway induction, 2) T-cell activity and 3) cell cycle/proliferation. (Middle) The heatmap of mRNA expression of the representative genes corresponding to the gene set enrichments above. (Bottom) The heatmap of progression free- and overall survival of each patient (in log2 scale). MHC: major histocompatibility complex. OS: overall survival. PFS: progression-free survival.
Figure 3.. Multiplex immunofluorescence imaging of tumor…
Figure 3.. Multiplex immunofluorescence imaging of tumor samples demonstrates varying degrees of PD-L1 expression and CD8+ T cell infiltration.
In the top panel, high CD8 infiltrate and focal PD-L1 expression are demonstrated in a patient tumor sample from the neoadjuvant group. In the second panel, low CD8+ T cell infiltrate and negative PD-L1 expression are seen in a patient tumor sample from the adjuvant-only group. In the third panel, a patient tumor sample from the neoadjuvant group demonstrates low CD8 infiltration with constitutive PD-L1 expression. In the bottom panel, a tumor sample from a patient in the adjuvant-only group demonstrates high CD8 infiltration with focal PD-L1 expression; however, the degree of PD-L1 expression is lower. Images on the left are depicted at lower magnification to demonstrate the difference between focal and constitutive PD-L1 expression and are representative of the entire tumor section/slide. Images second from the left show staining for CD8; second from the right, PD-1 stains are superimposed; on the right, the images depict co-staining for CD8, PD-1 and PD-L1. Multiplex staining was performed in one standardized run per patient. For each staining run, two sequential slides were used as duplicates for each patient. Computational analysis was performed two or more times per sample.
Figure 4.. Neoadjuvant PD-1 blockade alters correlative…
Figure 4.. Neoadjuvant PD-1 blockade alters correlative relationships between blood and tumor repertoire features and alters circulating immune cell phenotypes.
(a) Box and whisker plots comparing the number of expanded T cell receptor clones between baseline and surgery (left; P = 0.07, two-tailed t test, t = 1.98, df = 14.33, 95% CI −1.94 to 50.7, n = 26 patients) and surgery and 1-2 cycles of pembrolizumab (right; P = 0.85, two-tailed t test, t = 0.19, df = 26.0, 95% CI −30.2 to 36.3, n = 28 patients). The Y axis denotes the number of expanded clones. (b) Box and whisker plot comparing the T cell receptor overlap between peripheral blood and tumor at the time of surgery (P = 0.59, two-tailed t test, t = 0.54, df = 24.8, 95% CI −0.01 to 0.008, n = 27 patients). On the Y axis, 0 indicates no clonal overlap and 1 indicates complete overlap. For panels (a) and (b), whiskers represent minima and maxima, boxes extend from 25th to 75th percentiles, middle line represents median. (c) Hierarchically ordered Spearman correlation plots of the T cell receptor sequencing data. Numbers indicate the Spearman correlation coefficient; boxes marked with an X had Benjamini-Hochberg adjusted, two-tailed P > 0.01 by asymptotic t approximation, n = 25 patients. Patients who received neoadjuvant PD-1 blockade (left) demonstrated significant relationships between multiple variables. (d) Scatter plot of the proportions of a peripheral cluster of intermediate monocytes (CD11b+CD11c+CD14+CD16+HLA-DRhi) at baseline and after 1-2 cycles of adjuvant therapy. The Y axis indicates percent of live mononuclear cells. n = 28 patients; Benjamini-Hochberg-corrected, two-sided P = 0.007 by general linear hypothesis test. (e) Scatter plot of selected cell surface markers on CD4+ T-cells before and after the first dose of pembrolizumab. Note the decreased PD-1 expression and increased CD152 (Benjamini-Hochberg corrected, two-sided P = 0.025 and 0.0015, respectively, general linear hypothesis test, n = 28 patients) in the neoadjuvant group (left). *: P < 0.05; **: P < 0.01; ns: non-significant; N+A: neoadjvant + adjuvant; A: adjuvant only.
Figure 5.. Proposed mechanism of neoadjuvant PD-1…
Figure 5.. Proposed mechanism of neoadjuvant PD-1 blockade in recurrent GBM.
Tumor infiltrating lymphocytes, if present, are rendered ineffective through the PD-1/PD-L1 axis. Neoadjuvant PD-1 blockade releases this checkpoint, enabling modulation of the T cell receptor (TCR) clonotypes with systemic activation and clonal selection of tumor-specific T cells. Such T cell activation in turn upregulates interferon-γ-related signaling, while downregulating tumor cell cycle related genes. After surgery, and with continued adjuvant anti-PD-1 monoclonal antibody administration, tumor-specific T cells continue to eliminate residual tumor cells and begin transitioning toward a T memory phenotype. In the adjuvant-only group, surgery occurs before checkpoint blockade release. Because of the reduced residual antigenic burden, TCR modulation is less robust and fewer tumor-specific T cells are activated. With fewer tumor-specific T cells, the remaining tumor cells are able to proliferate at a more rapid pace.

References

    1. Ostrom QT, et al. CBTRUS Statistical Report: Primary brain and other central nervous system tumors diagnosed in the United States in 2010-2014. Neuro Oncol 19, v1–v88 (2017).
    1. Stupp R, et al. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med 352, 987–996 (2005).
    1. Lamborn KR, et al. Progression-free survival: an important end point in evaluating therapy for recurrent high-grade gliomas. Neuro Oncol 10, 162–170 (2008).
    1. Wu W, et al. Joint NCCTG and NABTC prognostic factors analysis for high-grade recurrent glioma. Neuro Oncol 12, 164–172 (2010).
    1. Clarke JL, et al. Is surgery at progression a prognostic marker for improved 6-month progression-free survival or overall survival for patients with recurrent glioblastoma? Neuro Oncol 13, 1118–1124 (2011).
    1. Brahmer JR, et al. Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. N Engl J Med 366, 2455–2465 (2012).
    1. Burki TK Pembrolizumab for patients with advanced melanoma. Lancet Oncol 16, e264 (2015).
    1. Garon EB, et al. Pembrolizumab for the treatment of non-small-cell lung cancer. N Engl J Med 372, 2018–2028 (2015).
    1. Hamid O, et al. Safety and tumor responses with lambrolizumab (anti-PD-1) in melanoma. N Engl J Med 369, 134–144 (2013).
    1. Ribas A Tumor immunotherapy directed at PD-1. N Engl J Med 366, 2517–2519 (2012).
    1. Topalian SL, et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med 366, 2443–2454 (2012).
    1. Wolchok JD, et al. Nivolumab plus ipilimumab in advanced melanoma. N Engl J Med 369, 122–133 (2013).
    1. Ribas A Adaptive Immune Resistance: How Cancer Protects from Immune Attack. Cancer Discov 5, 915–919 (2015).
    1. Ribas A, et al. Association of Pembrolizumab With Tumor Response and Survival Among Patients With Advanced Melanoma. JAMA 315, 1600–1609 (2016).
    1. Reck M, et al. Pembrolizumab versus Chemotherapy for PD-L1-Positive Non-Small-Cell Lung Cancer. N Engl J Med 375, 1823–1833 (2016).
    1. Redman JM, Gibney GT & Atkins MB Advances in immunotherapy for melanoma. BMC Med 14, 20 (2016).
    1. Liu J, et al. Improved Efficacy of Neoadjuvant Compared to Adjuvant Immunotherapy to Eradicate Metastatic Disease. Cancer Discov 6, 1382–1399 (2016).
    1. Forde PM, et al. Neoadjuvant PD-1 blockade in resectable lung cancer. N Engl J Med 378, 1976–1986 (2018).
    1. Blank CU, et al. Neoadjuvant versus adjuvant ipilimumab plus nivolumab in macroscopic stage III melanoma. Nat. Med. 24, 1655–1661 (2018).
    1. Amaria RN, et al. Neoadjuvant immune checkpoint blockade in high-risk resectable melanoma. Nat. Med. 24, 1649–1654 (2018).
    1. Blumenthal DT, et al. Pembrolizumab: first experience with recurrent primary central nervous system (CNS) tumors. J Neurooncol 129, 453–460 (2016).
    1. Bouffet E, et al. Immune Checkpoint Inhibition for Hypermutant Glioblastoma Multiforme Resulting From Germline Biallelic Mismatch Repair Deficiency. J Clin Oncol 34, 2206–2211 (2016).
    1. Johanns TM, et al. Immunogenomics of Hypermutated Glioblastoma: A Patient with Germline POLE Deficiency Treated with Checkpoint Blockade Immunotherapy. Cancer Discov 6, 1230–1236 (2016).
    1. Reardon DA, et al. Randomized phase 3 study evaluating the efficacy and safety of nivolumab vs bevacizumab in patients with recurrent glioblastoma: Checkmate 143. Neuro-Oncology 19, 21–21 (2017).
    1. Antonios JP, et al. Immunosuppressive tumor-infiltrating myeloid cells mediate adaptive immune resistance via a PD-1/PD-L1 mechanism in glioblastoma. Neuro Oncol 19, 796–807 (2017).
    1. Antonios JP, et al. PD-1 blockade enhances the vaccination-induced immune response in glioma. JCI Insight 1(2016).
    1. Bloch O, et al. Gliomas promote immunosuppression through induction of B7-H1 expression in tumor-associated macrophages. Clin. Cancer. Res 19, 3165–3175 (2013).
    1. Reardon DA, et al. Glioblastoma Eradication Following Immune Checkpoint Blockade in an Orthotopic, Immunocompetent Model. Cancer Immunol Res 4, 124–135 (2016).
    1. Wainwright DA, et al. Durable therapeutic efficacy utilizing combinatorial blockade against IDO, CTLA-4, and PD-L1 in mice with brain tumors. Clin. Cancer. Res 20, 5290–5301 (2014).
    1. Zeng J, et al. Anti-PD-1 blockade and stereotactic radiation produce long-term survival in mice with intracranial gliomas. Int. J. Radiat. Oncol. Biol. Phys 86, 343–349 (2013).
    1. Okada H, et al. Immunotherapy response assessment in neuro-oncology: a report of the RANO working group. Lancet Oncol 16, e534–e542 (2015).
    1. Pollack IF, et al. Antigen-specific immune responses and clinical outcome after vaccination with glioma-associated antigen peptides and polyinosinic-polycytidylic acid stabilized by lysine and carboxymethylcellulose in children with newly diagnosed malignant brainstem and nonbrainstem gliomas. J Clin Oncol 32, 2050–2058 (2014).
    1. Prins RM, et al. Gene expression profile correlates with T-cell infiltration and relative survival in glioblastoma patients vaccinated with dendritic cell immunotherapy. Clin. Cancer. Res. 17, 1603–1615 (2011).
    1. Ayers M, et al. IFN-gamma-related mRNA profile predicts clinical response to PD-1 blockade. J. Clin. Invest. 127, 2930–2940 (2017).
    1. Urup T, et al. Transcriptional changes induced by bevacizumab combination therapy in responding and non-responding recurrent glioblastoma patients. BMC Cancer 17, 278 (2017).
    1. Brennan CW, et al. The somatic genomic landscape of glioblastoma. Cell 155, 462–477 (2013).
    1. Chen J, et al. Interferon-gamma-induced PD-L1 surface expression on human oral squamous carcinoma via PKD2 signal pathway. Immunobiology 217, 385–393 (2012).
    1. Spranger S, et al. Up-regulation of PD-L1, IDO, and T(regs) in the melanoma tumor microenvironment is driven by CD8(+) T cells. Sci Transl Med 5, 200ra116 (2013).
    1. Taube JM, et al. Colocalization of inflammatory response with B7-h1 expression in human melanocytic lesions supports an adaptive resistance mechanism of immune escape. Sci Transl Med 4, 127ra137 (2012).
    1. Farmer P, et al. Identification of molecular apocrine breast tumours by microarray analysis. Oncogene 24, 4660–4671 (2005).
    1. Li G & Wang X Prediction Accuracy Measures for a Nonlinear Model and for Right-Censored Time-to-Event Data. Journal of the American Statistical Association, 1–25 (2018).
    1. Einat M, Resnitzky D & Kimchi A Close link between reduction of c-myc expression by interferon and, G0/G1 arrest. Nature 313, 597–600 (1985).
    1. Shearer M & Taylor-Papadimitriou J Regulation of cell growth by interferon. Cancer Metastasis Rev 6, 199–221 (1987).
    1. Yung WK, Steck PA, Kelleher PJ, Moser RP & Rosenblum MG Growth inhibitory effect of recombinant alpha and beta interferon on human glioma cells. J Neurooncol 5, 323–330 (1987).
    1. Robert L, et al. Distinct immunological mechanisms of CTLA-4 and PD-1 blockade revealed by analyzing TCR usage in blood lymphocytes. Oncoimmunology 3, e29244 (2014).
    1. Omuro A, et al. Nivolumab with or without ipilimumab in patients with recurrent glioblastoma: results from exploratory phase I cohorts of CheckMate 143. Neuro Oncol 20, 674–686 (2018).
    1. Kamphorst AO, et al. Proliferation of PD-1+ CD8 T cells in peripheral blood after PD-1-targeted therapy in lung cancer patients. Proc Natl Acad Sci U S A 114, 4993–4998 (2017).
    1. Huang AC, et al. T-cell invigoration to tumour burden ratio associated with anti-PD-1 response. Nature 545, 60–65 (2017).
    1. Krieg C, et al. High-dimensional single-cell analysis predicts response to anti-PD-1 immunotherapy. Nat. Med 24, 144–153 (2018).
    1. Arbour KC, et al. Impact of baseline steroids on efficacy of programmed cell death-1 and programmed death-ligand 1 blockade in patients with non-small-cell lung cancer. J Clin Oncol 36, 2872–2878 (2018).
    1. Weber JS, et al. Safety profile of nivolumab monotherapy: A pooled analysis of patients with advanced melanoma. J Clin Oncol 35, 785–792 (2017).
    1. Hamza MA, et al. Survival outcome of early versus delayed bevacizumab treatment in patients with recurrent glioblastoma. J Neurooncol 119, 135–140 (2014).
    1. Kim D, Langmead B & Salzberg SL HISAT: a fast spliced aligner with low memory requirements. Nat. Methods 12, 357–360 (2015).
    1. Anders S, Pyl PT & Huber W HTSeq--a Python framework to work with high-throughput sequencing data. Bioinformatics 31, 166–169 (2015).
    1. Hanzelmann S, Castelo R & Guinney J GSVA: gene set variation analysis for microarray and RNA-seq data. BMC Bioinformatics 14, 7 (2013).
    1. Subramanian A, et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci U S A 102, 15545–15550 (2005).
    1. Newman AM, et al. Robust enumeration of cell subsets from tissue expression profiles. Nat. Methods 12, 453–457 (2015).
    1. Tirosh I, et al. Dissecting the multicellular ecosystem of metastatic melanoma by single-cell RNA-seq. Science 352, 189–196 (2016).
    1. Grossman RL, et al. Toward a Shared Vision for Cancer Genomic Data. N Engl J Med 375, 1109–1112 (2016).
    1. Carlson CS, et al. Using synthetic templates to design an unbiased multiplex PCR assay. Nat Commun 4, 2680 (2013).
    1. Robins H, et al. Ultra-sensitive detection of rare T cell clones. J. Immunol. Methods 375, 14–19 (2012).
    1. Robins HS, et al. Comprehensive assessment of T-cell receptor beta-chain diversity in alphabeta T cells. Blood 114, 4099–4107 (2009).
    1. Emerson RO, et al. High-throughput sequencing of T-cell receptors reveals a homogeneous repertoire of tumour-infiltrating lymphocytes in ovarian cancer. J. Pathol 231, 433–440 (2013).
    1. Daley T & Smith AD Predicting the molecular complexity of sequencing libraries. Nat. Methods 10, 325–327 (2013).
    1. Nowicka M, et al. CyTOF workflow: differential discovery in high-throughput high-dimensional cytometry datasets. F1000Res 6, 748 (2017).
    1. Ellingson BM, et al. Consensus recommendations for a standardized Brain Tumor Imaging Protocol in clinical trials. Neuro Oncol 17, 1188–1198 (2015).
    1. Ellingson BM, et al. Volumetric response quantified using T1 subtraction predicts long-term survival benefit from cabozantinib monotherapy in recurrent glioblastoma. Neuro Oncol 20, 1411–1418 (2018).
    1. Ellingson BM, et al. Recurrent glioblastoma treated with bevacizumab: contrast-enhanced T1-weighted subtraction maps improve tumor delineation and aid prediction of survival in a multicenter clinical trial. Radiology 271, 200–210 (2014).
    1. Ellingson BM, et al. Validation of post-operative residual contrast enhancing tumor volume as an independent prognostic factor for overall survival in newly diagnosed glioblastoma. Neuro Oncol (2018).
    1. Ellingson BM, et al. Baseline pretreatment contrast enhancing tumor volume including central necrosis is a prognostic factor in recurrent glioblastoma: evidence from single and multicenter trials. Neuro Oncol 19, 89–98 (2017).
    1. Mootha VK, et al. PGC-1alpha-responsive genes involved in oxidative phosphorylation are coordinately downregulated in human diabetes. Nat. Genet 34, 267–273 (2003).
    1. Zhao K, Lu ZX, Park JW, Zhou Q & Xing Y GLiMMPS: robust statistical model for regulatory variation of alternative splicing using RNA-seq data. Genome Biol 14, R74 (2013).

Source: PubMed

3
Abonnieren