First-in-human phase 1 study of budigalimab, an anti-PD-1 inhibitor, in patients with non-small cell lung cancer and head and neck squamous cell carcinoma

Antoine Italiano, Philippe A Cassier, Chia-Chi Lin, Tuomo Alanko, Katriina J Peltola, Anas Gazzah, Her-Shyong Shiah, Emiliano Calvo, Andrés Cervantes, Desamparados Roda, Diego Tosi, Bo Gao, Michael Millward, Lydia Warburton, Minna Tanner, Stefan Englert, Stacie Lambert, Apurvasena Parikh, Daniel E Afar, Gregory Vosganian, Victor Moreno, Antoine Italiano, Philippe A Cassier, Chia-Chi Lin, Tuomo Alanko, Katriina J Peltola, Anas Gazzah, Her-Shyong Shiah, Emiliano Calvo, Andrés Cervantes, Desamparados Roda, Diego Tosi, Bo Gao, Michael Millward, Lydia Warburton, Minna Tanner, Stefan Englert, Stacie Lambert, Apurvasena Parikh, Daniel E Afar, Gregory Vosganian, Victor Moreno

Abstract

Background: Budigalimab is a humanized, recombinant immunoglobulin G1 monoclonal antibody targeting programmed cell death protein 1 (PD-1). We present the safety, efficacy, pharmacokinetic (PK), and pharmacodynamic data from patients enrolled in the head and neck squamous cell carcinoma (HNSCC) and non-small cell lung cancer (NSCLC) expansion cohorts of the phase 1 first-in-human study of budigalimab monotherapy (NCT03000257; registered 15 December 2016).

Patients and methods: Patients with recurrent/metastatic HNSCC or locally advanced/metastatic NSCLC naive to PD-1/PD-1-ligand inhibitors were enrolled; patients were not selected on the basis of oncogene driver mutations or PD-L1 status. Budigalimab was administered at 250 mg intravenously Q2W or 500 mg intravenously Q4W until disease progression/unacceptable toxicity. The primary endpoints were safety and PK; the secondary endpoint was efficacy. Exploratory endpoints included biomarker assessments.

Results: In total, 81 patients were enrolled (HNSCC: N = 41 [PD-L1 positive: n = 19]; NSCLC: N = 40 [PD-L1 positive: n = 16]); median treatment duration was 72 days (range, 1-617) and 71 days (range, 1-490) for the HNSCC and NSCLC cohorts, respectively. The most frequent grade ≥ 3 treatment-emergent adverse event was anemia (HNSCC: n = 9, 22%; NSCLC: n = 5, 13%). Both dosing regimens had comparable drug exposure and increased interferon gamma-induced chemokines, monokine induced by gamma interferon, and interferon-gamma-inducible protein 10. Objective response rates were 13% (90% CI, 5.1-24.5) in the HNSCC cohort and 19% (90% CI, 9.2-32.6) in the NSCLC cohort. Median progression-free survival was 3.6 months (95% CI, 1.7-4.7) and 1.9 months (95% CI, 1.7-3.7) in the HNSCC and NSCLC cohorts.

Conclusions: The safety, efficacy and biomarker profiles of budigalimab are similar to other PD-1 inhibitors. Development of budigalimab in combination with novel anticancer agents is ongoing.

Keywords: Budigalimab; Head and neck squamous cell cancer; Non-small cell lung cancer; PD-1 inhibitor.

Conflict of interest statement

Antoine Italiano: Consulting/advisory role: Roche, Daiichi Sankyo, Immune Design, Epizyme, Bayer, Lilly; honoraria: Bayer, Daiichi Sankyo, Lilly, Epizyme, Novartis, Roche; research funding: Roche, Bayer, AstraZeneca/MedImmune, PharmaMar, MSD Oncology, Merck Serono. Philippe A. Cassier: Honoraria: Novartis, Roche/Genentech, Blueprint Medicines, Amgen; research funding: Novartis, Roche/Genentech, Lilly, Blueprint Medicines, Bayer, AstraZeneca, Celgene, Plexxikon, AbbVie, Bristol-Myers Squibb, Merck Serono, Merck Sharp & Dohme; Consultancy/advisory role: Merck Serono, Roche/Genentech. Chia-Chi Lin: Consulting/advisory role: Novartis, Boehringer Ingelheim, Blueprint Medicines; travel/accommodations/expenses: Lilly, Daiichi Sankyo, BeiGene, Novartis; honoraria: Novartis, Roche, Daiichi Sankyo. Tuomo Alanko: Consulting/advisory role: Bayer, Baxalta/Shire, BMS, Celgene, Eli Lilly, MSD, Nordic Drugs, Roche, Kaiku Health; research funding: AbbVie, Bayer, Boehringer Ingelheim, BMS, Debiopharm, Eli Lilly, Incyte, MSD, Pfizer, Roche; travel/accommodations/expenses: Baxalta/Shire, BMS, MSD, Pfizer, Roche. Katriina J. Peltola: Consulting/advisory role: Orion Pharma, BMS, MSD, Novartis, Pfizer, Ipsen, Roche, Varian; stockholder: Faron Pharmaceuticals; speakers’ bureau: BMS, Pfizer, MSD; expert testimony: Ipsen; travel/accommodations/expenses: Roche, BMS; research funding: AbbVie, Bayer, BMS, MSD, Roche, Exelixis, Orion Pharma, Eisai, Novartis. Anas Gazzah: Travel, accommodations, congress registration expenses: Boehringer Ingelheim, Novartis, Pfizer, Roche; consultant/expert role: Novartis; principal/sub-investigator of clinical trials: Aduro Biotech, Agios Pharmaceuticals, Amgen, Argen-X BVBA, Arno Therapeutics, Astex Pharmaceuticals, AstraZeneca, Aveo, Bayer HealthCare Ag, BBB Technologies BV, BeiGene, BioAlliance Pharma, BioNTech AG, Blueprint Medicines, Boehringer Ingelheim, Bristol-Myers Squibb, Ca, Celgene Corporation, Chugai Pharmaceutical Co., Clovis Oncology, Daiichi Sankyo, Debiopharm SA, Eisai, Exelixis, Forma, GamaMabs, Genentech, Inc., Gilead Sciences, Inc, GlaxoSmithKline, Glenmark Pharmaceuticals, H3 Biomedicine, Inc, F. Hoffmann-La Roche AG, Incyte Corporation, Innate Pharma, Servier IRIS, Janssen, Kura Oncology, Kyowa Kirin Pharmaceutical Development, Lilly, Loxo Oncology, Lytix Biopharma AS, MedImmune, Menarini Ricerche, Merck Sharp & Dohme Chibret, Merrimack Pharmaceuticals, Merus, Millennium Pharmaceuticals, Nanobiotix, Nektar Therapeutics, Novartis Pharma, Octimet Oncology NV, OncoEthix, OncoMed, Oncopeptides, Onyx Therapeutics, Orion Pharma, Oryzon Genomics, Pfizer, PharmaMar, Pierre Fabre, Rigontec GmbH, Roche, Sanofi Aventis, Sierra Oncology, Taiho Pharma, Tesaro, Inc, Tioma Therapeutics, Inc., Xencor; research grants: AstraZeneca, BMS, Boehringer Ingelheim, Janssen Cilag, Merck, Novartis, Pfizer, Roche, Sanofi; nonfinancial support (drug supplied): AstraZeneca, Bayer, BMS, Boehringer Ingelheim, Johnson & Johnson, Lilly, MedImmune, Merck, NH TherAGuix, Pfizer, Roche. Her-Shyong Shiah: The author declares no potential conflicts of interest. Emiliano Calvo: Consulting/advisory role: Novartis, Nanobiotix, Janssen-Cilag, PsiOxus, Seattle Genetics, EUSA Pharma, AbbVie, Celgene, AstraZeneca, Guidepoint Global, Roche/Genentech, GLG, Pfizer, Servier, amcure; speakers’ bureau: Novartis; research funding: AstraZeneca, BeiGene, Novartis, START; travel/accommodations/expenses reimbursement: Roche/Genentech; honoraria: HM Hospitales Group; stock/ownership interests: START, Oncoart Associated, International Cancer Consultants; president and founder of Foundation INTHEOS. Andrés Cervantes: Institutional research funding: AbbVie, Genentech, Merck Serono, BMS, MSD, Roche, BeiGene, Bayer, Servier, Lilly, Novartis, Takeda, Astellas, FibroGen; advisory board or speaker fees: Merck Serono, Roche, Bayer, Servier, Pierre Fabre. Desamparados Roda: The author declares no potential conflicts of interest. Diego Tosi: Consulting/advisory role: BioMarin (immediate family member); research funding: Novartis, Astellas, Janssen; patent pending on a new drug combination for prostate cancer treatment; travel/accommodations/expenses: Janssen, Pfizer, Astellas Pharma; immediate family member had travel/accommodations/expenses from Nutricia and Amicus. Bo Gao: Consulting/advisory role: MSD. Michael Millward: Consulting/advisory role: Merck Sharp & Dohme, Bristol-Myers Squibb, AstraZeneca, Roche, Pfizer, Takeda, Novartis; conference travel/support: Merck Sharp & Dohme, Bristol-Myers Squibb, AstraZeneca, Roche. Lydia Warburton: Travel/accommodations/expenses: MSD, Merck. Minna Tanner: Consulting/advisory role: Roche, Novartis, Pfizer; speakers’ bureau: Roche, Novartis, Pfizer, Amgen. Gregory Vosganian: Former employee of AbbVie and may own stock. Stefan Englert, Stacie Lambert, Apurvasena Parikh, Daniel E. Afar: AbbVie employees and may own stock. Victor Moreno: Consulting fees: Merck, BMS, Janssen, Pieris; travel/accommodations: Regeneron/Sanofi; presentations: Nanobiotix; educational grant: Medscape/Bayer.

© 2021. The Author(s).

Figures

Fig. 1
Fig. 1
PD-1 receptor saturation and pharmacodynamic effects of budigalimab administration by dose level. Data shown for each patient with assay baseline and at least 1 postbaseline value. Individual patient data shown for patients with ≥ twofold change in CD8 Ki67 staining. Mean + /– 95% CI shown for PD-1 staining, T-cell counts, and cytokines (number of patients for each mean shown in graph). C, cycle; D, day; HNSCC, head and neck squamous cell carcinoma; hr, hour; IP-10, interferon gamma-induced protein 10; mAb, monoclonal antibody; MIG, monokine induced by gamma interferon; NSCLC, non-small cell lung cancer; PD-1, programmed cell death protein 1; Q, every; TCM, central memory T cells; W, weeks
Fig. 2
Fig. 2
Best percentage change in target lesions from baseline for a HNSCC and b NSCLC cohorts receiving budigalimab monotherapy. aPD-L1 + status; bPD-L1 status missing (i.e., unknown). ALK, anaplastic lymphoma kinase; EGFR, epidermal growth factor receptor; HNSCC, head and neck squamous cell carcinoma; NSCLC, non-small cell lung cancer; PD-L1 + , programmed cell death protein 1 ligand 1 positive; Q, every; W, weeks
Fig. 3
Fig. 3
Percentage change in target lesions from baseline for a HNSCC and b NSCLC cohorts receiving budigalimab monotherapy. ALK, anaplastic lymphoma kinase; EGFR, epidermal growth factor receptor; HNSCC, head and neck squamous cell carcinoma; NSCLC, non-small cell lung V; PD-1, programmed cell death protein 1; Q, every; W, weeks

References

    1. Herzberg B, Campo MJ, Gainor JF. Immune checkpoint inhibitors in non-small cell lung cancer. Oncologist. 2017;22(1):81–88. doi: 10.1634/theoncologist.2016-0189.
    1. Hamanishi J, Mandai M, Matsumura N, Abiko K, Baba T, Konishi I. PD-1/PD-L1 blockade in cancer treatment: perspectives and issues. Int J Clin Oncol. 2016;21(3):462–473. doi: 10.1007/s10147-016-0959-z.
    1. Keir ME, Butte MJ, Freeman GJ, Sharpe AH. PD-1 and its ligands in tolerance and immunity. Annu Rev Immunol. 2008;26:677–704. doi: 10.1146/annurev.immunol.26.021607.090331.
    1. Dong H, Strome SE, Salomao DR, Tamura H, Hirano F, Flies DB, Roche PC, Lu J, Zhu G, Tamada K, Lennon VA, Celis E, Chen L. Tumor-associated B7–H1 promotes T-cell apoptosis: a potential mechanism of immune evasion. Nat Med. 2002;8(8):793–800. doi: 10.1038/nm730.
    1. Mandai M, Hamanishi J, Abiko K, Matsumura N, Baba T, Konishi I. Dual faces of IFNγ in cancer progression: a role of PD-L1 induction in the determination of pro- and antitumor immunity. Clin Cancer Res. 2016;22(10):2329–2334. doi: 10.1158/1078-0432.CCR-16-0224.
    1. Sanmamed MF, Chen L. Inducible expression of B7–H1 (PD-L1) and its selective role in tumor site immune modulation. Cancer J. 2014;20:256–261. doi: 10.1097/PPO.0000000000000061.
    1. KEYTRUDA® (pembrolizumab) injection [prescribing information]. Whitehouse Station, NJ: Merck Sharp & Dohme Corp.; 2020. . Accessed October 2, 2020.
    1. OPDIVO (nivolumab) injection [prescribing information]. Princeton, NJ: Bristol-Myers Squibb Company; 2020. . Accessed October 2, 2020.
    1. LIBTAYO® (cemiplimab-rwlc) injection [prescribing information]. Tarrytown, NY: Regeneron Pharmaceuticals, Inc.; 2020. . Accessed October 2, 2020.
    1. Lambert SL, Vosganian G, Harding F, Sheridan J, Englert S, Siggelkow S, Parikh A, Wang B, Hew K, Bankoti J, Stickler M, Lee E, Mcclellan M, Afar D, Reddy A, Lambert S. Biomarker and preliminary pharmacodynamic evaluations of the PD-1 inhibitor ABBV-181 from an ongoing phase 1 clinical trial in patients with advanced solid tumors. J Immunother Cancer. 2018 doi: 10.1186/s40425-018-0422-y.
    1. European Medicines Agency (2015) Assessment report. OPDIVO. International non-proprietary name: nivolumab. . Accessed March 17, 2021.
    1. European Medicines Agency (2015) Assessment report. Keytruda. International non-proprietary name: pembrolizumab. . Accessed March 17, 2021.
    1. Powderly J, Cassier PA, Cervantes A, Gao B, Gazzah A, Italiano A, Lin C, Luke JJ, Moreno V, Peltola K, Rasco D, Spira AI, Tanner MME, Tosi D, Afar D, Englert S, Parikh A, Reddy A, Vosganian G, Tolcher AW. Safety and efficacy of the PD-1 inhibitor ABBV-181 in patients with advanced solid tumors: preliminary phase 1 results from study M15–891. Ann Oncol. 2018 doi: 10.1093/annonc/mdy279.
    1. Powderly J, Spira A, Kondo S, Doi T, Luke JJ, Rasco D, Gao B, Tanner M, Cassier PA, Gazzah A, Italiano A, Tosi D, Afar DE, Parikh A, Engelhardt B, Englert S, Lambert SL, Kasichayanula S, Mensing S, Menon R, Vosganian G, Tolcher A. Model informed dosing regimen and phase I results of the anti-PD1 antibody budigalimab (ABBV-181) Clin Transl Sci. 2020 doi: 10.1111/cts.12855.
    1. Parikh A, Kasichayanula S, Menon R, Afar D, Lambert S, Engelhardt B, Mensing S. Pharmacokinetics (PK) and dosing regimen selection of the PD-1 inhibitor ABBV-181 in patients with advanced solid tumors: preliminary phase 1 results from study M15–891. J Immunother Cancer. 2018 doi: 10.1186/s40425-018-0423-x.
    1. Eisenhauer EA, Therasse P, Bogaerts J, Schwartz LH, Sargent D, Ford R, Dancey J, Arbuck S, Gwyther S, Mooney M, Rubinstein L, Shankar L, Dodd L, Kaplan R, Lacombe D, Verweij J. New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1) Eur J Cancer. 2009;45(2):228–247. doi: 10.1016/j.ejca.2008.10.026.
    1. Seymour L, Bogaerts J, Perrone A, Ford R, Schwartz LH, Mandrekar S, Lin NU, Litière S, Dancey J, Chen A, Hodi FS, Therasse P, Hoekstra OS, Shankar LK, Wolchok JD, Ballinger M, Caramella C, de Vries EGE, RECIST working group, iRECIST: guidelines for response criteria for use in trials testing immunotherapeutics. Lancet Oncol. 2017;18(3):e143–e152. doi: 10.1016/S1470-2045(17)30074-8.
    1. Haanen JBAG, Carbonnel F, Robert C, Kerr KM, Peters S, Larkin J, Jordan K. ESMO Guidelines committee (2017) Management of toxicities from immunotherapy: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2017;28:iv119–iv142. doi: 10.1093/annonc/mdx225.
    1. Puzanov I, Diab A, Abdallah K, Bingham CO, Brogdon C, Dadu R, Hamad L, Kim S, Lacouture ME, LeBoeuf NR, Lenihan D, Onofrei C, Shannon V, Sharma R, Silk AW, Skondra D, Suarez-Almazor ME, Wang Y, Wiley K, Kaufman HL, Ernstoff MS. Society for Immunotherapy of Cancer Toxicity Management Working Group Managing toxicities associated with immune checkpoint inhibitors: consensus recommendations from the Society for Immunotherapy of Cancer (SITC) Toxicity Management Working Group. J immunother Cancer. 2017;5(1):95. doi: 10.1186/s40425-017-0300-z.
    1. Thompson JA, Schneider BJ, Brahmer J, Andrews S, Armand P, Bhatia S, Budde LE, Costa L, Davies M, Dunnington D, Ernstoff MS, Frigault M, Hoffner B, Hoimes CJ, Lacouture M, Locke F, Lunning M, Mohindra NA, Naidoo J, Olszanski AJ, Oluwole O, Patel SP, Reddy S, Ryder M, Santomasso B, Shofer S, Sosman JA, Wahidi M, Wang Y, Johnson-Chilla A, Scavone JL. Management of immunotherapy-related toxicities version 1. J Natl Compr Canc Network. 2019;17(3):255–289. doi: 10.6004/jnccn.2019.0013.
    1. Brahmer J, Reckamp KL, Baas P, Crinò L, Eberhardt WEE, Poddubskaya E, Antonia S, Pluzanski A, Vokes EE, Holgado E, Waterhouse D, Ready N, Gainor J, Frontera OA, Havel L, Steins M, Garassino MC, Aerts JG, Domine M, Paz-Ares L, Reck M, Baudelet C, Harbison CT, Lestini B, Spigel DR. Nivolumab versus docetaxel in advanced squamous-cell non-small-cell lung cancer. N Engl J Med. 2015;373(2):123–135. doi: 10.1056/NEJMoa1504627.
    1. Borghaei H, Paz-Ares L, Horn L, Spigel DR, Steins M, Ready NE, Chow LQ, Vokes EE, Felip E, Holgado E, Barlesi F, Kohlhäufl M, Arrieta O, Burgio MA, Fayette J, Lena H, Poddubskaya E, Gerber DE, Gettinger SN, Rudin CM, Rizvi N, Crinò L, Blumenschein GR, Jr, Antonia SJ, Dorange C, Harbison CT, Finckenstein FG, Brahmer JR. Nivolumab versus docetaxel in advanced nonsquamous non-small-cell lung cancer. N Engl J Med. 2015;373(17):1627–1639. doi: 10.1056/NEJMoa1507643.
    1. Herbst RS, Baas P, Kim DW, Felip E, Pérez-Gracia JL, Han JY, Molina J, Kim JH, Arvis CD, Ahn MJ, Majem M, Fidler MJ, de Castro G, Garrido M, Lubiniecki GM, Shentu Y, Im E, Dolled-Filhart M, Garon EB. Pembrolizumab versus docetaxel for previously treated, PD-L1-positive, advanced non-small-cell lung cancer (KEYNOTE-010): a randomised controlled trial. Lancet. 2016;387(10027):1540–1550. doi: 10.1016/S0140-6736(15)01281-7.
    1. Jain P, Jain C, Velcheti V. Role of immune-checkpoint inhibitors in lung cancer. Ther Adv Respir Dis. 2018;12:1753465817750075. doi: 10.1177/1753465817750075.
    1. Khoja L, Day D, Chen TWW, Siu LL, Hansen AR. Tumour- and class-specific patterns of immune-related adverse events of immune checkpoint inhibitors: a systematic review. Ann Oncol. 2017;28(10):2377–2385. doi: 10.1093/annonc/mdx286.
    1. Martin-Romano P, Castanon E, Ammari S, Champiat S, Hollebecque A, Postel-Vinay S, Baldini C, Varga A, Michot JM, Vuagnat P, Marabelle A, Soria JC, Ferté C, Massard C. Evidence of pseudoprogression in patients treated with PD1/PDL1 antibodies across tumor types. Cancer Med. 2020;9(8):2643–2652. doi: 10.1002/cam4.2797.
    1. Chanana R, Noronha V, Joshi A, Patil V, Prabhash K. Evolving role of immunotherapy in head-and-neck cancers: a systemic review. J Head Neck Phys Surg. 2018;6(1):2–11. doi: 10.4103/jhnps.jhnps_10_18.
    1. Tazdait M, Mezquita L, Lahmar J, Ferrara R, Bidault F, Ammari S, Balleyguier C, Planchard D, Gazzah A, Soria JC, Marabelle A, Besse B, Caramella C. Patterns of responses in metastatic NSCLC during PD-1 or PDL-1 inhibitor therapy: comparison of RECIST 1.1, irRECIST and iRECIST criteria. Eur J Cancer. 2018;88:38–47. doi: 10.1016/j.ejca.2017.10.017.
    1. Gettinger SN, Horn L, Gandhi L, Spigel DR, Antonia SJ, Rizvi NA, Powderly JD, Heist RS, Carvajal RD, Jackman DM, Sequist LV, Smith DC, Leming P, Carbone DP, Pinder-Schenck MC, Topalian SL, Hodi FS, Sosman JA, Sznol M, McDermott DF, Pardoll DM, Sankar V, Ahlers CM, Salvati M, Wigginton JM, Hellman MD, Kollia GD, Gupta AK, Brahmer JR. Overall survival and long-term safety of nivolumab (anti-programmed death 1 antibody, BMS-936558, ONO-4538) in patients with previously treated advanced non-small-cell lung cancer. J Clin Oncol. 2015;33(18):2004–2012. doi: 10.1200/JCO.2014.58.3708.
    1. Jia W, Gao Q, Han A, Zhu H, Yu J. The potential mechanism, recognition and clinical significance of tumor pseudoprogression after immunotherapy. Cancer Biol Med. 2019;16(4):655–670. doi: 10.20892/j.issn.2095-3941.2019.0144.
    1. Berghoff AS, Bellosillo B, Caux C, de Langen A, Mazieres J, Normanno N, Preusser M, Provencio M, Rojo F, Wolf J, Zielinski CC. Immune checkpoint inhibitor treatment in patients with oncogene-addicted non-small cell lung cancer (NSCLC): summary of a multidisciplinary round-table discussion. ESMO Open. 2019;4(3):e000498. doi: 10.1136/esmoopen-2019-000498.
    1. Kamphorst AO, Pillai RN, Yang S, Nasti TH, Akondy RS, Wieland A, Sica GL, Yu K, Koenig L, Patel NT, Behera M, Wu H, McCausland M, Chen Z, Zhang C, Khuri FR, Owonikoko TK, Ahmed R, Ramalingam SS. 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(19):4993–4998. doi: 10.1073/pnas.1705327114.
    1. Huang AC, Postow MA, Orlowski RJ, Mick R, Bengsch B, Manne S, Xu W, Harmon S, Giles JR, Wenz B, Adamow M, Kuk D, Panageas KS, Carrera C, Wong P, Quagliarello F, Wubbenhorst B, D’Andrea K, Pauken KE, Herati RS, Staupe RP, Schenkel JM, McGettigan S, Kothari S, George SM, Vonderheide RH, Amaravadi RK, Karakousis GC, Schuchter LM, Xu X, Nathanson KL, Wolchok JD, Gangadhar TC, Wherry EJ. T-cell invigoration to tumour burden ratio associated with anti-PD-1 response. Nature. 2017;545(7652):60–65. doi: 10.1038/nature22079.
    1. Das R, Verma R, Sznol M, Boddupalli CS, Gettinger SN, Kluger H, Callahan M, Wolchok JD, Halaban R, Dhodapkar MV, Dhodapkar KM. Combination therapy with anti-CTLA-4 and anti-PD-1 leads to distinct immunologic changes in vivo. J Immunol. 2015;194(3):950–959. doi: 10.4049/jimmunol.1401686.
    1. Choueiri TK, Fishman MN, Escudier B, McDermott DF, Drake CG, Kluger H, Stadler WM, Perez-Gracia JL, McNeel DG, Curti B, Harrison MR, Plimack ER, Appleman L, Fong L, Albiges L, Cohen L, Young TC, Chasalow SD, Ross-Macdonald P, Srivastava S, Jure-Kunkel M, Kurland JF, Simon JS, Sznol M. Immunomodulatory activity of nivolumab in metastatic renal cell carcinoma. Clin Cancer Res. 2016;22(22):5461–5471. doi: 10.1158/1078-0432.CCR-15-2839.
    1. Brahmer JR, Drake CG, Wollner I, Powderly JD, Picus J, Sharfman WH, Stankevich E, Pons A, Salay TM, McMiller TL, Gilson MM, Wang C, Selby M, Taube JM, Anders R, Chen L, Korman AJ, Pardoll DM, Lowy I, Topalian SL. Phase I study of single-agent anti-programmed death-1 (MDX-1106) in refractory solid tumors: safety, clinical activity, pharmacodynamics, and immunologic correlates. J Clin Oncol. 2010;28:3167–3175. doi: 10.1200/JCO.2009.26.7609.

Source: PubMed

3
Subscribe