Anti-PD-1 monoclonal antibody MEDI0680 in a phase I study of patients with advanced solid malignancies

Aung Naing, Jeffrey Infante, Sanjay Goel, Howard Burris, Chelsea Black, Shannon Marshall, Ikbel Achour, Susannah Barbee, Rena May, Chris Morehouse, Kristen Pollizzi, Xuyang Song, Keith Steele, Nairouz Elgeioushi, Farzana Walcott, Joyson Karakunnel, Patricia LoRusso, Amy Weise, Joseph Eder, Brendan Curti, Michael Oberst, Aung Naing, Jeffrey Infante, Sanjay Goel, Howard Burris, Chelsea Black, Shannon Marshall, Ikbel Achour, Susannah Barbee, Rena May, Chris Morehouse, Kristen Pollizzi, Xuyang Song, Keith Steele, Nairouz Elgeioushi, Farzana Walcott, Joyson Karakunnel, Patricia LoRusso, Amy Weise, Joseph Eder, Brendan Curti, Michael Oberst

Abstract

Background: The safety, efficacy, pharmacokinetics, and pharmacodynamics of the anti-programmed cell death-1 antibody MEDI0680 were evaluated in a phase I, multicenter, dose-escalation study in advanced solid malignancies.

Methods: MEDI0680 was administered intravenously once every 2 weeks (Q2W) or once every 3 weeks at 0.1, 0.5, 2.5, 10 or 20 mg/kg. Two cohorts received 20 mg/kg once a week for 2 or 4 weeks, then 20 mg/kg Q2W. All were treated for 12 months or until progression. The primary endpoint was safety. Secondary endpoints were efficacy and pharmacokinetics. Exploratory endpoints included pharmacodynamics.

Results: Fifty-eight patients were treated. Median age was 62.5 years and 81% were male. Most had kidney cancer (n = 36) or melanoma (n = 9). There were no dose-limiting toxicities. Treatment-related adverse events occurred in 83% and were grade ≥ 3 in 21%. Objective clinical responses occurred in 8/58 patients (14%): 5 with kidney cancer, including 1 with a complete response, and 3 with melanoma. The relationship between dose and serum levels was predictable and linear, with apparent receptor saturation at 10 mg/kg Q2W and all 20 mg/kg cohorts.

Conclusions: MEDI0680 induced peripheral T-cell proliferation and increased plasma IFNγ and associated chemokines regardless of clinical response. CD8+ T-cell tumor infiltration and tumoral gene expression of IFNG, CD8A, CXCL9, and granzyme K (GZMK) were also increased following MEDI0680 administration.

Trial registration: NCT02013804 ; date of registration December 12, 2013.

Keywords: Immunotherapy; Kidney cancer; MEDI0680; Melanoma; PD-1.

Conflict of interest statement

Aung Naing: Research grants from NCI, EMD Serono, AstraZeneca, Healios Onc., Nutrition, Atterocor, Amplimmune, Karyopharm Therapeutics, Incyte, Regeneron, Merck, BMS, Pfizer, Neon Therapeutics, Calithera BioSciences, and TopAlliance BioSciences; grants and advisory board membership with ARMO BioSciences, Novartis, and CytomX Therapeutics; grants from Immune Deficiency Foundation (spouse).

Jeffrey Infante: Consultant for Armo Biosciences and Biomed Valley; employee of Janssen Oncology.

Sanjay Goel: Funding to conduct clinical trial from AstraZeneca.

Howard Burris: Consultant or advisor to Mersana, AstraZeneca, Bristol-Myers Squibb, FORMA Therapeutics, Janssen, Novartis, Roche/Genentech, and TG Therapeutics; research funding from Roche/Genentech, Bristol-Myers Squibb, Incyte, Tarveda Therapeutics, Mersana, AstraZeneca, Macrogenics, Novartis, Boehringer-Ingelheim, Eli Lilly, Seattle Genetics, AbbVie, Bayer, Celldex, Merck, Celgene, Agios, Jounce Therapeutics, Moderna Therapeutics, CytomX Therapeutics, GlaxoSmithKline, Verastem, Tesaro, Immunocore, Takeda, Millennium, BioMed Valley Discoveries, Pfizer, PTC Therapeutics, TG Therapuetics, Loxo, Vertex, eFFECTOR Therapetics, Janssen, Gilead Sciences, Valent Technologies, BioAtla, CicloMed, Harpoon Therapeutics, Jiangsu Hengrui Medicine, Revolution Medicines, Daiichi Sankyo, H3 Biomedicine, Neon Therapeutics, OncoMed, Regeneron, and Sanofi; expert testimony for Novartis.

Patricia LoRusso: Advisory boards for Genentech, AstraZeneca, Ipsen, AbbVie, Genmab, Roche, CytomX, and SOTIO; Data Safety Monitoring Boards for Agios, Five Prime, Halozyme, and Tyme.

Amy Weise: Speakers’ bureau for Array Biopharma.

Joseph Eder: No conflicts to disclose.

Brendan Curti: Institution receives research funding from AstraZeneca.

Chelsea Black, Shannon Marshall, Ikbel Achour, Rena May, Chris Morehouse, Kristen Pollizzi, Xuyang Song, Keith Steele, Nairouz Elgeioushi, Farzana Walcott, Joyson Karakunnel, and Michael Oberst are current or former employees of AstraZeneca and own stocks/shares in AstraZeneca. Susannah Barbee is a former employee of Amplimmune.

Figures

Fig. 1
Fig. 1
Response to MEDI0680 therapy. a Tumor size change from baseline in the as-treated population. b The timing and duration of response and onset of progressive disease or new lesions in the responding population. Blue bars indicate treatment initiation to censoring date or progression date. One patient with kidney cancer and PR had an ongoing response but did not have a disease assessment at the time of the last dose
Fig. 2
Fig. 2
Pharmacokinetic and receptor occupancy analysis of MEDI0680. a Pharmacokinetic analysis of MEDI0680 in patient serum. Data represent time points up to 150 days. Abbreviation: LLOQ lower limit of quantitation. b PD-1 receptor occupancy by MEDI0680 on CD45 RO+ CD3 T cells among patients treated at various drug doses and schedules, as indicated. Measurements were done at baseline, during the first cycle of MEDI0680 treatment, and on the first day after the completion of the first cycle
Fig. 3
Fig. 3
Peripheral and intratumoral measures of MEDI0680 activity. a Peripheral CD4+ and CD8+ T-cell activation and proliferation among treatment groups, as indicated. Shown are the fold changes in the percentages of CD4+ and CD8+ Ki67+ and CD4+ TEM CD38high/HLA-DRhigh cells in whole blood post-treatment. Abbreviation: TEM effector memory T cells. b Change in plasma cytokines among treatment groups, as indicated. Shown are the fold change in the plasma levels of IFNγ, CXCL-9, CXCL-10, and CXCL-11 at day 8 post-treatment with MEDI0680. c Examples of PD-L1+ and CD8+ IHC images (20× magnification) from matched pre- and on-treatment biopsies from an RCC patient. The tumor at screening is characterized by abundant CD8+ TILs and PD-L1 on immune cells but not on tumor cells (* symbols on IHC images). The on-treatment tumor has greater CD8+ T-cell infiltration and PD-L1 immunoreactivity on both immune and tumor cells (*). d Levels of CD8+ TILs in tumor biopsies pre- and on-treatment at various dose levels. Abbreviation: hpf high power field. (e) Log2 fold change in on-treatment versus pretreatment CD8A, IFNG, CXCL9, and GZMK gene expression in RCC and melanoma tumor biopsies. A 1.5-fold change is indicated by the dotted line

References

    1. Dong H, Zhu G, Tamada K, Chen L. B7-H1, a third member of the B7 family, co-stimulates T-cell proliferation and interleukin-10 secretion. Nat Med. 1999;5:1365–1369. doi: 10.1038/70932.
    1. Latchman Y, Wood CR, Chernova T, Chaudhary D, Borde M, Chernova I, et al. PD-L2 is a second ligand for PD-1 and inhibits T cell activation. Nat Immunol. 2001;2:261–268. doi: 10.1038/85330.
    1. Pardoll DM. The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer. 2012;12:252–264. doi: 10.1038/nrc3239.
    1. Zheng H, Liu X, Zhang J, Rice SJ, Wagman M, Kong Y, et al. Expression of PD-1 on CD4+ T cells in peripheral blood associates with poor clinical outcome in non-small cell lung cancer. Oncotarget. 2016;7:56233–56240.
    1. Goldberg MV, Maris CH, Hipkiss EL, Flies AS, Zhen L, Tuder RM, et al. Role of PD-1 and its ligand, B7-H1, in early fate decisions of CD8 T cells. Blood. 2007;110:186–192. doi: 10.1182/blood-2006-12-062422.
    1. Topalian SL, Drake CG, Pardoll DM. Immune checkpoint blockade: a common denominator approach to cancer therapy. Cancer Cell. 2015;27:450–461. doi: 10.1016/j.ccell.2015.03.001.
    1. OPDIVO® (nivolumab) US Prescribing Information. Updated November 2018. Available at: . Last Accessed 20 Feb 2019.
    1. KEYTRUDA® (pembrolizumab) US Prescribing Information. Updated December 2018. Available at: . Last Accessed 20 Feb 2019).
    1. Libtayo (cemiplimab) Prescribing Information. Available at: . Last Accessed 20 Feb 2019.
    1. Yarchoan M, Hopkins A, Jaffee EM. Tumor mutational burden and response rate to PD-1 inhibition. N Engl J Med. 2017;377:2500–2501. doi: 10.1056/NEJMc1713444.
    1. Lawrence MS, Stojanov P, Polak P, Kryukov GV, Cibulskis K, Sivachenko A, et al. Mutational heterogeneity in cancer and the search for new cancer genes. Nature. 2013;499:214–218. doi: 10.1038/nature12213.
    1. Yi M, Jiao D, Xu H, Liu Q, Zhao W, Han X, et al. Biomarkers for predicting efficacy of PD-1/ PD-L1 inhibitors. Mol Cancer. 2018;17:129. doi: 10.1186/s12943-018-0864-3.
    1. Bermejo MB, Jaffee EM, Davar D, Cardarelli J, Williams D, Phillips P. Phase 1b/2 study of nivolumab in combination with an anti–IL-8 monoclonal antibody, BMS-986253, in a biomarker-enriched population of patients with advanced cancer. J Clin Oncol. 2018;36(15_Suppl).
    1. Wrangle JM, Velcheti V, Patel MR, Garrett-Mayer E, Hill EG, Ravenel JG, et al. ALT-803, an IL-15 superagonist, in combination with nivolumab in patients with metastatic non-small cell lung cancer: a non-randomised, open-label, phase 1b trial. Lancet. 2018;19:694–704. doi: 10.1016/S1470-2045(18)30148-7.
    1. Zibelman MR, Macfarlane A, Alpaugh RK, Dulaimi E, Costello K, O’Neill J, et al. Effect of exogenous interferon-gamma (IFN-gamma) on peripheral blood immune markers as part of a phase I clinical trial of combined IFN-gamma with nivolumab (Nivo) in patients (pts) with select solid tumors. J Clin Oncol. 2017;35(7_Suppl):97.
    1. Chung V, Kos FJ, Hardwick N, et al. Evaluation of safety and efficacy of p53MVA vaccine combined with pembrolizumab in patients with advanced solid cancers. Clin Transl Oncol. 2019;21:363–372. doi: 10.1007/s12094-018-1932-2.
    1. Hamid O, Chow LQM, Tavakkoli F, Marshall S, Gribbin MJ, Karakunnel JJ, et al. Phase I, open-label study of MEDI0680, an anti-programmed cell death-1 (PD-1) antibody, in combination with MEDI4736, an anti-programmed cell death ligand-1 (PD-L1) antibody, in patients with advanced malignancies. J Clin Oncol. 2015;33(15_suppl) (Abstract TPS3087).
    1. Janku F, Tan DS, Martin-Liberal J, Takahashi S, Geva R, Gucalp A, et al. First-in-human study of FAZ053, an anti-PD-L1 mAb, alone and in combination with spartalizumab, an anti-PD-1 mAb, in patients with advanced malignancies. J Immunother Cancer. 2018;6(suppl 1) Abstract P651.
    1. Butte MJ, Keir ME, Phamduy TB, Sharpe AH, Freeman GJ. Programmed death-1 ligand 1 interacts specifically with the B7-1 costimulatory molecule to inhibit T cell responses. Immunity. 2007;27:111–122. doi: 10.1016/j.immuni.2007.05.016.
    1. Park JJ, Omiya R, Matsumura Y, Sakoda Y, Kuramasu A, Augustine MM, et al. B7-H1/CD80 interaction is required for the induction and maintenance of peripheral T-cell tolerance. Blood. 2010;116:1291–1298. doi: 10.1182/blood-2010-01-265975.
    1. Paterson AM, Brown KE, Keir ME, Vanguri VK, Riella LV, Chandraker A, et al. The programmed death-1 ligand 1:B7-1 pathway restrains diabetogenic effector T cells in vivo. J Immunol. 2011;187:1097–1105. doi: 10.4049/jimmunol.1003496.
    1. Mazza C, Escudier B, Albiges L. Nivolumab in renal cell carcinoma: latest evidence and clinical potential. Ther Adv Med Oncol. 2017;9:171–181. doi: 10.1177/1758834016679942.
    1. Weinstock M, McDermott D. Targeting PD-1/PD-L1 in the treatment of metastatic renal cell carcinoma. Ther Adv Urol. 2015;7:365–377. doi: 10.1177/1756287215597647.
    1. Ivashko IN, Kolesar JM. Pembrolizumab and nivolumab: PD-1 inhibitors for advanced melanoma. Am J Health Syst Pharm. 2016;73:193–201. doi: 10.2146/ajhp140768.
    1. Galluzzi L, Kroemer G, Eggermont A. Novel immune checkpoint blocker approved for the treatment of advanced melanoma. OncoImmunology. 2014;3:e967147. doi: 10.4161/21624011.2014.967147.
    1. Brahmer JR, Hammers H, Lipson EJ. Nivolumab: targeting PD-1 to bolster antitumor immunity. Future Oncol. 2015;11:1307–1326. doi: 10.2217/fon.15.52.
    1. McDermott David F., Lee Jae-Lyun, Szczylik Cezary, Donskov Frede, Malik Jahangeer, Alekseev Boris Yakovlevich, Larkin James M. G., Matveev Vsevolod Borisovich, Gafanov Rustem Airatovich, Tomczak Piotr, Tykodi Scott S., Geertsen Poul F., Wiechno Pawel J., Shin Sang Joon, Pouliot Frederic, Alonso Gordoa Teresa, Li Wenting, Perini Rodolfo F., Schloss Charles, Atkins Michael B. Pembrolizumab monotherapy as first-line therapy in advanced clear cell renal cell carcinoma (accRCC): Results from cohort A of KEYNOTE-427. Journal of Clinical Oncology. 2018;36(15_suppl):4500–4500. doi: 10.1200/JCO.2018.36.15_suppl.4500.
    1. Shim H. One target, different effects: a comparison of distinct therapeutic antibodies against the same targets. Exp Mol Med. 2011;43:539–549. doi: 10.3858/emm.2011.43.10.063.
    1. Kim MS, Lee SH, Song MY, Yoo TH, Lee BK, Kim YS. Comparative analyses of complex formation and binding sites between human tumor necrosis factor-alpha and its three antagonists elucidate their different neutralizing mechanisms. J Mol Biol. 2007;374:1374–1388. doi: 10.1016/j.jmb.2007.10.034.
    1. Emi Aikawa N, de Carvalho JF, Artur Almeida Silva C, Bonfá E. Immunogenicity of anti-TNF-alpha agents in autoimmune diseases. Clin Rev Allergy Immunol. 2010;38:82–89. doi: 10.1007/s12016-009-8140-3.
    1. Topalian SL, Sznol M, McDermott DF, Kluger HM, Carvajal RD, Sharfman WH, et al. Survival, durable tumor remission, and long-term safety in patients with advanced melanoma receiving nivolumab. J Clin Oncol. 2014;32:1020–1030. doi: 10.1200/JCO.2013.53.0105.
    1. Hamid O, Robert C, Daud A, Hodi FS, Hwu WJ, Kefford R, et al. Safety and tumor responses with lambrolizumab (anti-PD-1) in melanoma. N Engl J Med. 2013;369:134–144. doi: 10.1056/NEJMoa1305133.
    1. Robert C, Long GV, Brady B, Dutriaux C, Maio M, Mortier L, et al. Nivolumab in previously untreated melanoma without BRAF mutation. N Engl J Med. 2015;372:320–330. doi: 10.1056/NEJMoa1412082.
    1. Patnaik A, Kang SP, Rasco D, Papadopoulos KP, Elassaiss-Schaap J, Beeram M, et al. Phase I study of pembrolizumab (MK-3475; anti-PD-1 monoclonal antibody) in patients with advanced solid tumors. Clin Cancer Res. 2015;21:4286–4293. doi: 10.1158/1078-0432.CCR-14-2607.
    1. Robert C, Ribas A, Wolchok JD, Hodi FS, Hamid O, Kefford R, et al. Anti-programmed-death-receptor-1 treatment with pembrolizumab in ipilimumab-refractory advanced melanoma: a randomised dose-comparison cohort of a phase 1 trial. Lancet. 2014;384:1109–1117. doi: 10.1016/S0140-6736(14)60958-2.
    1. Rebelatto MC, Midha A, Mistry A, Sabalos C, Schechter N, Li X, et al. Development of a programmed cell death ligand-1 immunohistochemical assay validated for analysis of non-small cell lung cancer and head and neck squamous cell carcinoma. Diagn Pathol. 2016;11:95. doi: 10.1186/s13000-016-0545-8.
    1. Silva J-P, Vetterlein O, Jose J, Peters S, Kirby H. The S228P mutation prevents in vivo and in vitro IgG4 fab-arm exchange as demonstrated using a combination of novel quantitative immunoassays and physiological matrix preparation. J Biol Chem. 2015;290:5462–5469. doi: 10.1074/jbc.M114.600973.
    1. Song X, Gao X, Zheng Bo, Black C, Gribbin M, Karakunnel J, et al. Pharmacokinetics and pharmacodynamics of MEDI0680, a fully human anti-PD1 monoclonal antibody, in patients with advanced malignancies. Cancer Res. 2017;77(13 suppl) (Abstract 5045).
    1. Thurber GM, Schmidt MM, Wittrup KD. Antibody tumor penetration: transport opposed by systemic and antigen-mediated clearance. Adv Drug Deliv Rev. 2008;60:1421–1434. doi: 10.1016/j.addr.2008.04.012.
    1. Weber JS, Postow M, Lao CD, Schadendorf D. Management of adverse events following treatment with anti-programmed death-1 agents. Oncologist. 2016;21:1230–1240. doi: 10.1634/theoncologist.2016-0055.
    1. Wang M, Ma X, Guo L, Xia F. Safety and efficacy profile of pembrolizumab in solid cancer: pooled reanalysis based randomized controlled trials. Drug Des Devel Ther. 2017;11:2851–2860. doi: 10.2147/DDDT.S146286.
    1. Wong AC, Ma B. An update on the pharmacodynamics, pharmacokinetics, safety and clinical activity of nivolumab in the treatment of solid cancers. Expert Opin Drug Metab Toxicol. 2016;12:1255–1261. doi: 10.1080/17425255.2016.1223047.
    1. Topalian SL, Hodi FS, Brahmer JR, Gettinger SN, Smith DC, McDermott DF, et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med. 2012;366:2443–2454. doi: 10.1056/NEJMoa1200690.
    1. Choueiri TK, Fishman MN, Escudier B, McDermott DF, Drake CG, Kluger H, et al. Immunomodulatory activity of nivolumab in metastatic renal cell carcinoma. Clin Cancer Res. 2016;22:5461–5471. doi: 10.1158/1078-0432.CCR-15-2839.
    1. Motzer RJ, Rini BI, McDermott DF, Redman BG, Kuzel TM, Harrison MR, et al. Nivolumab for metastatic renal cell carcinoma: results of a randomized phase II trial. J Clin Oncol. 2015;33:1430–1437. doi: 10.1200/JCO.2014.59.0703.
    1. Huang AC, Postow MA, Orlowski RJ, Mick R, Bengsch B, Manne S, et al. T cell invigoration to tumor burden ratio associated with PD-1 response. Nature. 2017;545:60–65. doi: 10.1038/nature22079.
    1. Kamphorst AO, Pillai RN, Yang S, Nasti TH, Akondy RS, Wieland A, 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. 2017;114:4993–4998. doi: 10.1073/pnas.1705327114.
    1. Murphy KM, Reiner SL. Decision making in the immune system: the lineage decisions of helper T cells. Nat Rev Immunol. 2002;2:933–944. doi: 10.1038/nri954.
    1. Das R, Verma R, Sznol M, Boddupalli CS, Gettinger SN, Kluger H, et al. Combination therapy with anti-CTLA4 and anti-PD1 leads to distinct immunologic changes in-vivo. J Immunol. 2015;194:950–959. doi: 10.4049/jimmunol.1401686.
    1. Tumeh PC, Harview CL, Yearly JH, Shintaku IP, Taylor EJ, Robert L, et al. PD-1 blockade induces responses by inhibiting adaptive immune resistance. Nature. 2014;515:568–571. doi: 10.1038/nature13954.
    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. Diggs LP, Hsueh EC. Utility of PD-L1 immunohistochemistry assays for predicting PD-1/PD-L1 inhibitor response. Biomark Res. 2017;5:12. doi: 10.1186/s40364-017-0093-8.
    1. Festino L, Botti G, Lorigan P, Masucci GV, Hipp JD, Horak CE, et al. Cancer treatment with anti-PD-1/PD-L1 agents: is PD-L1 expression a biomarker for patient selection? Drugs. 2016;76:925–945. doi: 10.1007/s40265-016-0588-x.
    1. Taube J, Klein A, Brahmer J, Xu H, Pan X, Kim JH, et al. Association of PD-1, PD-1 ligands, and other features of the tumor immune microenvironment with response to anti-PD-1 therapy. Clin Cancer Res. 2014;20:5064–5074. doi: 10.1158/1078-0432.CCR-13-3271.
    1. Chen PL, Roh W, Reuben A, Cooper ZA, Spencer CN, Prieto PA, et al. Analysis of immune signatures in longitudinal tumor samples yields insight into biomarkers of response and mechanisms of resistance to immune checkpoint blockade. Cancer Discov. 2016;6:827–837. doi: 10.1158/-15-1545.
    1. Daud A, Loo K, Pauli ML, Sanchez-Rodriguez R, Sandoval PM, Taravati K, et al. Tumor immune profiling predicts response to anti-PD-1 therapy in human melanoma. J Clin Invest. 2016;126:3447–3452. doi: 10.1172/JCI87324.
    1. Inoue H, Park JH, Kiyotani K, Zewde M, Miyashita A, Jinnin M, et al. Intratumoral expression levels of PD-L1, GZMA, and HLA-A along with oligoclonal T cell expansion associate with response to nivolumab in metastatic melanoma. Oncoimmunology. 2016;5:e1204507. doi: 10.1080/2162402X.2016.1204507.
    1. Seiwert Tanguy Y., Burtness Barbara, Weiss Jared, Eder Joseph Paul, Yearley Jennifer, Murphy Erin, Nebozhyn Michael, McClanahan Terri, Ayers Mark, Lunceford Jared K., Mehra Ranee, Heath Karl, Cheng Jonathan D., Chow Laura Q. Inflamed-phenotype gene expression signatures to predict benefit from the anti-PD-1 antibody pembrolizumab in PD-L1+ head and neck cancer patients. Journal of Clinical Oncology. 2015;33(15_suppl):6017–6017. doi: 10.1200/jco.2015.33.15_suppl.6017.
    1. Ayers M, Luceford J, Nebozhyn M, Murphy E, Loboda A, Kaufman DR, et al. IFN-γ–related mRNA profile predicts clinical response to PD-1 blockade. J Clin Invest. 2017;127:2930–2940. doi: 10.1172/JCI91190.
    1. Ribas A, Shin DS, Zaretsky J, Frederiksen J, Cornish A, Avramis E, et al. PD-1 blockade expands intratumoral memory T cells. Cancer Immunol Res. 2016;4:194–203. doi: 10.1158/2326-6066.CIR-15-0210.
    1. Piha-Paul Sarina Anne, Bennouna Jaafar, Albright Andrew, Nebozhyn Michael, McClanahan Terrill, Ayers Mark, Lunceford Jared K., Ott Patrick Alexander. T-cell inflamed phenotype gene expression signatures to predict clinical benefit from pembrolizumab across multiple tumor types. Journal of Clinical Oncology. 2016;34(15_suppl):1536–1536. doi: 10.1200/JCO.2016.34.15_suppl.1536.
    1. Topalian SL, Taube JM, Anders RA, Pardoll DM. Mechanism-driven biomarkers to guide immune checkpoint blockade in cancer therapy. Nat Rev Cancer. 2016;16:275–287. doi: 10.1038/nrc.2016.36.

Source: PubMed

3
Předplatit