PD-L1 targeting and subclonal immune escape mediated by PD-L1 mutations in metastatic colorectal cancer

Alexander Stein, Donjete Simnica, Christoph Schultheiß, Rebekka Scholz, Joseph Tintelnot, Eray Gökkurt, Lisa von Wenserski, Edith Willscher, Lisa Paschold, Markus Sauer, Sylvie Lorenzen, Jorge Riera-Knorrenschild, Reinhard Depenbusch, Thomas J Ettrich, Steffen Dörfel, Salah-Eddin Al-Batran, Meinolf Karthaus, Uwe Pelzer, Lisa Waberer, Axel Hinke, Marcus Bauer, Chiara Massa, Barbara Seliger, Claudia Wickenhauser, Carsten Bokemeyer, Susanna Hegewisch-Becker, Mascha Binder, Alexander Stein, Donjete Simnica, Christoph Schultheiß, Rebekka Scholz, Joseph Tintelnot, Eray Gökkurt, Lisa von Wenserski, Edith Willscher, Lisa Paschold, Markus Sauer, Sylvie Lorenzen, Jorge Riera-Knorrenschild, Reinhard Depenbusch, Thomas J Ettrich, Steffen Dörfel, Salah-Eddin Al-Batran, Meinolf Karthaus, Uwe Pelzer, Lisa Waberer, Axel Hinke, Marcus Bauer, Chiara Massa, Barbara Seliger, Claudia Wickenhauser, Carsten Bokemeyer, Susanna Hegewisch-Becker, Mascha Binder

Abstract

Background: In patients with microsatellite stable (MSS) metastatic colorectal cancer (mCRC), immune checkpoint blockade is ineffective, and combinatorial approaches enhancing immunogenicity need exploration.

Methods: We treated 43 patients with predominantly microsatellite stable RAS/BRAF wild-type mCRC on a phase II trial combining chemotherapy with the epidermal growth factor receptor antibody cetuximab and the programmed cell death ligand 1 (PD-L1) antibody avelumab. We performed next-generation gene panel sequencing for mutational typing of tumors and liquid biopsy monitoring as well as digital droplet PCR to confirm individual mutations. Translational analyses included tissue immunohistochemistry, multispectral imaging and repertoire sequencing of tumor-infiltrating T cells. Detected PD-L1 mutations were mechanistically validated in CRISPR/Cas9-generated cell models using qRT-PCR, immunoblotting, flow cytometry, complement-dependent cytotoxicity assay, antibody-dependent cytotoxicity by natural killer cell degranulation assay and LDH release assay as well as live cell imaging of T cell mediated tumor cell killing.

Results: Circulating tumor DNA showed rapid clearance in the majority of patients mirroring a high rate of early tumor shrinkage. In 3 of 13 patients expressing the high-affinity Fcγ receptor 3a (FcγR3a), tumor subclones with PD-L1 mutations were selected that led to loss of tumor PD-L1 by nonsense-mediated RNA decay in PD-L1 K162fs and protein degradation in PD-L1 L88S. As a consequence, avelumab binding and antibody-dependent cytotoxicity were impaired, while T cell killing of these variant clones was increased. Interestingly, PD-L1 mutant subclones showed slow selection dynamics reversing on avelumab withdrawal and patients with such subclones had above-average treatment benefit. This suggested that the PD-L1 mutations mediated resistance to direct antitumor effects of avelumab, while at the same time loss of PD-L1 reduced biological fitness by enhanced T cell killing limiting subclonal expansion.

Conclusion: The addition of avelumab to standard treatment appeared feasible and safe. PD-L1 mutations mediate subclonal immune escape to avelumab in some patients with mCRC expressing high-affinity FcγR3a, which may be a subset experiencing most selective pressure. Future trials evaluating the addition of avelumab to standard treatment in MSS mCRC are warranted especially in this patient subpopulation.

Trial registration number: NCT03174405.

Keywords: biomarkers; clinical trials; gastrointestinal neoplasms; immunotherapy; phase II as topic; translational medical research; tumor.

Conflict of interest statement

Competing interests: AS received institutional research grants from Merck, BMS, Roche, Sanofi, Servier and honoraria for lectures and advisory board meetings by Merck, Roche, Amgen, Lilly, Sanofi-Aventis, Servier, Bayer, BMS, MSD and Sirtex. S-E A-B has an advisory role with Merck, Roche, Celgene, Lilly, Nordic Pharma, Bristol-Myers Squibb, Astellas and MSD Sharp & Dohme; is a speaker for Roche, Celgene, Lilly, Nordic Pharma, AIO gGmbH, MCI, promedicis, Forum für Medizinische Fortbildung and Taiho pharma; he is CEO/founder of IKF Klinische Krebsforschung GmbH at Northwest Hospital; and has received research grants from Sanofi, Merck, Roche, Celgene, Vifor, Medac, Hospira, Lilly, Eurozyto, German Cancer Aid (Krebshilfe), German Research Foundation and the Federal Ministry of Education and Research. UP received institutional research grants from Celgene, BMS, Amgen, Lilly, Roche, Sanofi and Servier and honoraria for lectures and advisory board meetings by Roche, Celgene, Amgen, Lilly, Sanofi-Aventis, Servier, Bayer and BMS. AH received honoraria for lectures from Roche. CB received institutional research grants and honoraria for lectures and advisory board meetings from Merck, BMS, Roche, Sanofi, Servier, Bayer, BMS, Astrazeneca, Lilly, Mundipharma, Hexal, MSD and GSO. MB received institutional research grants from Merck, BMS, Hexal, German Cancer Aid (Krebshilfe), German Research Foundation and the Federal Ministry of Education and Research as well as honoraria for lectures and advisory board meetings by Celgene, Janssen, Gilead, Merck, Roche, Amgen, Sanofi-Aventis and BMS.

© Author(s) (or their employer(s)) 2021. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.

Figures

Figure 1
Figure 1
Efficacy of AVETUX protocol. (A) Consort diagram. (B) Kaplan-Meier estimates of progression-free survival. (C) Waterfall plot of best responses in target lesions (PD in responding patient was new lesion). (D) Spider plot depicting tumor measurements over time. Patients with MSI are marked with asterisk. (E) Kaplan-Meier estimates of overall survival. PD, progressive disease.
Figure 2
Figure 2
Mutational profiling and liquid biopsy disease monitoring. (A) Distribution of mutation spectra in FFPE tumor tissue and liquid biopsy at baseline evaluation. (B) Venn diagram of patients of which tumor driver mutations were detected by gene panel sequencing in FFPE tissue and/or liquid biopsy, respectively. (C) Circulating tumor (CT) DNA clearance from baseline to week 4 after treatment initiation. (D) Serial liquid biopsy testing in patients with disease progression during observational period. Gray box: increase or reappearance of ctDNA prior to clinical PD in weeks. Line indicates median. (E) KRAS and NRAS circulating tumor DNA monitoring during AVETUX therapy regimen. Respective patient number in bold. Patients with MSI are marked with asterisk. CR, complete response; FFPE, formalin-fixed paraffin-embedded; PR, partial response; PD, progressive disease; SD, stable disease; VAF, variant allele frequency; SFU, safety follow-up.
Figure 3
Figure 3
Selection of resistance variants on AVETUX protocol and clearance after avelumab withdrawal. (A) Overview of B2M, JAK1 and PD-L1 mutations in baseline and on-treatment tumor and liquid biopsy samples. Tumor samples collected under treatment or at EOT originate from: patient 12: liver metastasis, patient 22: tumor DNA 1: rectum and tumor DNA 2: bladder (same metastatic site as used for baseline testing),. (B) Localization of B2M, JAK1 and PD-L1 mutations. (C) Distribution of emerging resistance mutations to avelumab and cetuximab as well as FcγR3a genotype (rs396991) over the cohort. Asterisk on KRAS indicates that mutation disappeared in follow-up liquid biopsy samples. In total five on-treatment tumor samples were available. (D) ddPCR validation of immune checkpoint blockade resistance variants in patient 22. (E) PFS of patients with F/F versus F/V or V/V FcγR3a genotype (rs396991). Dotted line indicates median PFS of entire patient cohort. Statistical test: one-sided, unpaired t-test. (F) Longitudinal ctDNA and biopsy (hemicolectomy) mutational and CEA profile of patient 21 with a treatment-induced PD-L1 K162fs mutation and patient 30 with a treatment-induced PD-L1 L88S mutation. Treatment is indicated above each plot. Highlighted area indicates time during AVETUX regimen. Patients with MSI are marked with asterisk. APC, adenomatous polyposis coli; B2M, β2-microglobulin; CEA, carcinoembryonic antigen; JAK1, Janus kinase 1; PD-L1, programmed cell death ligand 1; PFS, progression-free survival; PD, progressive disease; PR, partial response; TP53, tumor protein p53;
Figure 4
Figure 4
IHC staining of PD-L1 in pre-treatment and on-treatment tumor tissue of patient 22 with genetic evidence of a treatment-induced PD-L1 L88S mutation. Representative micrographs of H&E and PD-L1 stainings of tumor tissue at 100× and 400× magnification, respectively. Moreover, multiplex IHC is shown using antibodies against PanCK (tumor cells, green), CD3 (T cells, turquoise) and PD-L1 (magenta). Nuclei are stained with DAPI. IC, immune cell score; IHC, immunohistochemistry; PD-L1, programmed cell death protein ligand 1; TPS, Tumor proportional score; Tx, treatment.
Figure 5
Figure 5
Selected PD-L1 mutations reduce protein abundancy, surface expression, ADCC and T cell suppression. (A) Schematic representation of gRNAs targeting the PD-L1 coding sequence (CDS). (B) CRISPR/Cas9-mediated depletion of PD-L1 as detected in crude cell extracts via immunoblotting. (C) PD-L1 protein levels after lentiviral redelivery of PD-L1 variants into PD-L1KO2 human cell lines and the murine Ba/F3 cell line as detected by immunoblotting. (D) Enrichment of PD-L1 in the membranous fraction (M) of HT-29 cell variants from panel C. (C) Cytosolic, non-membranous fraction. (E) Flow cytometric detection of PD-L1 surface expression displayed as mean relative fluorescence intensity (RI) after staining with avelumab. n(DLD-1)=7, n(HT-29)=8, n(UT-SCC-14)=9, n(UT-SCC-29)=8, n(Ba/F3)=4. (F) NK cell degranulation induced by coculturing primary NK cells and cell lines expressing indicated PD-L1 variants in the presence of avelumab (n=4 for DLD-1, HT-29, UT-SCC-29; n=5 for UT-SCC-14). Percent degranulated NK cells normalized to spontaneous NK degranulation is shown for all cocultures. (G) Representative images and time course of T cell mediated tumor cell killing. CD8+ T cells were cocultured with HT-29, DLD-1 and UT-SCC-14 cells expressing WT PD-L1 or the L88S and K162fs variants (red fluorescence). Caspase 3/7 activity (relative intensity of green fluorescence) was monitored every 90 min for 24 hours with an Incuyte S3. Scale bar, 200 µM. Asterisks indicate p value range (*p<0.05; **p<0.01; ***p<0.001; ***p<0.0001). Statistics for time course: two-tailed paired t test. All other statistics: two-tailed unpaired t test. NK, natural killer; PD-L1, programmed cell death protein ligand 1.
Figure 6
Figure 6
PD-L1 L88S exhibits enhanced phosphorylation-dependent proteasomal degradation. (A) immunoblot analysis of PD-L1 abundancy in HT-29 (n=6) and UT-SCC-14 (n=4) cells overexpressing PD-L1 variants after blocking protein synthesis with 20 µM CHX for 3, 6 and 9 hours (h). (B) Quantification of (A) and four replicates using ImageJ. (C) Relative stability of PD-L1 protein (=signal intensity relative to control as quantified using ImageJ) in PD-L1 overexpressing cells after abrogation of N-glycosylation for 18 hours using tunicamycin (Tm) as determined by immunoblotting (n=5). (D) Quantification of PD-L1 protein abundancy in PD-L1 overexpressing cells after 2-hour and 4-hour blocking of AMPK with 10 µM compound C (Comp C) as determined by immunoblotting (n=6). (E) Enrichment of PD-L1 variants after inhibition of the proteasome using 20 µM MG132 for 4 hours. Quantification of glycosylated (·) and non-glycosylated (··) PD-L1 from four replicates of PD-L1 transduced HT-29 and UT-SCC-14 cells. Statistics: two-tailed unpaired t-test. Asterisks indicate p value range (*p<0.05; **p<0.01; ***p<0.001; ***p<0.0001; ns>0.05). AMPK, AMP-activated protein kinase; PD-L1, programmed cell death protein ligand 1.

References

    1. Overman MJ, McDermott R, Leach JL, et al. . Nivolumab in patients with metastatic DNA mismatch repair-deficient or microsatellite instability-high colorectal cancer (CheckMate 142): an open-label, multicentre, phase 2 study. Lancet Oncol 2017;18:1182–91. 10.1016/S1470-2045(17)30422-9
    1. Overman MJ, Lonardi S, Wong KYM, et al. . Durable clinical benefit with nivolumab plus ipilimumab in DNA mismatch Repair-Deficient/Microsatellite Instability-High metastatic colorectal cancer. J Clin Oncol 2018;36:773–9. 10.1200/JCO.2017.76.9901
    1. André T, Shiu K-K, Kim TW, et al. . Pembrolizumab in Microsatellite-Instability-High advanced colorectal cancer. N Engl J Med 2020;383:2207–18. 10.1056/NEJMoa2017699
    1. Grothey A, Tabernero J, Arnold D. Fluoropyrimidine (FP) + bevacizumab (BEV) + atezolizumab vs FP/BEV in BRAFwt metastatic colorectal cancer (mCRC): findings from cohort 2 of MODUL. Ann Oncol suppl 2018;29:714–5.
    1. Le DT, Uram JN, Wang H, et al. . Pd-1 blockade in tumors with mismatch-repair deficiency. N Engl J Med 2015;372:2509–20. 10.1056/NEJMoa1500596
    1. Eng C, Kim TW, Bendell J, et al. . Atezolizumab with or without cobimetinib versus regorafenib in previously treated metastatic colorectal cancer (IMblaze370): a multicentre, open-label, phase 3, randomised, controlled trial. Lancet Oncol 2019;20:849–61. 10.1016/S1470-2045(19)30027-0
    1. Duffy AG, Greten TF. Immunological off-target effects of standard treatments in gastrointestinal cancers. Ann Oncol 2014;25:24–32. 10.1093/annonc/mdt349
    1. Pozzi C, Cuomo A, Spadoni I, et al. . The EGFR-specific antibody cetuximab combined with chemotherapy triggers immunogenic cell death. Nat Med 2016;22:624–31. 10.1038/nm.4078
    1. Tesniere A, Schlemmer F, Boige V, et al. . Immunogenic death of colon cancer cells treated with oxaliplatin. Oncogene 2010;29:482–91. 10.1038/onc.2009.356
    1. Woolston A, Khan K, Spain G, et al. . Genomic and transcriptomic determinants of therapy resistance and immune landscape evolution during anti-EGFR treatment in colorectal cancer. Cancer Cell 2019;36:35–50. 10.1016/j.ccell.2019.05.013
    1. Boyerinas B, Jochems C, Fantini M, et al. . Antibody-Dependent cellular cytotoxicity activity of a novel anti-PD-L1 antibody Avelumab (MSB0010718C) on human tumor cells. Cancer Immunol Res 2015;3:1148–57. 10.1158/2326-6066.CIR-15-0059
    1. Wickenhauser C, Bethmann D, Feng Z, et al. . Multispectral fluorescence imaging allows for distinctive topographic assessment and subclassification of tumor-infiltrating and surrounding immune cells. Methods Mol Biol 2019;1913:13–31. 10.1007/978-1-4939-8979-9_2
    1. Akyüz N, Brandt A, Stein A, et al. . T-Cell diversification reflects antigen selection in the blood of patients on immune checkpoint inhibition and may be exploited as liquid biopsy biomarker. Int J Cancer 2017;140:2535–44. 10.1002/ijc.30549
    1. Mährle T, Akyüz N, Fuchs P, et al. . Deep sequencing of bone marrow microenvironments of patients with del(5q) myelodysplastic syndrome reveals imprints of antigenic selection as well as generation of novel T-cell clusters as a response pattern to lenalidomide. Haematologica 2019;104:2018.208223. 10.3324/haematol.2018.208223
    1. Mohme M, Schliffke S, Maire CL, et al. . Immunophenotyping of newly diagnosed and recurrent glioblastoma defines distinct immune exhaustion profiles in peripheral and tumor-infiltrating lymphocytes. Clin Cancer Res 2018;24:4187–200. 10.1158/1078-0432.CCR-17-2617
    1. Schliffke S, Akyüz N, Ford CT, et al. . Clinical response to ibrutinib is accompanied by normalization of the T-cell environment in CLL-related autoimmune cytopenia. Leukemia 2016;30:2232–4. 10.1038/leu.2016.157
    1. Schliffke S, Carambia A, Akyüz N, et al. . T-Cell repertoire profiling by next-generation sequencing reveals tissue migration dynamics of TRBV13-family clonotypes in a common experimental autoimmune encephalomyelitis mouse model. J Neuroimmunol 2019;332:49–56. 10.1016/j.jneuroim.2019.03.014
    1. Simnica D, Akyüz N, Schliffke S, et al. . T cell receptor next-generation sequencing reveals cancer-associated repertoire metrics and reconstitution after chemotherapy in patients with hematological and solid tumors. Oncoimmunology 2019;8:e1644110. 10.1080/2162402X.2019.1644110
    1. Simnica D, Schliffke S, Schultheiß C, et al. . High-Throughput immunogenetics reveals a lack of physiological T cell clusters in patients with autoimmune cytopenias. Front Immunol 2019;10:1897. 10.3389/fimmu.2019.01897
    1. Bolotin DA, Poslavsky S, Mitrophanov I, et al. . MiXCR: software for comprehensive adaptive immunity profiling. Nat Methods 2015;12:380–1. 10.1038/nmeth.3364
    1. R. C. Team . R foundation for statistical computing. Vienna, Austria, 2018.
    1. Nazarov VI, Pogorelyy MV, Komech EA, et al. . tcR: an R package for T cell receptor repertoire advanced data analysis. BMC Bioinformatics 2015;16:175. 10.1186/s12859-015-0613-1
    1. Cerami E, Gao J, Dogrusoz U, et al. . The cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics data. Cancer Discov 2012;2:401–4. 10.1158/-12-0095
    1. Xu C, Gu X, Padmanabhan R, et al. . smCounter2: an accurate low-frequency variant caller for targeted sequencing data with unique molecular identifiers. Bioinformatics 2019;35:1299–309. 10.1093/bioinformatics/bty790
    1. Tintelnot J, Baum N, Schultheiß C, et al. . Nanobody targeting of epidermal growth factor receptor (EGFR) ectodomain variants overcomes resistance to therapeutic EGFR antibodies. Mol Cancer Ther 2019;18:823–33. 10.1158/1535-7163.MCT-18-0849
    1. Alter G, Malenfant JM, Altfeld M. CD107a as a functional marker for the identification of natural killer cell activity. J Immunol Methods 2004;294:15–22. 10.1016/j.jim.2004.08.008
    1. Yamashita K, Iwatsuki M, Harada K, et al. . Can PD-L1 expression evaluated by biopsy sample accurately reflect its expression in the whole tumour in gastric cancer? Br J Cancer 2019;121:278–80. 10.1038/s41416-019-0515-5
    1. Li C, Huang C, Mok TS, et al. . Comparison of 22C3 PD-L1 expression between surgically resected specimens and paired tissue microarrays in non-small cell lung cancer. J Thorac Oncol 2017;12:1536–43. 10.1016/j.jtho.2017.07.015
    1. Kvistborg P, Philips D, Kelderman S, et al. . Anti-Ctla-4 therapy broadens the melanoma-reactive CD8+ T cell response. Sci Transl Med 2014;6:ra128. 10.1126/scitranslmed.3008918
    1. Robert L, Tsoi J, Wang X, et al. . Ctla4 blockade broadens the peripheral T-cell receptor repertoire. Clin Cancer Res 2014;20:2424–32. 10.1158/1078-0432.CCR-13-2648
    1. Van Emburgh BO, Arena S, Siravegna G, et al. . Acquired Ras or EGFR mutations and duration of response to EGFR blockade in colorectal cancer. Nat Commun 2016;7:13665. 10.1038/ncomms13665
    1. Misale S, Yaeger R, Hobor S, et al. . Emergence of KRAS mutations and acquired resistance to anti-EGFR therapy in colorectal cancer. Nature 2012;486:532–6. 10.1038/nature11156
    1. Diaz LA, Williams RT, Wu J, et al. . The molecular evolution of acquired resistance to targeted EGFR blockade in colorectal cancers. Nature 2012;486:537–40. 10.1038/nature11219
    1. Arena S, Bellosillo B, Siravegna G, et al. . Emergence of multiple EGFR extracellular mutations during cetuximab treatment in colorectal cancer. Clin Cancer Res 2015;21:2157–66. 10.1158/1078-0432.CCR-14-2821
    1. Montagut C, Dalmases A, Bellosillo B, et al. . Identification of a mutation in the extracellular domain of the epidermal growth factor receptor conferring cetuximab resistance in colorectal cancer. Nat Med 2012;18:221–3. 10.1038/nm.2609
    1. Braig F, März M, Schieferdecker A, et al. . Epidermal growth factor receptor mutation mediates cross-resistance to panitumumab and cetuximab in gastrointestinal cancer. Oncotarget 2015;6:12035–47. 10.18632/oncotarget.3574
    1. Vidal J, Muinelo L, Dalmases A, et al. . Plasma ctDNA Ras mutation analysis for the diagnosis and treatment monitoring of metastatic colorectal cancer patients. Ann Oncol 2017;28:1325–32. 10.1093/annonc/mdx125
    1. Morelli MP, Overman MJ, Dasari A, et al. . Characterizing the patterns of clonal selection in circulating tumor DNA from patients with colorectal cancer refractory to anti-EGFR treatment. Ann Oncol 2015;26:731-736. 10.1093/annonc/mdv005
    1. Kim TW, Peeters M, Thomas A, et al. . Impact of Emergent Circulating Tumor DNA RAS Mutation in Panitumumab-Treated Chemoresistant Metastatic Colorectal Cancer. Clin Cancer Res 2018;24:5602-5609. 10.1158/1078-0432.CCR-17-3377
    1. Siravegna G, Mussolin B, Buscarino M, et al. . Clonal evolution and resistance to EGFR blockade in the blood of colorectal cancer patients. Nat Med 2015;21:795–801. 10.1038/nm.3870
    1. Zaretsky JM, Garcia-Diaz A, Shin DS, et al. . Mutations associated with acquired resistance to PD-1 blockade in melanoma. N Engl J Med 2016;375:819–29. 10.1056/NEJMoa1604958
    1. Bibeau F, Lopez-Crapez E, Di Fiore F, et al. . Impact of Fc{gamma}RIIa-Fc{gamma}RIIIa polymorphisms and KRAS mutations on the clinical outcome of patients with metastatic colorectal cancer treated with cetuximab plus irinotecan. J Clin Oncol 2009;27:1122–9. 10.1200/JCO.2008.18.0463
    1. Trotta AM, Ottaiano A, Romano C, et al. . Prospective evaluation of Cetuximab-Mediated antibody-dependent cell cytotoxicity in metastatic colorectal cancer patients predicts treatment efficacy. Cancer Immunol Res 2016;4:366–74. 10.1158/2326-6066.CIR-15-0184
    1. European medicines Agency. (2017), vol. August 2019.
    1. Li C-W, Lim S-O, Xia W, et al. . Glycosylation and stabilization of programmed death ligand-1 suppresses T-cell activity. Nat Commun 2016;7:12632. 10.1038/ncomms12632
    1. Cha J-H, Yang W-H, Xia W, et al. . Metformin promotes antitumor immunity via endoplasmic-reticulum-associated degradation of PD-L1. Mol Cell 2018;71:e607. 10.1016/j.molcel.2018.07.030
    1. Bokemeyer C, Bondarenko I, Hartmann JT, et al. . Efficacy according to biomarker status of cetuximab plus FOLFOX-4 as first-line treatment for metastatic colorectal cancer: the OPUS study. Ann Oncol 2011;22:1535–46. 10.1093/annonc/mdq632
    1. Venook AP, Niedzwiecki D, Lenz H-J, et al. . Effect of first-line chemotherapy combined with cetuximab or bevacizumab on overall survival in patients with KRAS wild-type advanced or metastatic colorectal cancer: a randomized clinical trial. JAMA 2017;317:2392–401. 10.1001/jama.2017.7105
    1. Qin S, Li J, Wang L, et al. . Efficacy and Tolerability of First-Line Cetuximab Plus Leucovorin, Fluorouracil, and Oxaliplatin (FOLFOX-4) Versus FOLFOX-4 in Patients With RAS Wild-Type Metastatic Colorectal Cancer: The Open-Label, Randomized, Phase III TAILOR Trial. J Clin Oncol 2018;36:JCO2018783183. 10.1200/JCO.2018.78.3183
    1. Innocenti F, Ou F-S, Qu X, et al. . Mutational analysis of patients with colorectal cancer in CALGB/SWOG 80405 identifies new roles of microsatellite instability and tumor mutational burden for patient outcome. J Clin Oncol 2019;37:1217–27. 10.1200/JCO.18.01798

Source: PubMed

3
S'abonner