Anti-CSF-1R emactuzumab in combination with anti-PD-L1 atezolizumab in advanced solid tumor patients naïve or experienced for immune checkpoint blockade

Carlos Gomez-Roca, Philippe Cassier, Dmitriy Zamarin, Jean-Pascal Machiels, Jose Luis Perez Gracia, F Stephen Hodi, Alvaro Taus, Maria Martinez Garcia, Valentina Boni, Joseph P Eder, Navid Hafez, Ryan Sullivan, David Mcdermott, Stephane Champiat, Sandrine Aspeslagh, Catherine Terret, Anna-Maria Jegg, Wolfgang Jacob, Michael A Cannarile, Carola Ries, Konstanty Korski, Francesca Michielin, Randolph Christen, Galina Babitzki, Carl Watson, Georgina Meneses-Lorente, Martin Weisser, Dominik Rüttinger, Jean-Pierre Delord, Aurelien Marabelle, Carlos Gomez-Roca, Philippe Cassier, Dmitriy Zamarin, Jean-Pascal Machiels, Jose Luis Perez Gracia, F Stephen Hodi, Alvaro Taus, Maria Martinez Garcia, Valentina Boni, Joseph P Eder, Navid Hafez, Ryan Sullivan, David Mcdermott, Stephane Champiat, Sandrine Aspeslagh, Catherine Terret, Anna-Maria Jegg, Wolfgang Jacob, Michael A Cannarile, Carola Ries, Konstanty Korski, Francesca Michielin, Randolph Christen, Galina Babitzki, Carl Watson, Georgina Meneses-Lorente, Martin Weisser, Dominik Rüttinger, Jean-Pierre Delord, Aurelien Marabelle

Abstract

Background: This phase 1b study (NCT02323191) evaluated the safety, antitumor activity, pharmacokinetics, and pharmacodynamics of colony-stimulating factor-1 receptor-blocking monoclonal antibody (mAb) emactuzumab in combination with the programmed cell death-1 ligand (PD-L1)-blocking mAb atezolizumab in patients with advanced solid tumors naïve or experienced for immune checkpoint blockers (ICBs).

Methods: Emactuzumab (500-1350 mg flat) and atezolizumab (1200 mg flat) were administered intravenously every 3 weeks. Dose escalation of emactuzumab was conducted using the 3+3 design up to the maximum tolerated dose (MTD) or optimal biological dose (OBD). Extension cohorts to evaluate pharmacodynamics and clinical activity were conducted in metastatic ICB-naive urothelial bladder cancer (UBC) and ICB-pretreated melanoma (MEL), non-small cell lung cancer (NSCLC) and UBC patients.

Results: Overall, 221 patients were treated. No MTD was reached and the OBD was determined at 1000 mg of emactuzumab in combination with 1200 mg of atezolizumab. Grade ≥3 treatment-related adverse events occurred in 25 (11.3%) patients of which fatigue and rash were the most common (14 patients (6.3%) each). The confirmed objective response rate (ORR) was 9.8% for ICB-naïve UBC, 12.5% for ICB-experienced NSCLC, 8.3% for ICB-experienced UBC and 5.6% for ICB-experienced MEL patients, respectively. Tumor biopsy analyses demonstrated increased activated CD8 +tumor infiltrating T lymphocytes (TILs) associated with clinical benefit in ICB-naïve UBC patients and less tumor-associated macrophage (TAM) reduction in ICB-experienced compared with ICB-naïve patients.

Conclusion: Emactuzumab in combination with atezolizumab demonstrated a manageable safety profile with increased fatigue and skin rash over usual atezolizumab monotherapy. A considerable ORR was particularly seen in ICB-experienced NSCLC patients. Increase ofCD8 +TILs under therapy appeared to be associated with persistence of a TAM subpopulation.

Keywords: Clinical Trials as Topic; Drug Therapy, Combination; Immunotherapy; Macrophages; T-Lymphocytes.

Conflict of interest statement

Competing interests: CG-R: Invited Speaker: BMS, Eisai, Pierre Fabre, Roche/Genentech; Coordinating PI: BMS; Steering Committee Member: BMS; Local PI: Foundation Medicine; Steering Committee Member: Genentech; Research Grant: Roche/Genentech; AM: Stock ownership Pegascy, Hifibio Therapeutics, Shattuck Labs, Centessa Pharmaceuticals; Honoraria: BMS, AstraZeneca/MedImmune, Oncovir; Consulting and advisory activities: Lytix Biopharma, Eisai, Pierre Fabre, AstraZeneca, Servier, Roche, Redx Pharma, Sotio, Innate Pharma, ImCheck Therapeutics, MSD, OSE Immunotherapeutics, HIFIBIO Therapeutics, MedinCell, Centessa Pharmaceuticals; Speaker’s bureau: BMS; Research funding: BMS, Boehringer Ingelheim, Transgene, MSD; Travel expenses: MSD, AstraZeneca; SC: Honoraria: Amgen, AstraZeneca, BMS, EISAI, Janssen, MSD, Novartis and Roche; Principal Investigator of Clinical Trials for: Amgen, MSD, Sanofi Aventis, Transgene; Advisory Board: Alderaan Biotechnology, Amgen, AstraZeneca, Oncovita, Seagen, Ultrahuman; Travel and congress: AstraZeneca, MSD, Roche; Principal/sub-investigator of clinical trials for: Abbvie, Adaptimmune, Adlai Nortye USA Inc, Aduro Biotech, Agios Pharmaceuticals, Amgen, Argen-X Bvba, Astex Pharmaceuticals, Astra Zeneca Ab, Aveo, Basilea Pharmaceutica International Ltd, Bayer Healthcare Ag, Bbb Technologies Bv, Beigene, BicycleTx Ltd, Blueprint Medicines, Boehringer Ingelheim, Boston Pharmaceuticals, Bristol Myers Squibb, Ca, Celgene Corporation, Chugai Pharmaceutical Co, Clovis Oncology, Cullinan-Apollo, Curevac, Daiichi Sankyo, Debiopharm, Eisai, Eisai Limited, Eli Lilly, Exelixis, Faron Pharmaceuticals Ltd, Forma Tharapeutics, Gamamabs, Genentech, Glaxosmithkline, H3 Biomedicine, Hoffmann La Roche Ag, Imcheck Therapeutics, Innate Pharma, Institut De Recherche Pierre Fabre, Iris Servier, Iteos Belgium SA, Janssen Cilag, Janssen Research Foundation, Kura Oncology, Kyowa Kirin Pharm. Dev, Lilly France, Loxo Oncology, Lytix Biopharma As, Medimmune, Menarini Ricerche, Merck Sharp & Dohme Chibret, Merrimack Pharmaceuticals, Merus, Millennium Pharmaceuticals, Molecular Partners Ag, Nanobiotix, Nektar Therapeutics, Novartis Pharma, Octimet Oncology Nv, Oncoethix, Oncopeptides, Orion Pharma, Ose Pharma, Pfizer, Pharma Mar, Pierre Fabre, Medicament, Roche, Sanofi Aventis, Seattle Genetics, Sotio A.S, Syros Pharmaceuticals, Taiho Pharma, Tesaro, Turning Point Therapeutics, Xencor; Research Grants from: Astrazeneca, BMS, Boehringer Ingelheim, GSK, INCA, Janssen Cilag, Merck, Novartis, Pfizer, Roche, SanofiNon-financial support (drug supplied) from Astrazeneca, Bayer, BMS, Boringher Ingelheim, GSK, Medimmune, Merck, NH TherAGuiX, Pfizer, Roche; SA: Speakers bureau: Pfizer, Roche, Sanofi and BMSAdvisory board: Sanofi; PC: Honoraria: Novartis, Roche/Genentech, Amgen, Astra Zeneca, Merck Serono; Research Funding: Novartis, Roche/Genentech, Lilly, lueprint Medicines, Bayer, Astra Zeneca, Celgene, Plexxikon, Abbvie, BMS, Merck Serono, Merck Sharp and Dohme, Taiho Pharmaceutical, Toray Industries, Transgene, Loxo, GSK, Innate Pharma, Janssen; Travel expenses: Roche, Amgen, Novartis, BMS, MSD, Netris Pharma, Bayer, Merck Serono; DZ: Reports research support from: Roche, Astra Zeneca, and Plexxikon; Personal/consultancy fees from Synlogic Therapeutics, GSK, Roche, Xencor, Memgen, Immunos, Celldex, Calidi, and Agenus; J-PM: Advisory board member or speaker with honoraria: Pfizer, Roche, Astra/Zeneca, Bayer, Innate, Merck Serono, Boerhinger, BMS, Novartis, Janssen, Incyte, Cue Biopharma, ALX Oncology, iTEOS, eTheRNATravel expenses: Amgen, BMS, Pfizer, MSDData safety monitoring board with honoraria: Debio, Nanobiotix, Psioxus; Uncompensated advisory role: MSD; JLPG; Research grants and support: Roche, BMS, MSD, Seattle Genetics. Speakers bureau and advisory boards: Roche, BMS, Ipsen, MSD, Seattle Genetics. Travel support: Roche, MSD, BMS; FSH: Consulting: BMS, Merck, EMD Serono, Novartis, Sanofi, Psioxus Therapeutics, Pieris Pharmacutical, Corner Therapeutics, Eisai, Idera, Takeda, Genentech/Roche; Advisory Board: Compass Therapeutics, Apricity Scientific, Pionyr, Torque, Rheos, Bicara, Checkpoint Therapeutics, Bioentre, Gossamer, Iovance; ATG: Personal fees from: Boehringer-Ingelheim, BMS, MSD, Roche, Pfizer, Astra Zeneca, Tesaro-GSK and non-financial support from Boehringer-Ingelheim, Lilly and RocheMaria Martinez Garcia; Research grants and support: Roche, BMS, MSD, Seattle Genetics; Speakers bureau and advisory boards: Roche, BMS, Ipsen, MSD, Seattle Genetics; Travel support: Roche, MSD, BMS; VB: Consulting or Advisory Role: Puma Biotechnology; Ideaya Biosciences; Loxo Therapeutics, CytomX Therapeutics; Guidepoint; Oncoart; Amunix; Institutional financial support for clinical trials from: Abbvie, ACEO, Adaptaimmune, Amcure, AMGEN, AstraZeneca, BMS, Cytomx, GSK, Genentech/Roche, H3, Incyte, Janssen, Kura, Lilly, Loxo, Nektar, Macrogenics, Menarini, Merck, Merus, Nanobiotix, Novartis, Pfizer, PharmaMar, Principia, PUMA, Sanofi, Taiho, Tesaro, BeiGene, Transgene, Takeda, Incyte, Innovio, MSD, PsiOxus, Seattle Genetics, Mersana, GSK, Daiichi, Nektar, Astellas, ORCA, Boston Therapeutics, Dynavax, DebioPharm, Boehringen Ingelheim, Regeneron, Millenium, Synthon, Spectrum, Rigontec, Zenith; JPE: The author declares no potential conflicts of interest. NH: The author declares no potential conflicts of interest. RS: Consultant/advisory boards: Asana Biosciences, AstraZeneca, Bristol-Myers Squibb, Eisai, Iovance, Merck, Novartis, OncoSec, Pfizer, Replimune; Research funding: Amgen, Merck; DM: Consulting and honoraria: BMS, Pfizer, Merck, Alkermes Inc., EMD Serono, Eli Lilly and Company, Iovance, Eisai Inc., Werewolf Therapeutics, Calithera Biosciences; Research support: BMS, Merck, Genentech, Pfizer, Exelixis, X4 Pharma, Alkermes Inc; MAC: Sponsor employee and sponsor stock ownership; A-MJ: Former sponsor employee and has patent issued in the use of emactuzumab; WJ: Sponsor employee and sponsor stock ownership; CR: Former Roche employee and has patent issued in the use of emactuzumab. Consultant for Verseau Therapeutics, Ridgeline Discovery, iOmx Therapeutics AG; KK: Sponsor employee and Roche stocks; GB: Sponsor employee; FM: Sponsor employee; RC: Sponsor employee and Roche stocks; CW: Sponsor consultantGeorgina Meneses-LorenteSponsor employee; MW: Sponsor employee, stock options, and has patent issued in the use of emactuzumab; DR: Sponsor employee, sponsor stock ownership and has patent issued in the use of emactuzumab; J-PD: Consulting/Advisory: Novartis, Roche/Genentech, BMS, MSD; Research funding: Genentech, BMS, MSD, Astra Zeneca, Transgene; CT: Research funding GSK, travel expenses Mundipharma.

© Author(s) (or their employer(s)) 2022. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.

Figures

Figure 1
Figure 1
Flow diagram of study design, patient enrolment and emactuzumab dose. All patients received 1200 mg of atezolizumab in combination with emactuzumab q3w. AE, adverse event; GC, gastric cancer; MEL, melanoma; n, number of patients; NSCLC, non-small cell lung cancer; OvCa, ovarian carcinoma; PD, progressive disease; STS, soft tissue sarcoma; TNBC, triple-negative breast cancer; UBC, urothelial bladder cancer cancer.
Figure 2
Figure 2
Waterfall plot based on RECIST criteria per investigator assessment (A) ICB-naïve UBC patients (B) ICB-experienced melanoma patients (C) ICB-experienced NSCLC patients (D) ICB-experienced UBC patients. ICB, immune checkpoint blocker; NSCLC, non-small cell lung cancer; UBC, urothelial bladder cancer.
Figure 3
Figure 3
Change from baseline of tumor-associated macrophages and tumor-infiltrating T cells in paired biopsies and comparison of progressive disease patients versus non-progressive disease patients in the UBC ICB-naïve cohort. Clinical responses are indicated. CR, complete response; ICB, immune checkpoint blocker; PD, progressive disease; PR, partial response; SD, stable disease; UBC, urothelial bladder cancer.
Figure 4
Figure 4
Baseline comparison of tumor-associated macrophages and tumor-infiltrating T cells and comparison of progressive disease (PD) patients versus non-PD patients in the ICB-naïve UBC and in the ICB-experienced UBC, NSCLC and MEL cohort. Clinical responses are indicated. ICB, immune checkpoint blocker; MEL, melanoma; NSCLC, non-small cell lung cancer; PR, partial response; SD, stable disease; UBC, urothelial bladder cancer.

References

    1. Cohen J, Sznol M. Therapeutic combinations of immune-modulating antibodies in melanoma and beyond. Semin Oncol 2015;42:488–94. 10.1053/j.seminoncol.2015.02.014
    1. Pennock GK, Chow LQM. The evolving role of immune checkpoint inhibitors in cancer treatment. Oncologist 2015;20:812–22. 10.1634/theoncologist.2014-0422
    1. West HJ. JAMA oncology patient page. Immune Checkpoint Inhibitors. JAMA Oncol 2015;1:115.
    1. Zhang Q-wen, Liu L, Gong C-yang, et al. . Prognostic significance of tumor-associated macrophages in solid tumor: a meta-analysis of the literature. PLoS One 2012;7:e50946. 10.1371/journal.pone.0050946
    1. Komohara Y, Jinushi M, Takeya M. Clinical significance of macrophage heterogeneity in human malignant tumors. Cancer Sci 2014;105:1–8. 10.1111/cas.12314
    1. Ries CH, Cannarile MA, Hoves S, et al. . Targeting tumor-associated macrophages with anti-CSF-1R antibody reveals a strategy for cancer therapy. Cancer Cell 2014;25:846–59. 10.1016/j.ccr.2014.05.016
    1. Biswas SK, Mantovani A. Macrophage plasticity and interaction with lymphocyte subsets: cancer as a paradigm. Nat Immunol 2010;11:889–96. 10.1038/ni.1937
    1. Ruffell B, Coussens LM. Macrophages and therapeutic resistance in cancer. Cancer Cell 2015;27:462–72. 10.1016/j.ccell.2015.02.015
    1. Yu J, Green MD, Li S, et al. . Liver metastasis restrains immunotherapy efficacy via macrophage-mediated T cell elimination. Nat Med 2021;27:152–64. 10.1038/s41591-020-1131-x
    1. Gubin MM, Esaulova E, Ward JP, et al. . high-dimensional analysis delineates myeloid and lymphoid compartment remodeling during successful immune-checkpoint cancer therapy. Cell 2018;175:1443. 10.1016/j.cell.2018.11.003
    1. Neubert NJ, Schmittnaegel M, Bordry N, et al. . T cell-induced CSF1 promotes melanoma resistance to PD1 blockade. Sci Transl Med 2018;10. 10.1126/scitranslmed.aan3311. [Epub ahead of print: 11 04 2018].
    1. Peranzoni E, Lemoine J, Vimeux L, et al. . Macrophages impede CD8 T cells from reaching tumor cells and limit the efficacy of anti-PD-1 treatment. Proc Natl Acad Sci U S A 2018;115:E4041–50. 10.1073/pnas.1720948115
    1. Zhu Y, Knolhoff BL, Meyer MA, et al. . CSF1/CSF1R blockade reprograms tumor-infiltrating macrophages and improves response to T-cell checkpoint immunotherapy in pancreatic cancer models. Cancer Res 2014;74:5057–69. 10.1158/0008-5472.CAN-13-3723
    1. Shi G, Yang Q, Zhang Y, et al. . Modulating the tumor microenvironment via oncolytic viruses and CSF-1R inhibition synergistically enhances anti-PD-1 immunotherapy. Mol Ther 2019;27:244–60. 10.1016/j.ymthe.2018.11.010
    1. Ries CH, Hoves S, Cannarile MA, et al. . CSF-1/CSF-1R targeting agents in clinical development for cancer therapy. Curr Opin Pharmacol 2015;23:45–51. 10.1016/j.coph.2015.05.008
    1. Cassier PA, Italiano A, Gomez-Roca CA, et al. . CSF1R inhibition with emactuzumab in locally advanced diffuse-type tenosynovial giant cell tumours of the soft tissue: a dose-escalation and dose-expansion phase 1 study. Lancet Oncol 2015;16:949–56. 10.1016/S1470-2045(15)00132-1
    1. Cassier PA, Italiano A, Gomez-Roca C, et al. . Long-term clinical activity, safety and patient-reported quality of life for emactuzumab-treated patients with diffuse-type tenosynovial giant-cell tumour. Eur J Cancer 2020;141:162–70. 10.1016/j.ejca.2020.09.038
    1. Gomez-Roca CA, Italiano A, Le Tourneau C, et al. . Phase I study of emactuzumab single agent or in combination with paclitaxel in patients with advanced/metastatic solid tumors reveals depletion of immunosuppressive M2-like macrophages. Ann Oncol 2019;30:1381–92. 10.1093/annonc/mdz163
    1. Herbst RS, Soria J-C, Kowanetz M, et al. . Predictive correlates of response to the anti-PD-L1 antibody MPDL3280A in cancer patients. Nature 2014;515:563–7. 10.1038/nature14011
    1. Rosenberg JE, Hoffman-Censits J, Powles T, 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;387:1909–20. 10.1016/S0140-6736(16)00561-4
    1. Horn L, Mansfield AS, Szczęsna A, et al. . First-line atezolizumab plus chemotherapy in extensive-stage small-cell lung cancer. N Engl J Med 2018;379:2220–9. 10.1056/NEJMoa1809064
    1. Schmid P, Adams S, Rugo HS, et al. . Atezolizumab and nab-paclitaxel in advanced triple-negative breast cancer. N Engl J Med 2018;379:2108–21. 10.1056/NEJMoa1809615
    1. Finn RS, Qin S, Ikeda M, et al. . Atezolizumab plus bevacizumab in unresectable hepatocellular carcinoma. N Engl J Med 2020;382:1894–905. 10.1056/NEJMoa1915745
    1. West H, McCleod M, Hussein M, et al. . Atezolizumab in combination with carboplatin plus nab-paclitaxel chemotherapy compared with chemotherapy alone as first-line treatment for metastatic non-squamous non-small-cell lung cancer (IMpower130): a multicentre, randomised, open-label, phase 3 trial. Lancet Oncol 2019;20:924–37. 10.1016/S1470-2045(19)30167-6
    1. Smart K, Bröske A-M, Rüttinger D, et al. . PK/PD mediated dose optimization of Emactuzumab, a CSF1R inhibitor, in patients with advanced solid tumors and diffuse-type Tenosynovial giant cell tumor. Clin Pharmacol Ther 2020;108:616–24. 10.1002/cpt.1964
    1. Eisenhauer EA, Therasse P, Bogaerts J, et al. . New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1). Eur J Cancer 2009;45:228–47. 10.1016/j.ejca.2008.10.026
    1. Fehrenbacher L, Spira A, Ballinger M, et al. . Atezolizumab versus docetaxel for patients with previously treated non-small-cell lung cancer (POPLAR): a multicentre, open-label, phase 2 randomised controlled trial. Lancet 2016;387:1837–46. 10.1016/S0140-6736(16)00587-0
    1. Cassier PA, Italiano A, Gomez-Roca CA, et al. . CSF1R inhibition with emactuzumab in locally advanced diffuse-type tenosynovial giant cell tumours of the soft tissue: a dose-escalation and dose-expansion phase 1 study. Lancet Oncol 2015;16:949–56. 10.1016/S1470-2045(15)00132-1
    1. Harb WA, Johnson ML, Goldman JW. Phase 1b/2 dose-escalation study of ARRY-382, an oral inhibitor of colony-stimulating factor-1 receptor (CSF1R), in combination with pembrolizumab for treatment of patients with advanced solid tumors. Journal for Immunotherapy of Cancer 2017;5.
    1. Wainberg Z, Piha‐Paul S, Luke J. First‐in‐human phase 1 dose escalation and expansion of a novel combination, anti–CSF‐1 receptor (cabiralizumab) plus anti– PD‐1 (nivolumab), in patients with advanced solid tumors. Journal for ImmunoTherapy of Cancer 2017;5:89. 10.13140/RG.2.2.28962.53443
    1. Calvo A, Joensuu H, Sebastian M, et al. . Phase Ib/II study of lacnotuzumab (MCS110) combined with spartalizumab (PDR001) in patients (PTS) with advanced tumors. J Clin Oncol 2018;36:3014. 10.1200/JCO.2018.36.15_suppl.3014
    1. Razak AR, Cleary JM, Moreno V, et al. . Safety and efficacy of AMG 820, an anti-colony-stimulating factor 1 receptor antibody, in combination with pembrolizumab in adults with advanced solid tumors. J Immunother Cancer 2020;8:e001006. 10.1136/jitc-2020-001006
    1. Cassier PA, Garin G, Eberst L, et al. . MEDIPLEX: a phase 1 study of durvalumab (D) combined with pexidartinib (P) in patients (PTS) with advanced pancreatic ductal adenocarcinoma (PDAC) and colorectal cancer (CRC). JCO 2019;37:2579. 10.1200/JCO.2019.37.15_suppl.2579
    1. Falchook GS, Peeters M, Rottey S, et al. . A phase 1a/1b trial of CSF-1R inhibitor LY3022855 in combination with durvalumab or tremelimumab in patients with advanced solid tumors. Invest New Drugs 2021;39:1284–97. 10.1007/s10637-021-01088-4
    1. Borghaei H, Paz-Ares L, Horn L, et al. . Nivolumab versus docetaxel in advanced nonsquamous non-small-cell lung cancer. N Engl J Med 2015;373:1627–39. 10.1056/NEJMoa1507643
    1. Brahmer J, Reckamp KL, Baas P, et al. . Nivolumab versus docetaxel in advanced squamous-cell non-small-cell lung cancer. N Engl J Med 2015;373:123–35. 10.1056/NEJMoa1504627
    1. Robert C, Schachter J, Long GV, et al. . Pembrolizumab versus ipilimumab in advanced melanoma. N Engl J Med 2015;372:2521–32. 10.1056/NEJMoa1503093
    1. Bellmunt J, de Wit R, Vaughn DJ, et al. . Pembrolizumab as second-line therapy for advanced urothelial carcinoma. N Engl J Med 2017;376:1015–26. 10.1056/NEJMoa1613683
    1. Rittmeyer A, Barlesi F, Waterkamp D, et al. . Atezolizumab versus docetaxel in patients with previously treated non-small-cell lung cancer (oak): a phase 3, open-label, multicentre randomised controlled trial. Lancet 2017;389:255–65. 10.1016/S0140-6736(16)32517-X
    1. Barlesi F, Vansteenkiste J, Spigel D, et al. . Avelumab versus docetaxel in patients with platinum-treated advanced non-small-cell lung cancer (JAVELIN lung 200): an open-label, randomised, phase 3 study. Lancet Oncol 2018;19:1468–79. 10.1016/S1470-2045(18)30673-9
    1. Powles T, Durán I, van der Heijden MS, et al. . Atezolizumab versus chemotherapy in patients with platinum-treated locally advanced or metastatic urothelial carcinoma (IMvigor211): a multicentre, open-label, phase 3 randomised controlled trial. Lancet 2018;391:748–57. 10.1016/S0140-6736(17)33297-X
    1. Wolchok JD, Rollin L, Larkin J. Nivolumab and ipilimumab in advanced melanoma. N Engl J Med 2017;377:2503–4. 10.1056/NEJMc1714339
    1. Larkin J, Chiarion-Sileni V, Gonzalez R, et al. . Five-year survival with combined nivolumab and ipilimumab in advanced melanoma. N Engl J Med 2019;381:1535–46. 10.1056/NEJMoa1910836
    1. Martini DJ, Lalani A-KA, Bossé D, et al. . Response to single agent PD-1 inhibitor after progression on previous PD-1/PD-L1 inhibitors: a case series. J Immunother Cancer 2017;5:66. 10.1186/s40425-017-0273-y
    1. Fujita K, Uchida N, Yamamoto Y, et al. . Retreatment with anti-PD-L1 antibody in advanced non-small cell lung cancer previously treated with anti-PD-1 antibodies. Anticancer Res 2019;39:3917–21. 10.21873/anticanres.13543
    1. Fujita K, Yamamoto Y, Kanai O, et al. . Retreatment with anti-PD-1 antibody in non-small cell lung cancer patients previously treated with anti-PD-L1 antibody. Thorac Cancer 2020;11:15–18. 10.1111/1759-7714.13241
    1. Evren E, Ringqvist E, Willinger T. Origin and ontogeny of lung macrophages: from mice to humans. Immunology 2020;160:126–38. 10.1111/imm.13154
    1. Salvagno C, Ciampricotti M, Tuit S, et al. . Therapeutic targeting of macrophages enhances chemotherapy efficacy by unleashing type I interferon response. Nat Cell Biol 2019;21:511–21. 10.1038/s41556-019-0298-1
    1. Lu D, Ni Z, Liu X, et al. . Beyond T cells: understanding the role of PD-1/PD-L1 in tumor-associated macrophages. J Immunol Res 2019;2019:1–7. 10.1155/2019/1919082
    1. Xiong H, Mittman S, Rodriguez R, et al. . Anti-PD-L1 treatment results in functional remodeling of the macrophage compartment. Cancer Res 2019;79:1493–506. 10.1158/0008-5472.CAN-18-3208
    1. Wallin JJ, Bendell JC, Funke R, et al. . Atezolizumab in combination with bevacizumab enhances antigen-specific T-cell migration in metastatic renal cell carcinoma. Nat Commun 2016;7:12624. 10.1038/ncomms12624
    1. Powles T, Kockx M, Rodriguez-Vida A, et al. . Clinical efficacy and biomarker analysis of neoadjuvant atezolizumab in operable urothelial carcinoma in the ABACUS trial. Nat Med 2019;25:1706–14. 10.1038/s41591-019-0628-7
    1. Limagne E, Richard C, Thibaudin M, et al. . Tim-3/galectin-9 pathway and mMDSC control primary and secondary resistances to PD-1 blockade in lung cancer patients. Oncoimmunology 2019;8:e1564505. 10.1080/2162402X.2018.1564505

Source: PubMed

3
订阅