A first-in-human phase 1 dose escalation study of spartalizumab (PDR001), an anti-PD-1 antibody, in patients with advanced solid tumors

Aung Naing, Justin F Gainor, Hans Gelderblom, Patrick M Forde, Marcus O Butler, Chia-Chi Lin, Sunil Sharma, Maria Ochoa de Olza, Andrea Varga, Matthew Taylor, Jan H M Schellens, Hongqian Wu, Haiying Sun, Antonio P Silva, Jason Faris, Jennifer Mataraza, Scott Cameron, Todd M Bauer, Aung Naing, Justin F Gainor, Hans Gelderblom, Patrick M Forde, Marcus O Butler, Chia-Chi Lin, Sunil Sharma, Maria Ochoa de Olza, Andrea Varga, Matthew Taylor, Jan H M Schellens, Hongqian Wu, Haiying Sun, Antonio P Silva, Jason Faris, Jennifer Mataraza, Scott Cameron, Todd M Bauer

Abstract

Background: Spartalizumab is a humanized IgG4κ monoclonal antibody that binds programmed death-1 (PD-1) and blocks its interaction with PD-L1 and PD-L2. This phase 1/2 study was designed to assess the safety, pharmacokinetics, and preliminary efficacy of spartalizumab in patients with advanced or metastatic solid tumors.

Methods: In the phase 1 part of the study, 58 patients received spartalizumab, intravenously, at doses of 1, 3, or 10 mg/kg, administered every 2 weeks (Q2W), or 3 or 5 mg/kg every 4 weeks (Q4W).

Results: Patients had a wide range of tumor types, most commonly sarcoma (28%) and metastatic renal cell carcinoma (10%); other tumor types were reported in ≤3 patients each. Most patients (93%) had received prior antineoplastic therapy (median three prior lines) and two-thirds of the population had tumor biopsies negative for PD-L1 expression at baseline. The maximum tolerated dose was not reached. The recommended phase 2 doses were selected as 400 mg Q4W or 300 mg Q3W. No dose-limiting toxicities were observed, and adverse events included those typical of other PD-1 antibodies. The most common treatment-related adverse events of any grade were fatigue (22%), diarrhea (17%), pruritus (14%), hypothyroidism (10%), and nausea (10%). Partial responses occurred in two patients (response rate 3.4%); one with atypical carcinoid tumor of the lung and one with anal cancer. Paired tumor biopsies from patients taken at baseline and on treatment suggested an on-treatment increase in CD8+ lymphocyte infiltration in patients with clinical benefit.

Conclusions: Spartalizumab was well tolerated at all doses tested in patients with previously treated advanced solid tumors. On-treatment immune activation was seen in tumor biopsies; however, limited clinical activity was reported in this heavily pretreated, heterogeneous population. The phase 2 part of this study is ongoing in select tumor types.

Trial registration number: NCT02404441.

Keywords: clinical trials as topic; immunotherapy; programmed cell death 1 receptor.

Conflict of interest statement

Competing interests: AN received research funding from NCI, EMD Serono, MedImmune, Healios Oncology Nutrition, Atterocor, Amplimmune, ARMO BioSciences, Eli Lilly, Karyopharm Therapeutics, Incyte, Regeneron, Merck, BMS, Pfizer, Neon Therapeutics, Calithera Biosciences, TopAlliance Biosciences, Kymab, PsiOxus, and Immune Deficiency Foundation (spouse); travel/accommodation expense from ARMO BioSciences; and consultancy fees and research funding from CytomX Therapeutics and Novartis. JG received grant funding from Array, Tesaro, Moderna, Adaptimmune, and Alexo; personal fees from Oncorus, Regeneron, Pfizer, Incyte, Agios, Amgen, and Ironwood Pharmaceuticals; and grant funding and personal fees from Bristol-Myers Squibb, Genentech/Roche, Takeda, Blueprint, Loxo, Novartis, and Merck. PMF received research funding from Corvus and Kyowa; consultancy fees from AbbVie, Boehringer, EMD Serono, Inivata, Janssen, Lilly, and Merck; and research funding and consultancy fees from AstraZeneca, BMS, and Novartis. MOB received funding from Novartis, personal fees from BMS, Adaptimmune, GSK, Novartis, EMD Serono, Sanofi, Immunocore, and Turnstone; grant funding from Takara Bio; and personal fees and grant funding from Merck. C-CL received personal fees from BeiGene, Daiichi Sankyo, Roche, and Novartis. SS received research funding from GSK, Millennium, MedImmune, Johnson & Johnson, Gilead, Plexxikon, Onyx, Bayer, Blueprint, XuanZhu, Incyte, Toray, Celgene, Hengrui, OncoMed, Tesaro, AADi, Merck, Inhibrx, AMAL, and Syndax; personal fees and research funding from Novartis; equity from Iterion Therapeutics, Proterus Therapeutics, ConverGene, and Stingray Therapeutics; honoraria from Exelixis, Loxo Oncology, Natera, Hengrui Therapeutics, Tarveda Therapeutics, Dracen Pharmaceuticals, and Barricade Therapeutics; and owns stock with LSK BioPharma and Salarius Pharmaceuticals. MT received consultancy fees from Eisai, Bristol-Myers Squibb, Array Biopharma, Blueprint Medicines, Arqule, Loxo Oncology, Bayer, Novartis, and Genentech. JHMS received personal fees from Modra Pharmaceuticals. TMB received grant funding from Daiichi Sankyo, Medpacto, Incyte, Mirati Therapeutics, MedImmune, AbbVie, AstraZeneca, MabVax, Stemline Therapeutics, Merck, Lilly, GlaxoSmithKline, Novartis, Genentech, Deciphera, Merrimack, Immunogen, Millennium, Phosplatin Therapeutics, Calithera Biosciences, Kolltan Pharmaceuticals, Principa Biopharma, Peleton, Immunocore, Roche, Aileron Therapeutics, Bristol-Myers Squibb, Amgen, Onyx, Sanofi, Boehringer-Ingelheim, Astellas Pharma, Five Prime Therapeutics, Jacobio, Top Alliance BioScience, Janssen, Clovis Oncology, Takeda, Karyopharm Therapeutics, Foundation Medicine, and ARMO Biosciences; grant funding and consultancy fees from Leap Therapeutics; grant funding, consultancy fees, and non-financial support from Ignyta and Moderna Therapeutics; grant funding, personal fees, and consultancy fees from Pfizer; grant funding, personal fees, and non-financial support from Loxo and Bayer; personal fees and non-financial support from Guardant Health; and personal fees from Exelesis. HW, HS, and JM are employees of Novartis. APS and JF are employees of Novartis and own stock with Novartis. SC is an employee of Novartis and has patents with Novartis.

© Author(s) (or their employer(s)) 2020. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.

Figures

Figure 1
Figure 1
Median concentration–time profiles for spartalizumab, by treatment group. (A) Cycle 1. (B) Cycle 3. Q2W, once every 2 weeks; Q4W, once every 4 weeks.
Figure 2
Figure 2
Percentage change from baseline in target lesions and duration of exposure to spartalizumab. (A) Best percentage change from baseline in target lesions, by PD-L1 expression at baseline. Best overall response is shown for each patient according to RECIST v1.1. (B) Duration of exposure to spartalizumab, by treatment group. Best overall response and response at last assessment are shown. (C) Percentage change from baseline in target lesions over time, by PD-L1 expression at baseline. Best overall response is shown for each patient according to RECIST v1.1. * Unavailable; # Non-measurable disease. ATC, anaplastic thyroid cancer; BOR, best overall response; EMC, extraskeletal myxoid chondrosarcoma; H&N, head and neck; HCC, hepatocellular carcinoma; mRCC, metastatic renal cell carcinoma; non-CR/non-PD, non-complete response/non-progressive disease; NSCLC, non-small cell lung cancer; PD, progressive disease; PD-L1, programmed death ligand 1; PR, partial response; Q2W, once every 2 weeks; Q4W, once every 4 weeks; RECIST, Response Evaluation Criteria In Solid Tumors; SCC, squamous cell carcinoma; SCLC, small cell lung cancer; SD, stable disease; TNBC, triple-negative breast cancer; UNK, unknown.
Figure 3
Figure 3
Tumor response assessments and immunohistochemistry of CD8+ lymphocyte infiltration in patient with atypical carcinoid tumor of the lung with a partial response to treatment. (A) Reduction in overall tumor burden (upper) and individual lesions (lower). (B) CT scans of the liver: (i) liver metastasis pretreatment; (ii) pseudo-progression at first restaging; (iii) response at second restaging. (c) High levels of CD8 detected by immunohistochemistry during Cycle 2. PD, progressive disease; PR, partial response; Q2W, every 2 weeks; Q4W, every 4 weeks; SD, stable disease.

References

    1. Keir ME, Butte MJ, Freeman GJ, et al. . PD-1 and its ligands in tolerance and immunity. Annu Rev Immunol 2008;26:677–704. 10.1146/annurev.immunol.26.021607.090331
    1. Pardoll DM. The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer 2012;12:252–64. 10.1038/nrc3239
    1. Freeman GJ, Long AJ, Iwai Y, et al. . Engagement of the PD-1 immunoinhibitory receptor by a novel B7 family member leads to negative regulation of lymphocyte activation. J Exp Med 2000;192:1027–34. 10.1084/jem.192.7.1027
    1. Latchman Y, Wood CR, Chernova T, et al. . PD-L2 is a second ligand for PD-1 and inhibits T cell activation. Nat Immunol 2001;2:261–8. 10.1038/85330
    1. Ahmadzadeh M, Johnson LA, Heemskerk B, et al. . Tumor antigen-specific CD8 T cells infiltrating the tumor express high levels of PD-1 and are functionally impaired. Blood 2009;114:1537–44. 10.1182/blood-2008-12-195792
    1. Dong H, Strome SE, Salomao DR, et al. . Tumor-associated B7-H1 promotes T-cell apoptosis: a potential mechanism of immune evasion. Nat Med 2002;8:793–800. 10.1038/nm730
    1. Wong RM, Scotland RR, Lau RL, et al. . Programmed death-1 blockade enhances expansion and functional capacity of human melanoma antigen-specific CTLs. Int Immunol 2007;19:1223–34. 10.1093/intimm/dxm091
    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. 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. Nishino M, Giobbie-Hurder A, Gargano M, et al. . Developing a common language for tumor response to immunotherapy: immune-related response criteria using unidimensional measurements. Clin Cancer Res 2013;19:3936–43. 10.1158/1078-0432.CCR-13-0895
    1. Sun H, Sy S, Xu J, et al. . The recommended phase 2 dose selection for spartalizumab (PDR001), an anti-PD-1 antibody, in patients with advanced solid tumors. American Society of Clinical Pharmacology and Therapeutics, Annual Meeting, 2019: PII-109.
    1. Khoja L, Butler MO, Kang SP, et al. . Pembrolizumab. J Immunother Cancer 2015;3:36 10.1186/s40425-015-0078-9
    1. Brahmer JR, Hammers H, Lipson EJ. Nivolumab: targeting PD-1 to bolster antitumor immunity. Future Oncol 2015;11:1307–26. 10.2217/fon.15.52
    1. Patnaik A, Kang SP, Rasco D, et al. . Phase I study of pembrolizumab (MK-3475; anti-PD-1 monoclonal antibody) in patients with advanced solid tumors. Clin Cancer Res 2015;21:4286–93. 10.1158/1078-0432.CCR-14-2607
    1. Naing A, Gelderblom H, Gainor JF, et al. . A first-in-human phase I study of the anti-PD-1 antibody PDR001 in patients with advanced solid tumors. JCO 2016;34:3060 10.1200/JCO.2016.34.15_suppl.3060
    1. Garon EB, Rizvi NA, Hui R, et al. . Pembrolizumab for the treatment of non-small-cell lung cancer. N Engl J Med 2015;372:2018–28. 10.1056/NEJMoa1501824
    1. Weber JS, D'Angelo SP, Minor D, et al. . Nivolumab versus chemotherapy in patients with advanced melanoma who progressed after anti-CTLA-4 treatment (CheckMate 037): a randomised, controlled, open-label, phase 3 trial. Lancet Oncol 2015;16:375–84. 10.1016/S1470-2045(15)70076-8
    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. Khunger M, Hernandez AV, Pasupuleti V, et al. . Programmed cell death 1 (PD-1) ligand (PD-L1) expression in solid tumors as a predictive biomarker of benefit from PD-1/PD-L1 axis inhibitors: a systematic review and meta-analysis. JCO Precis Oncol 2017:1–15. 10.1200/PO.16.00030
    1. Lin C-C, Taylor M, Boni V, et al. . Phase I/II study of spartalizumab (PDR001), an anti-PD1 mAb, in patients with advanced melanoma or non-small cell lung cancer. Ann Oncol 2018;29:viii413 10.1093/annonc/mdy288.032
    1. Wirth LJ, Eigendorff E, Capdevila J, et al. . Phase I/II study of spartalizumab (PDR001), an anti-PD1 mAb, in patients with anaplastic thyroid cancer. J Clin Oncol 2018;36:6024 10.1200/JCO.2018.36.15_suppl.6024
    1. Dummer R, Schadendorf D, Nathan P, et al. . The anti–PD-1 antibody spartalizumab (PDR001) in combination with dabrafenib and trametinib in previously untreated patients with advanced BRAF V600-mutant melanoma: first efficacy, safety, and biomarker findings from the part 2 biomarker cohort of COMBI-i. Cancer Res 2018;78:CT182.

Source: PubMed

3
Prenumerera