Anti-PD-1 monoclonal antibody MEDI0680 in a phase I study of patients with advanced solid malignancies
暂无分享,去创建一个
P. LoRusso | H. Burris | J. Infante | A. Naing | Kristen N. Pollizzi | I. Achour | J. Eder | S. Goel | B. Curti | R. May | M. Oberst | J. Karakunnel | A. Weise | S. Barbee | S. Marshall | Xuyang Song | C. Black | C. Morehouse | Keith H. Steele | N. Elgeioushi | F. Walcott | Chelsea Black
[1] Kongming Wu,et al. Biomarkers for predicting efficacy of PD-1/PD-L1 inhibitors , 2018, Molecular cancer.
[2] J. Waisman,et al. Evaluation of safety and efficacy of p53MVA vaccine combined with pembrolizumab in patients with advanced solid cancers , 2018, Clinical and Translational Oncology.
[3] J. Larkin,et al. Pembrolizumab monotherapy as first-line therapy in advanced clear cell renal cell carcinoma (accRCC): Results from cohort A of KEYNOTE-427. , 2018 .
[4] M. Carleton,et al. 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. , 2018 .
[5] M. Robinson,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. , 2018, The Lancet. Oncology.
[6] E. Jaffee,et al. Tumor Mutational Burden and Response Rate to PD-1 Inhibition. , 2017, The New England journal of medicine.
[7] T. Chan,et al. Tumor and Microenvironment Evolution during Immunotherapy with Nivolumab , 2017, Cell.
[8] Linghong Guo,et al. Safety and efficacy profile of pembrolizumab in solid cancer: pooled reanalysis based on randomized controlled trials , 2017, Drug design, development and therapy.
[9] J. Lunceford,et al. IFN-&ggr;–related mRNA profile predicts clinical response to PD-1 blockade , 2017, The Journal of clinical investigation.
[10] L. Roskos,et al. Abstract 5045: Pharmacokinetics and pharmacodynamics of MEDI0680, a fully human anti-PD1 monoclonal antibody, in patients with advanced malignancies , 2017 .
[11] G. Sica,et al. Proliferation of PD-1+ CD8 T cells in peripheral blood after PD-1–targeted therapy in lung cancer patients , 2017, Proceedings of the National Academy of Sciences.
[12] Jedd D. Wolchok,et al. T-cell invigoration to tumour burden ratio associated with anti-PD-1 response , 2017, Nature.
[13] E. Hsueh,et al. Utility of PD-L1 immunohistochemistry assays for predicting PD-1/PD-L1 inhibitor response , 2017, Biomarker Research.
[14] B. Escudier,et al. Nivolumab in renal cell carcinoma: latest evidence and clinical potential , 2017, Therapeutic advances in medical oncology.
[15] E. Plimack,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. , 2017 .
[16] Nicole Schechter,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 , 2016, Diagnostic Pathology.
[17] D. Schadendorf,et al. Management of Adverse Events Following Treatment With Anti-Programmed Death-1 Agents. , 2016, The oncologist.
[18] L. Nardo,et al. Tumor immune profiling predicts response to anti-PD-1 therapy in human melanoma. , 2016, The Journal of clinical investigation.
[19] B. Ma,et al. An update on the pharmacodynamics, pharmacokinetics, safety and clinical efficacy of nivolumab in the treatment of solid cancers , 2016, Expert opinion on drug metabolism & toxicology.
[20] L. Chin,et al. Analysis of Immune Signatures in Longitudinal Tumor Samples Yields Insight into Biomarkers of Response and Mechanisms of Resistance to Immune Checkpoint Blockade. , 2016, Cancer discovery.
[21] Yusuke Nakamura,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 , 2016, Oncoimmunology.
[22] J. Hipp,et al. Cancer Treatment with Anti-PD-1/PD-L1 Agents: Is PD-L1 Expression a Biomarker for Patient Selection? , 2016, Drugs.
[23] J. Lunceford,et al. T-cell inflamed phenotype gene expression signatures to predict clinical benefit from pembrolizumab across multiple tumor types. , 2016 .
[24] Junjia Zhu,et al. Expression of PD-1 on CD4+ T cells in peripheral blood associates with poor clinical outcome in non-small cell lung cancer , 2016, Oncotarget.
[25] P. Ross-Macdonald,et al. Immunomodulatory Activity of Nivolumab in Metastatic Renal Cell Carcinoma , 2016, Clinical Cancer Research.
[26] J. Taube,et al. Mechanism-driven biomarkers to guide immune checkpoint blockade in cancer therapy , 2016, Nature Reviews Cancer.
[27] J. Kolesar,et al. Pembrolizumab and nivolumab: PD-1 inhibitors for advanced melanoma. , 2016, American journal of health-system pharmacy : AJHP : official journal of the American Society of Health-System Pharmacists.
[28] Dana Pe'er,et al. PD-1 Blockade Expands Intratumoral Memory T Cells , 2016, Cancer Immunology Research.
[29] D. McDermott,et al. Targeting PD-1/PD-L1 in the treatment of metastatic renal cell carcinoma , 2015, Therapeutic advances in urology.
[30] L. Chow,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. , 2015 .
[31] J. Lunceford,et al. Inflamed-phenotype gene expression signatures to predict benefit from the anti-PD-1 antibody pembrolizumab in PD-L1+ head and neck cancer patients. , 2015 .
[32] Lon Smith,et al. Phase I Study of Pembrolizumab (MK-3475; Anti–PD-1 Monoclonal Antibody) in Patients with Advanced Solid Tumors , 2015, Clinical Cancer Research.
[33] R. Motzer,et al. Nivolumab for Metastatic Renal Cell Carcinoma: Results of a Randomized Phase II Trial. , 2015, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.
[34] C. Drake,et al. Immune checkpoint blockade: a common denominator approach to cancer therapy. , 2015, Cancer cell.
[35] E. Lipson,et al. Nivolumab: targeting PD-1 to bolster antitumor immunity. , 2015, Future oncology.
[36] J. Wolchok,et al. Combination Therapy with Anti–CTLA-4 and Anti–PD-1 Leads to Distinct Immunologic Changes In Vivo , 2015, The Journal of Immunology.
[37] D. Schadendorf,et al. Nivolumab in previously untreated melanoma without BRAF mutation. , 2015, The New England journal of medicine.
[38] O. Vetterlein,et al. 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 , 2015, The Journal of Biological Chemistry.
[39] L. Galluzzi,et al. Novel immune checkpoint blocker approved for the treatment of advanced melanoma , 2014, Oncoimmunology.
[40] R. Emerson,et al. PD-1 blockade induces responses by inhibiting adaptive immune resistance , 2014, Nature.
[41] Antoni Ribas,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 , 2014, The Lancet.
[42] J. Taube,et al. Association of PD-1, PD-1 Ligands, and Other Features of the Tumor Immune Microenvironment with Response to Anti–PD-1 Therapy , 2014, Clinical Cancer Research.
[43] David C. Smith,et al. Survival, durable tumor remission, and long-term safety in patients with advanced melanoma receiving nivolumab. , 2014, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.
[44] Antoni Ribas,et al. Safety and tumor responses with lambrolizumab (anti-PD-1) in melanoma. , 2013, The New England journal of medicine.
[45] Steven A. Roberts,et al. Mutational heterogeneity in cancer and the search for new cancer genes , 2014 .
[46] David C. Smith,et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. , 2012, The New England journal of medicine.
[47] Drew M. Pardoll,et al. The blockade of immune checkpoints in cancer immunotherapy , 2012, Nature Reviews Cancer.
[48] H. Shim. One target, different effects: a comparison of distinct therapeutic antibodies against the same targets , 2011, Experimental & Molecular Medicine.
[49] G. Freeman,et al. The Programmed Death-1 Ligand 1:B7-1 Pathway Restrains Diabetogenic Effector T Cells In Vivo , 2011, The Journal of Immunology.
[50] S. Anand,et al. B7-H1/CD80 interaction is required for the induction and maintenance of peripheral T-cell tolerance. , 2010, Blood.
[51] E. Bonfá,et al. Immunogenicity of Anti-TNF-α Agents in Autoimmune Diseases , 2010, Clinical reviews in allergy & immunology.
[52] K Dane Wittrup,et al. Antibody tumor penetration: transport opposed by systemic and antigen-mediated clearance. , 2008, Advanced drug delivery reviews.
[53] Yong-Sung Kim,et al. Comparative analyses of complex formation and binding sites between human tumor necrosis factor-alpha and its three antagonists elucidate their different neutralizing mechanisms. , 2007, Journal of molecular biology.
[54] G. Freeman,et al. Programmed death-1 ligand 1 interacts specifically with the B7-1 costimulatory molecule to inhibit T cell responses. , 2007, Immunity.
[55] D. Getnet,et al. Role of PD-1 and its ligand, B7-H1, in early fate decisions of CD8 T cells. , 2007, Blood.
[56] Kenneth M. Murphy,et al. Decision making in the immune system: The lineage decisions of helper T cells , 2002, Nature Reviews Immunology.
[57] G. Freeman,et al. PD-L2 is a second ligand for PD-1 and inhibits T cell activation , 2001, Nature Immunology.
[58] G. Zhu,et al. B7-H1, a third member of the B7 family, co-stimulates T-cell proliferation and interleukin-10 secretion , 1999, Nature Medicine.
[59] J. Mackintosh. LETTERS OF DR. MACKINTOSH.: (Vide LANCET, No. 494, p. 662.) , 1833 .
[60] Hilde van der Togt,et al. Publisher's Note , 2003, J. Netw. Comput. Appl..