8
Journal of Chromatography B, 1023-1024 (2016) 9–16 Contents lists available at ScienceDirect Journal of Chromatography B jou rn al hom ep age: www.elsevier.com/locate/chromb Validated LC–MS/MS analysis of immune checkpoint inhibitor Nivolumab in human plasma using a Fab peptide-selective quantitation method: nano-surface and molecular-orientation limited (nSMOL) proteolysis Noriko Iwamoto a,1 , Takashi Shimada a,1 , Hiroyuki Terakado b , Akinobu Hamada c,a Life Science Research Center, SHIMADZU Corporation, Japan b Department of Pharmacy, National Cancer Center Hospital, Japan c Division of Clinical Pharmacology & Translational Research, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Japan a r t i c l e i n f o Article history: Received 16 January 2016 Received in revised form 1 April 2016 Accepted 22 April 2016 Available online 27 April 2016 Keywords: Nivolumab Immune checkpoint inhibitor Monoclonal antibody nSMOL Bioanalysis LCMS a b s t r a c t We previously reported the nano-surface and molecular-orientation limited (nSMOL) proteolysis, which is a novel method for selective quantitation of monoclonal antibody Fab. The nSMOL strategy is a Fab-selective limited proteolysis which utilizes the size difference between the protease nanoparticle (200 nm) and the antibody resin pore (100 nm). Here, we applied this method to a fully validated LCMS analysis of Nivolumab in human plasma. The immunoglobulin fraction was collected using Protein A resin, which was then followed by nSMOL reaction using the FG nanoparticle surface-immobilized trypsin under a nondenaturing physiological condition at 50 C for 7 h. After removal of resin and nanoparticles by filter centrifugation, signature peptides were separated using the ODS column liquid chromatogra- phy. The signature peptide ASGITFSNSGMHWVR from Nivolumab complementarity-determining region (CDR) and the P 14 R internal standard were simultaneously quantified by multiple-reaction monitoring (MRM) LCMS, with parent m/z 550.8 > fragment m/z 661.5 (y11 2+). The lower limit of quantification (LLOQ) of Nivolumab using the nSMOL method was 0.977 g/ml, with a linear dynamic range of from 0.977 to 250 g/ml. The intra- and inter-assay precision of LLOQ, low quality control (LQC), middle qual- ity control (MQC), and high quality control (HQC) were 7.56–17.9% and 15.6%, 6.99–9.25% and 7.51%, 2.51–8.85% and 8.01%, and 4.78–7.33% and 6.75%, respectively. Our study demonstrates that the nSMOL bioanalysis can be utilized as a reliable method for clinical pharmacokinetic studies of Nivolumab and other antibody drugs. © 2016 The Authors. Published by Elsevier B.V. This is an open access article under the CC BY license (http://creativecommons.org/licenses/by/4.0/). 1. Introduction Programmed cell death-1 (PD-1, CD279), originally identified by Honjo et al. [1], is a type-1 transmembrane immunoreceptor related to the CD28/cytotoxic T-lymphocyte-associated antigen 4 (CTCL-4) family, which negatively regulates the immune response pathways in T-cells [2,3]. This immunological significance led to its numerous clinical benefit such as for autoimmunity, infec- tious elements, and cancer immunotherapies [4]. It is conceivable Abbreviation: nSMOL, nano-surface and molecular-orientation limited proteol- ysis. Corresponding author at: Division of Clinical Pharmacology & Translational Research, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan. E-mail address: [email protected] (A. Hamada). 1 These authors contributed equally to this work. that there are three stages of immune editing systems in can- cer cells: elimination, equilibrium, and escape [5]. Therefore, blockade of the inhibitory antitumor T-cell response called the immune checkpoint has been a major therapeutic target for many cancer eradication. A highly specific monoclonal antibody Nivolumab is a fully human IgG4 monoclonal antibody targeting PD-1 and it is approved by the Food and Drug Administration in 2011 as a new drug for melanoma. T-cells assume definitive roles in immune cell-mediated cancer immunity via a complex cell-to-cell signaling. Long-term cancer controls is one clinical outcome achieved by immune checkpoint inhibitors, by regu- lating the accelerated and/or suppressive immune balance. This mechanism of immunotherapy has also received great atten- tion for the treatment of non-small-cell lung [6–8], renal cell carcinomas [9], ovary [10], bladder [11], and prostate cancers [12], glioblastoma [13], lymphoma [14], and many other tumour types. http://dx.doi.org/10.1016/j.jchromb.2016.04.038 1570-0232/© 2016 The Authors. Published by Elsevier B.V. This is an open access article under the CC BY license (http://creativecommons.org/licenses/by/4.0/).

Journal of Chromatography B - CORE · 2017. 2. 10. · 10 N. Iwamoto et al. / J. Chromatogr. B 1023-1024 (2016) 9–16 Fig. 1. Multiple sequence alignment of Nivolumab amino acid

  • Upload
    others

  • View
    0

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Journal of Chromatography B - CORE · 2017. 2. 10. · 10 N. Iwamoto et al. / J. Chromatogr. B 1023-1024 (2016) 9–16 Fig. 1. Multiple sequence alignment of Nivolumab amino acid

VNq(

Na

b

c

a

ARRAA

KNIMnBL

1

br(pit

y

RC

h1

Journal of Chromatography B, 1023-1024 (2016) 9–16

Contents lists available at ScienceDirect

Journal of Chromatography B

jou rn al hom ep age: www.elsev ier .com/ locate /chromb

alidated LC–MS/MS analysis of immune checkpoint inhibitorivolumab in human plasma using a Fab peptide-selectiveuantitation method: nano-surface and molecular-orientation limitednSMOL) proteolysis

oriko Iwamotoa,1, Takashi Shimadaa,1, Hiroyuki Terakadob, Akinobu Hamadac,∗

Life Science Research Center, SHIMADZU Corporation, JapanDepartment of Pharmacy, National Cancer Center Hospital, JapanDivision of Clinical Pharmacology & Translational Research, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Japan

r t i c l e i n f o

rticle history:eceived 16 January 2016eceived in revised form 1 April 2016ccepted 22 April 2016vailable online 27 April 2016

eywords:ivolumab

mmune checkpoint inhibitoronoclonal antibody

SMOLioanalysisCMS

a b s t r a c t

We previously reported the nano-surface and molecular-orientation limited (nSMOL) proteolysis, whichis a novel method for selective quantitation of monoclonal antibody Fab. The nSMOL strategy is aFab-selective limited proteolysis which utilizes the size difference between the protease nanoparticle(200 nm) and the antibody resin pore (100 nm). Here, we applied this method to a fully validated LCMSanalysis of Nivolumab in human plasma. The immunoglobulin fraction was collected using Protein Aresin, which was then followed by nSMOL reaction using the FG nanoparticle surface-immobilized trypsinunder a nondenaturing physiological condition at 50 ◦C for 7 h. After removal of resin and nanoparticlesby filter centrifugation, signature peptides were separated using the ODS column liquid chromatogra-phy. The signature peptide ASGITFSNSGMHWVR from Nivolumab complementarity-determining region(CDR) and the P14R internal standard were simultaneously quantified by multiple-reaction monitoring(MRM) LCMS, with parent m/z 550.8 > fragment m/z 661.5 (y11 2+). The lower limit of quantification(LLOQ) of Nivolumab using the nSMOL method was 0.977 �g/ml, with a linear dynamic range of from

0.977 to 250 �g/ml. The intra- and inter-assay precision of LLOQ, low quality control (LQC), middle qual-ity control (MQC), and high quality control (HQC) were 7.56–17.9% and 15.6%, 6.99–9.25% and 7.51%,2.51–8.85% and 8.01%, and 4.78–7.33% and 6.75%, respectively. Our study demonstrates that the nSMOLbioanalysis can be utilized as a reliable method for clinical pharmacokinetic studies of Nivolumab andother antibody drugs.

ublis

© 2016 The Authors. P

. Introduction

Programmed cell death-1 (PD-1, CD279), originally identifiedy Honjo et al. [1], is a type-1 transmembrane immunoreceptorelated to the CD28/cytotoxic T-lymphocyte-associated antigen 4CTCL-4) family, which negatively regulates the immune response

athways in T-cells [2,3]. This immunological significance led to

ts numerous clinical benefit such as for autoimmunity, infec-ious elements, and cancer immunotherapies [4]. It is conceivable

Abbreviation: nSMOL, nano-surface and molecular-orientation limited proteol-sis.∗ Corresponding author at: Division of Clinical Pharmacology & Translationalesearch, Exploratory Oncology Research & Clinical Trial Center, National Cancerenter 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan.

E-mail address: [email protected] (A. Hamada).1 These authors contributed equally to this work.

ttp://dx.doi.org/10.1016/j.jchromb.2016.04.038570-0232/© 2016 The Authors. Published by Elsevier B.V. This is an open access article u

hed by Elsevier B.V. This is an open access article under the CC BY license(http://creativecommons.org/licenses/by/4.0/).

that there are three stages of immune editing systems in can-cer cells: elimination, equilibrium, and escape [5]. Therefore,blockade of the inhibitory antitumor T-cell response called theimmune checkpoint has been a major therapeutic target formany cancer eradication. A highly specific monoclonal antibodyNivolumab is a fully human IgG4 monoclonal antibody targetingPD-1 and it is approved by the Food and Drug Administrationin 2011 as a new drug for melanoma. T-cells assume definitiveroles in immune cell-mediated cancer immunity via a complexcell-to-cell signaling. Long-term cancer controls is one clinicaloutcome achieved by immune checkpoint inhibitors, by regu-lating the accelerated and/or suppressive immune balance. Thismechanism of immunotherapy has also received great atten-

tion for the treatment of non-small-cell lung [6–8], renal cellcarcinomas [9], ovary [10], bladder [11], and prostate cancers[12], glioblastoma [13], lymphoma [14], and many other tumourtypes.

nder the CC BY license (http://creativecommons.org/licenses/by/4.0/).

Page 2: Journal of Chromatography B - CORE · 2017. 2. 10. · 10 N. Iwamoto et al. / J. Chromatogr. B 1023-1024 (2016) 9–16 Fig. 1. Multiple sequence alignment of Nivolumab amino acid

10 N. Iwamoto et al. / J. Chromatogr. B 1023-1024 (2016) 9–16

Fig. 1. Multiple sequence alignment of Nivolumab amino acid sequence by ClustalW analysis.A , Ipilima imilarp er is r

aaitcoeicaho

ti

mino acid sequences of four monoclonal antibodies of Nivolumab, Tremelimumabrea shows the matched sequence as common frameworks, and gray is to highlight sosition. (For interpretation of the references to color in this figure legend, the read

However, Nivolumab and other immune checkpoint agentsre known to cause several immune-related adverse events suchs dermatological, gastrointestinal, endocrine, hepatic, neurolog-cal, and other effects on other organs [15–17]. It is challengingo address the precision immunotherapy for cancers due to theomplexity of diverse immune interactions. Thus, identificationf predictive and pharmacodynamic biomarkers or marker pan-ls for drug efficacy will be crucial for a more effective cancermmunotherapy [18]. We expect that an overall clinical pharma-okinetics (PK) can be used as a good indicator for drug efficacynd for predicting the effects on the immune response. However,ow PK findings of these inhibitors may translate to their clinicalutcomes is yet to be determined.

In the field of clinical PK, ligand binding assay (LBA) is currentlyhe first choice for quantitating therapeutic antibodies. However,n some cases, LBA is not a reasonable method for antibody

umab, and Ramucirumab were aligned in (a) heavy chain and (b) light chain. Black amino acids. The red underline is shown to highlight the selected signature peptideeferred to the web version of this article.)

quantitation. For example, neutralizing antibodies called anti-drug antibodies (ADA) inhibit specific binding of antigen to drugs,contributing to unexpected errors [19]. Furthermore, species-specificity can cause limitation in translating findings obtainedusing preclinical animal models into clinical studies in human.Currently, cancer care utilizes combined chemotherapy strategiesusing next generation molecular target compounds, antibodies, andantibody-drug conjugates (ADC). Therefore, a more selective, sensi-tive, and universal technology independent of a variety of antibodyagents will be desirable. Nivolumab is a fully human antibody; thus,an accurate quantitation of signature peptides with a completestructural determination is especially important since the specificregions of antibody drugs are shorter than those of the endogenous

immunoglobulins with high sequence homology. Recently, liquidchromatography-triple quadrupole mass spectrometry (LC-TQMS)was applied to quantitate therapeutic antibodies in biological fluid.
Page 3: Journal of Chromatography B - CORE · 2017. 2. 10. · 10 N. Iwamoto et al. / J. Chromatogr. B 1023-1024 (2016) 9–16 Fig. 1. Multiple sequence alignment of Nivolumab amino acid

atogr.

Lads[

nsatEiriftowrtAairptmnctao

2

2

bT6wuf(DsMOW

2

oifa2A1tstv

N. Iwamoto et al. / J. Chrom

CMS is a more selective and sensitive method to quantitate thentibody specific regions. Further, it allows for multiplex assayevelopment in a more shorter period of time, without the has-le of specific collection and detection antibodies in LBA method20,21].

However, to develop LCMS at the level of regulated bioanalysis,ecessary improvements will need to be done for greater preci-ion and accuracy, dynamic range, and universal applicability forntibody drugs. Plasma is a complex biological mixture, and tryp-ic peptides from this matrix are large excess analytes for LCMS.xcess peptide mixtures and protease contamination can causeonization suppression, carryover, noise elevation, lower dynamicange and retention time reproducibility, and column sustainabil-ty. To overcome these issues, we recently reported a novel methodor selective proteolysis of the Fab by limiting protease access tohe substrate, which we termed the nano-surface and molecular-rientation limited (nSMOL) proteolysis [22]. The nSMOL chemistryas designed as a solid–solid proteolysis as follows: (1) protease

eaction on a uniform nanoparticle surface to raise the accessibilityo the substrate; (2) Fc immobilization onto the resin via Protein, such that the Fab is oriented outward to the reaction solution;nd (3) Fab-selective proteolysis by making use of the differencen the protease nanoparticle diameter (200 nm) and the antibodyesin pore (100 nm). The nSMOL has allowed us to minimize sam-le complexity while maintaining the specificity of peptides inhe complementarity-determining region (CDR). Furthermore, this

ethod has allowed for effective proteolysis on Fab by using aondenaturing physiological condition, absence of extra trypsinontamination in the analyte, absence of further peptide purifica-ion such as solid-phase extraction, and independent of a variety ofntibodies. In this study, we demonstrate a validated LCMS analysisf Nivolumab in plasma using the nSMOL [23,24].

. Materials and methods

.1. Reagents

Glycidyl methacrylate (GMA)-coated nano-ferrite particle FGeads with surface activation by NHS group was purchased fromamagawa Seiki (Nagano, Japan). Toyopearl AF-rProtein A HC-50F resin was from Tosoh (Tokyo, Japan). Nivolumab (OPDIVO)as purchased from Ono Pharmaceutical (Osaka, Japan). Individ-al male and female control human plasma (EDTA-2K treated) wasrom Kohjin Bio (Saitama, Japan). Trypsin gold was from PromegaFitchburg, WI). n-octyl-�-d-thioglucopyranoside (OTG) was fromojindo Laboratories (Kumamoto, Japan). P14R internal standard

ynthetic peptide was from Sigma Aldrich (St. Louis, MO). Ultrafree-C GV centrifugal filter was from Merck Millipore (Billerica, MA).ther reagents, buffers, and solvents were from Sigma-Aldrich andako Pure Chemical Industries (Osaka, Japan).

.2. Peptide structure identification of Nivolumab peptides

Two microlitre of Nivolumab (20 �g) was digested using 1 �gf trypsin in 150 �l of 25 mM tris–HCl buffer (pH 8.0) contain-ng 1 mM tris(2-carboxyethyl)phosphine-HCl salt (TCEP) at 37 ◦Cor 16 h. Trypsin digestion was quenched by adding 10% formiccid solution at a final concentration of 0.5%. For nSMOL reactions,0 �g of Nivolumab was collected with 10 �l of PBS-substitutedF-rProtein A resin (50% slurry) with gentle vortexing at 25 ◦C for0 min. Protein A resin was recovered on an Ultrafree filter, washed

wice using 300 �l of PBS by centrifugation (10,000g for 1 min), andubstituted with 150 �l of 25 mM tris–HCl (pH 8.0). nSMOL reac-ion was carried out using 1 �g of trypsin on FG-beads with gentleortexing at 37 ◦C for 16 h in saturated vapour atmosphere. After

B 1023-1024 (2016) 9–16 11

proteolysis, reaction was stopped by adding 10% formic acid at afinal concentration of 0.5%. The peptide solution was collected bycentrifugation (10,000g for 1 min) to remove Protein A resin andtrypsin FG-beads. Structure of tryptic peptides from Nivolumabwere analysed by high-resolution liquid chromatography-linearion trap time-of-flight MS (Nexera X2 ultra high performance liq-uid chromatograph and LCMS-IT-TOF, Shimadzu, Kyoto, Japan), andfragment ions were assigned using an in-house Mascot server andDistiller with Nivolumab amino acid sequence information (MatrixScience, London, UK). The LCMS conditions were as follows: sol-vent A, 0.1% aqueous formic acid; solvent B, acetonitrile with 0.1%formic acid; column, L-column2 ODS, 2.1 × 150 mm, 2 �m, 10 nmpore (Chemicals Evaluation and Research Institute, Tokyo, Japan);column temperature, 50 ◦C; flow rate, 0.2 ml/min; gradient pro-gram, 0–5 min: %B = 3, 5–35 min: %B = 3–30 gradient, 35–46 min:%B = 95, 46–55 min: %B = 3. MS and MS/MS spectra were obtainedusing desolvation line and heat block at 200 ◦C, respectively. Neb-ulizer nitrogen gas flows were set to 1.5 l/min. Drying gas pressurewas 100 kPa. Ion accumulation time was 30 ms for MS, and 70 msfor MS/MS analysis. MS/MS analysis was performed using the auto-mated data dependent mode. Ar pulse time into the ion trap cellwas 125 �s. The electrode of collision-induced dissociation (CID)cell was set at −1.5 V.

2.3. Prediction of Nivolumab signature peptides

Amino acid sequences of antibody drugs were obtained fromKyoto Encyclopedia of Genes and Genomes (KEGG) DRUG. Multi-ple alignment was performed using the amino acid sequence ofNivolumab (KEGG DRUG entry D10316), Tremelimumab (D06657),Ipilimumab (D04603), and Ramucirumab (D09371) by ClustalWalgorithm on GENETYX software (GENETYX, Tokyo, Japan). In thisanalysis, theoretical tryptic peptides containing the CDR sequence,amino acid substitution, positions of conserved cysteine residues,and insertion or deletion of amino acids were aligned.

2.4. Condition setting of multiple reaction monitoring forNivolumab peptides

The peptide quantitation was analysed using an LC–electrosprayionization-MS (LC–ESI-MS) with triple quadrupole (Nexera X2and LCMS-8050, Shimadzu). The LCMS conditions were as fol-lows: solvent A, 0.1% aqueous formic acid; solvent B, acetonitrilewith 0.1% formic acid; column, Shim-pack GISS C18, 2.1 × 50 mm,1.9 �m, 20 nm pore (Shimadzu); column temperature, 50 ◦C; flowrate, 0.4 ml/min; gradient program, 0–1.5 min: %B = 1, 1.5–4.5 min:%B = 1–40 gradient, 4.5–5.5 min: %B = 95 with flow rate 1 ml/min,5.5–6 min: %B = 1 with flow rate 1 mi/min, and 6–6.5 min: %B = 1.MS spectra were obtained with ESI probe temperature, desolva-tion line, and heat block at 300 ◦C, 250 ◦C and 400 ◦C, respectively.Nebulizer, heating, and drying nitrogen gas flows were set to 3, 10,and 10 l/min, respectively. The dwell time was set to 10 ms for eachtransition. MRM monitor ions of peptide fragments were importedfrom the measured values of structure-assigned fragments by high-resolution LCMS analysis. CID Ar partial pressure in the Q2 cell wasset to 270 kPa. The electrode voltage of Q1 pre bias, collision cell Q2,Q3 pre bias, and parent and fragment ion m/z were optimized usingthe LabSolution software (Shimadzu). For MRM transition, one frag-ment ion of b- or y-series was selected for quantitation, and twoions for structural confirmation according to the optimized MRMion yield.

2.5. Validation sample preparation of Nivolumab analysis

In the current study, we performed a bioanalytical validationof Nivolumab in plasma using the nSMOL method as described

Page 4: Journal of Chromatography B - CORE · 2017. 2. 10. · 10 N. Iwamoto et al. / J. Chromatogr. B 1023-1024 (2016) 9–16 Fig. 1. Multiple sequence alignment of Nivolumab amino acid

1 atogr. B 1023-1024 (2016) 9–16

ia−aiwgoOoTFafwtictplqr

3

3L

uForrsrccmos

3

h4NbiWpatte

3

GcLM

Fig. 2. Verification of dilution interference.Nivolumab tryptic peptides were quantitated by nSMOL in human plasma. Rep-resentative calibration curves of (a) ASGITFSNSGMHWVR, which showed goodcorrelation with Nivolumab concentrations, and (b) LLIYDASNR, with no correla-tion to Nivolumab concentrations demonstrating competition from endogenous

2 N. Iwamoto et al. / J. Chrom

n our previous report with a minor improvement [22]. Briefly,ll validation sample sets were prepared and stored at −20 ◦C or80 ◦C for 24 h or longer before each validation assay. A 20 �lliquot of Nivolumab-spiked human plasma was diluted 10-foldn PBS (pH 7.4) containing 0.1% OTG. The Ig fraction was collected

ith 50 �l of PBS-substituted AF-rProtein A resin (50% slurry) withentle vortexing at 25 ◦C for 15 min. Protein A resin was collectednto Ultrafree filter and washed twice with 300 �l PBS containingTG, centrifuged (10,000g for 1 min), and substituted with 150 �lf 25 mM tris-HCl (pH8.0) containing 10 pmol/ml P14R and 2 mMCEP. nSMOL proteolysis was carried out using 25 �g trypsin onG-beads with gentle vortexing at 50 ◦C for 7 h in saturated vapourtmosphere. After nSMOL, reaction was stopped by adding 10%ormic acid at a final concentration of 0.5%. The peptide solutionas collected by centrifugation (10,000g for 1 min) to remove Pro-

ein A resin and trypsin FG-beads. These analytes were transferrednto low protein binding polypropylene vials for LCMS analysis. Theoncentration of Nivolumab in plasma samples was set from 0.977o 250 �g/ml with two-fold serial dilution for nine calibration sam-les. The concentrations of lower limit of quantification (LLOQ),

ow quality control (LQC), middle quality control (MQC), and highuality control (HQC) were set to 0.977, 2.93, 23.4, and 200 �g/ml,espectively.

. Results

.1. Structural confirmation of Nivolumab peptides byCMS-IT-TOF MS and ClustalW analysis

The identification of tryptic Nivolumab peptides was performedsing a high-resolution LC-TOF MS/MS analysis and Mascot server.ive peptides from the heavy chain and three from the light chainn Fv were assigned with significant Mascot score, and summa-ized in Table 1. Peptide regions were shown as ClustalW alignmentesults of four fully human therapeutic antibodies (Fig. 1). To selectignature peptides, we have established a criteria in our previouseport: peptides with 8–15 amino acid sequences, with no missedleavage, no cysteine residue, and exclusion of the vicinity of theonserved disulphide bonding on the framework structure. Opti-ized MRM transitions of each peptide are shown in Table 2, with

ne fragment for quantitation and an additional two fragments fortructural confirmation.

.2. Nivolumab peptides interference from human plasma

To determine interference of Nivolumab peptides fromuman plasma, each candidate peptide was analysed using.69–37.5 �g/ml of sample with two-fold serial dilution in plasma.ot all CDR peptides can be used as the specific peptide for nSMOLioanalysis, due to the exceedingly diverse set of CDR sequences

n endogenous immunoglobulins. The peptide ASGITFSNSGMH-VR (MW 1649.8) was selected as the signature Nivolumab

eptide as we showed no interference from human plasma and good correlation with Nivolumab concentrations (Fig. 2a). In con-rast, for example of LLIYDASNR peptide, showed no correlationo Nivolumab concentrations demonstrating a competition fromndogenous immunoglobulins (Fig. 2b).

.3. Bioanalytical validation

The bioanalytical method was validated according to the

uideline on Bioanalytical Method Validation in Pharmaceuti-al Development from Notification 0711-1 of the Evaluation andicensing Division, Pharmaceutical and Food Safety Bureau, theinistry of Health, Labour and Welfare, dated July 11, 2013 for low

immunoglobulin are shown.

molecular weight drug compounds. The validation results of thisstudy are summarized in Supporting information.

3.3.1. SelectivityFor the individual human plasma controls from three males

and three females, no interference peak in the MRM chro-matograms were detected for the ASGITFSNSGMHWVR signatureNivolumab peptide. Minimal response was observed from inter-fering substance at the retention time of 3.75 min correspondingto the signature Nivolumab peptide. Representative MRM chro-matograms of the signature Nivolumab peptide are shown inFig. 3.

3.3.2. Matrix effectIndividual plasma samples from three males and three females

were analysed for matrix effect test at the LQC and HQC concentra-tions. The response ratio of Nivolumab peptide in plasma prior toeach sample preparation step was compared to that of PBS. Matrixfactors of P14R-corrected value at LQC and HQC were 9.72% and7.27%, respectively, which were within the accepted criteria of pre-cision (CV) of <15% (Table S1 in Supporting information).

3.3.3. CarryoverThe carryover test was performed by analysing three replicates

for Nivolumab peptide and one run for P14R immediately after thehigh concentration (250 �g/ml) analysis. The carryover was cal-culated as percent response in the blank plasma relative to thepreceding sample. In nSMOL analysis, carryover was observed at0.151–0.172% for the Nivolumab peptide and no carryover wasobserved for P14R, confirming minimal levels of carryover influ-encing our analysis (Table S2 in Supporting information).

3.3.4. Linearity and calibration standardThe linearity of nSMOL protocols was evaluated by analysing

nine calibration standards (zero sample, 0.977, 1.95, 3.91, 7.81,15.6, 31.3, 62.5, 125, and 250 �g/ml) using the linear regressionmodel. The calibration plot of weighting was done using the 1/area2

method. The calibration fit formulas of the triplicate runs wereY = 0.0627X + 0.0211 (r = 0.997), Y = 0.0585X + 0.00507 (r = 0.997),

and Y = 0.0750X + 0.0434202 (r = 0.996) (r: correlation coefficient).The accuracy at LLOQ was 91.9–97.0%, and at other concentrationswere 91.1–111% (Table S3 in Supporting information).
Page 5: Journal of Chromatography B - CORE · 2017. 2. 10. · 10 N. Iwamoto et al. / J. Chromatogr. B 1023-1024 (2016) 9–16 Fig. 1. Multiple sequence alignment of Nivolumab amino acid

N. Iwamoto et al. / J. Chromatogr. B 1023-1024 (2016) 9–16 13

Table 1Identified peptides from Nivolumab using the in-house Mascot analysis.

No. Observed m/z Sequence Region

1–16 796.9217 *QVQLVESGGGVVQPGR (*N-term Q as pyro-Glu) N-term of H-chain24–38 550.5983 ASGITFSNSGMHWVR CDR1 of H-chain44–57 593.6439 GLEWVAVIWYDGSKR CDR2 of H-chain59–65 423.2042 YYADSVK CDR2 of H-chain77–87 668.8559 NTLFLQMNSLR CDR2 of H-chain1–18 633.3453 EIVLTQSPATLSLSPGER N-term of L-chain25–45 784.7257 ASQSVSSYLAWYQQKPGQAPR CDR1 of L-chain46–54 532.7885 LLIYDASNR CDR2 of L-chain

Table 2Optimal MRM transition of signature candidate peptides from Nivolumab by nSMOL analysis.

Selected peptide Optimal MRM condition Role

Transition mass filter [m/z] Q1 [V] Collision [V] Q3 [V]

P14R (internalstandard)

512.1 → 292.3 (b3 +) −38 −20 −20 Quantitation512.1 → 389.3 (b4 +) −38 −16 −28 Structure512.1 → 660.4 (b6 +) −38 −17 −24 Structure

pQVQLVESGGGVVQPGR 797.1 → 329.3 (y3 +) −22 −20 −24 Quantitation797.1 → 457.3 (y4 +) −22 −21 −17 Structure797.1 → 551.3 (y5 +) −36 −26 −40 Structure

ASGITFSNSGMHWVR 550.8 → 661.5 (y11 ++) −24 −18 −24 Quantitation550.8 → 746.4 (y13 ++) −26 −16 −28 Structure550.8 → 785.4 (y6 +) −26 −25 −30 Structure

GLEWVAVIWYDGSKR 593.8 → 805.0 (y13 ++) −28 −17 −30 Quantitation593.8 → 740.4 (y12 ++) −26 −18 −40 Structure593.8 → 597.8 (y10 ++) −28 −17 −22 Structure

YYADSVK 423.3 → 519.4 (y5 +) −20 −16 −26 Quantitation423.3 → 682.4 (y6 +) −20 −18 −26 Structure423.3 → 333.1 (y3 +) −20 −20 −24 Structure

NTLFLQMNSLR 669.0 → 1008.6 (y8 +) −30 −24 −30 Quantitation669.0 → 861.5 (y7 +) −30 −25 −32 Structure669.0 → 748.4 (y6 +) −30 −25 −28 Structure

EIVLTQSPATLSLSPGER 949.7 → 1127.7 (y11 +) −26 −36 −44 Quantitation949.7 → 458.3 (y4 +) −26 −32 −17 Structure949.7 → 545.3 (y5 +) −26 −30 −28 Structure

ASQSVSSYLAWYQQKPGQAPR 785.0 → 940.6 (y16 ++) −36 −25 −36 Quantitation785.0 → 1033.8 (y18 ++) −36 −25 −30 Structure785.0 → 625.3 (y6 +) −36 −36 −32 Structure

3

pNapaaa8t6

3

bdaDNffnca

LLIYDASNR 532.9 → 838.5 (y7 +) −24

532.9 → 725.4 (y6 +) −24

532.9 → 447.3 (y7 +) −24

.3.5. Precision and accuracy in inter- and intra-assaysPrecision and accuracy were determined by analysis of human

lasma validation sample at LLOQ, LQC, MQC, and HQC ofivolumab concentrations. The intra-day and inter-day precisionnd accuracy were determined by analysing five sets of QC sam-les at four concentration levels on three different days. Precisionnd accuracy data were: run 1, 92.7% and 17.9% at LLOQ, 102–112%nd 7.11–9.25% at other concentrations; run 2, 109% and 11.2%t LLOQ, 99.0–103%, 2.51–7.86% at other concentrations; run 3,7.0% and 7.56% at LLOQ, 98.4–101%, 4.78–6.99% at other concen-rations; intra-assay (N = 15), 96.1% and 15.6% at LLOQ, 101–105%,.75–8.01% at other concentrations (Table 3).

.3.6. Lower Limit of Quantification (LLOQ)The LLOQ was defined as the lowest concentration of the cali-

ration curve with acceptable precision and accuracy, which wereetermined to be 0.977 �g/ml (Table S4). According to the pack-ge insert of Nivolumab from the Pharmaceuticals and Medicalevices Agency (PMDA) of Japan, the Cmax concentrations ofivolumab Phase I trial (CA209001) in a single-dose administration

or melanoma cancer patients were 6.7, 16.0, 60.0, and 196.3 �g/ml

or 0.3, 1, 3, and 10 mg/kg dose, respectively, indicating that theSMOL quantitation was sufficient to cover Nivolumab clinical con-entration spectrum in the plasma. Thus, the nSMOL method wasdequate to monitor PK of Nivolumab.

−19 −32 Quantitation−20 −38 Structure−28 −22 Structure

3.3.7. Dilution integrityThe effect of dilution on the analysis of Nivolumab concentration

was assessed by preparing spiked validation human plasma at aconcentration of 500 �g/ml. The 10- and 25-fold dilution of the fivevalidation samples were analysed within the calibration range. Theprecision and accuracy of the diluted samples were 7.92% and 95.4%,9.27% and 109%, for 10- and 25-fold dilutions, respectively. Thesevalues were within the criteria of decision, suggesting an absenceof interference from dilution (Table S4 in Supporting information).

3.3.8. StabilityThe stability following five freeze-thaw cycles at −20 ◦C and

−80 ◦C with at least 12 h of frozen time, short-term stability at roomtemperature for 4 h prior to sample treatment, long-term stabilityat −20 ◦C and −80 ◦C for 30 days, and processed sample stability at5 ◦C for 24 and 48 h were demonstrated at the LQC and HQC con-centrations. The accuracy of the initial value for five freeze-thawcycles at LQC and HQC in −20 ◦C was 95.6% and 96.1%, respectively,and in −80 ◦C was 87.5% and 101%, respectively (Table 4).

4. Discussion

We have developed a practical and versatile analytical methodto assess Nivolumab plasma concentration in patients. As demon-strated by the present validation study, this method was able to

Page 6: Journal of Chromatography B - CORE · 2017. 2. 10. · 10 N. Iwamoto et al. / J. Chromatogr. B 1023-1024 (2016) 9–16 Fig. 1. Multiple sequence alignment of Nivolumab amino acid

14 N. Iwamoto et al. / J. Chromatogr. B 1023-1024 (2016) 9–16

Table 3Precision and accuracy.

Run Nominal concentration Concentration (�g/ml)

0.977 2.93 23.4 200

1 Observed 0.786 2.60 26.9 1930.725 3.32 23.8 2160.918 3.11 29.1 2001.14 2.84 23.8 2080.957 3.05 27.4 232

Mean 0.906 2.98 26.2 210SD 0.162 0.276 2.32 14.9CV (%) 17.9 9.25 8.85 7.11Accuracy (%) 92.7 102 112 105

2 Observed 0.912 3.17 24.2 1891.04 2.75 23.3 1851.08 3.10 23.8 1961.24 3.14 24.0 2221.03 2.68 25.0 197

Mean 1.06 2.97 24.1 198SD 0.119 0.233 0.605 14.5CV (%) 11.2 7.86 2.51 7.33Accuracy (%) 109 101 103 99

3 Observed 0.900 3.21 22.2 2020.877 3.09 23.7 1870.781 2.75 21.5 1910.780 2.75 24.3 2110.910 2.94 25.5 193

Mean 0.850 2.95 23.4 197SD 0.0642 0.206 1.60 9.42CV (%) 7.56 6.99 6.84 4.78Accuracy (%) 87.0 101 100 98.4

Mean (N = 15) 0.939 2.97 24.6 202SD (N = 15) 0.146 0.223 1.97 13.6CV (%) 15.6 7.51 8.01 6.75Accuracy (%) 96.1 101 105 101

Table 4Confirmation of QC sample for stability.

Parameters for stabilitystudies

Concentrations of nivolumab in human plasma (�g/ml)

2.93 200

Mean (�g/ml) Accuracy (%) Mean (�g/ml) Accuracy (%)

Stability in plasma during freeze (−20 ◦C) and thaw cyclesCycle 5 2.73 95.6 183 96.1Stability in plasma during freeze (−80 ◦C) and thaw cyclesCycle 5 2.62 87.5 211 101Short-term stability in plasma for 4 h at room temperature

2.80 95.5 186 92.9Long-term stability in plasma for 30 days at −20 ◦C

3.03 104 213 107Long-term stability in plasma for 30 days at −80 ◦C

3.15 107 187 93.3Processed sample stability in HPLC set at 5 ◦C

97.8105

pActfsitaoa

ic

For 24 h 2.87

For 48 h 3.08

rovide a robust determination of Nivolumab in human plasma.ll concentration points have reproducibly passed the guidelineriteria for low molecular weight drug compounds. Especially men-ioned, nSMOL approach has been even acceptable for the normalreezing storage at −20 ◦C. These advantages will be expected toupport for the broad clinical PK study. We believe that this assays suitable for routine analyses of plasma Nivolumab concentrationo study the correlations between its PK profile and drug efficacynd/or adverse events in clinical practice, thereby aiding the devel-pment of a precision medicine for patients receiving Nivolumab

lone or in combination with other anticancer agents.

The essence of cancer immunotherapy is to control themmune cell activity in a complex immune system by immuneheckpoint blockade. However, immune checkpoint inhibitors

184 92.1 195 97.6

have been linked to side effects. Although various studies havehelped elucidate cancer immunity, a detailed mechanism-basedimmunotherapy which also takes into account the complex tumourmicroenvironment and diverse tumour tissue types will be neces-sary.

Expression of PD-1 ligand (PD-L1, PD-L2) in some cancer cellsor in immune cells within the tumour microenvironment has beenstrongly correlated to negative prognosis. The response rates of 44%for PD-L1-positive tumours and 17% for PD-L1-negative tumourswere reported in a phase I trial of Nivolumab [4,25]. However,

these response rates did not translate to overall survivals [26]. Todecide the optimal efficacy of immune checkpoint inhibitors, med-ical oncologists will require a conventional tool to predict clinicalefficacy and to manage adverse events.
Page 7: Journal of Chromatography B - CORE · 2017. 2. 10. · 10 N. Iwamoto et al. / J. Chromatogr. B 1023-1024 (2016) 9–16 Fig. 1. Multiple sequence alignment of Nivolumab amino acid

N. Iwamoto et al. / J. Chromatogr.

Fig. 3. MRM chromatogram of the signature Nivolumab peptide ASGITFSNSGMH-WVR and P14R in a selectivity test of the LLOQ sample.The signature Nivolumab peptide peak ASGITFSNSGMHWVR (fragment m/s 661.5from parent 550.8, retention time 3.75 min) was observed at the LQC (2.93 �g/ml)sample. The horizontal axis shows the retention time (min) and vertical axis showsthe ion count (cps). The red lines show baseline of the assigned peak. Representativechromatograms from (a) Nivolumab-spiked plasma of a male person, (b) Nivolumab-spiked plasma of a female person, (c) control blank sample, (d) internal peptide P14R(fragment m/z 292.3 from parent 512.1, retention time 3.35 min), and (e) blank sam-ple of P14R were shown. (For interpretation of the references to color in this figurel

moLiPdfsnatbs

yLo

[

[

[

[

[

Ligon, L. Zhu, J.F. Grosso, S.Y. Kim, J.M. Timmerman, M.A. Shipp, P. Armand,PD-1 blockade with Nivolumab in relapsed or refractory Hodgkin’s

egend, the reader is referred to the web version of this article.)

Recently, PK-guided treatment such that as by therapeutic drugonitoring for cancer chemotherapy has gained an increasing level

f interest. This is due to the current lack of available tools besidesBA to determine plasma concentration of antibody drugs in a clin-cal setting. Mass spectrometry has the potential to support theK study of antibody agents because of its superior selectivity forrug structure-indicated quantitation. nSMOL is an novel methodor the quantitation of antibody-specific CDRs from blood and tis-ue homogenate, independent of a variety of antibody agents [22].SMOL analysis will allow for accurate quantitation of antibodygents in the blood with the additional benefits of specific struc-ure confirmation and multiplex assay development. Based on ourioanalytical validation study, we have shown that nSMOL can beufficiently used to determine Nivolumab clinical PK.

In conclusion, we have first demonstrated a fully validated anal-

sis of Nivolumab in human plasma using nSMOL coupled withC–MS/MS. This analytical approach can be applied to other mon-clonal antibodies. Moreover, our study demonstrates that the

[

B 1023-1024 (2016) 9–16 15

nSMOL method can be expected in future clinical pharmacokineticstudies for antibody drug development and therapy.

Acknowledgement

This research was partly supported by “Practical Research forInnovative Cancer Control” funds from the Japan Agency for Medi-cal Research and Development, AMED.

Appendix A. Supplementary data

Supplementary data associated with this article can be found,in the online version, at http://dx.doi.org/10.1016/j.jchromb.2016.04.038.

References

[1] Y. Ishida, Y. Agata, K. Shibahara, T. Honjo, Induced expression of PD-1 a novelmember of the immunoglobulin gene superfamily, upon programmed celldeath, EMBO J. 11 (1992) 3887–3895.

[2] H. Nishimura, T. Okazaki, Y. Tanaka, K. Nakatani, M. Hara, A. Matsumori, S.Sasayama, A. Mizoguchi, H. Hiai, N. Minato, T. Honjo, Autoimmune dilatedcardiomyopathy in PD-1 receptor-deficient mice, Science 291 (2001)319–322.

[3] T. Okazaki, T. Honjo, PD-1 and PD-1 ligands: from discovery to clinicalapplication, Int. Immunol. 19 (2007) 813–824.

[4] P. Sharma, J.P. Allison, The future of immune checkpoint therapy, Science 348(2015) 56–61.

[5] R.D. Schreiber, L.J. Old, M.J. Smyth, Cancer immunoediting: integratingimmunity’s roles in cancer suppression and promotion, Science 331 (2011)1565–1570.

[6] S.N. Gettinger, L. Horn, L. Gandhi, D.R. Spigel, S.J. Antonia, N.A. Rizvi, J.D.Powderly, R.S. Heist, R.D. Carvajal, D.M. Jackman, L.V. Sequist, D.C. Smith, P.Leming, D.P. Carbone, M.C. Pinder-Schenck, S.L. Topalian, F.S. Hodi, J.A.Sosman, M. Sznol, D.F. McDermott, D.M. Pardoll, V. Sankar, C.M. Ahlers, M.Salvati, J.M. Wigginton, M.D. Hellmann, G.D. Kollia, A.K. Gupta, J.R. Brahmer,Overall survival and long-term safety of Nivolumab (anti-programmed death1 antibody, BMS-936558 ONO-4538) in patients with previously treatedadvanced non-small-cell lung cancer, J. Clin. Oncol. 33 (2015) 2004–2012.

[7] N.A. Rizvi, J. Mazieres, D. Planchard, T.E. Stinchcombe, G.K. Dy, S.J. Antonia, L.Horn, H. Lena, E. Minenza, B. Mennecier, G.A. Otterson, L.T. Campos, D.R.Gandara, B.P. Levy, S.G. Nair, G. Zalcman, J. Wolf, P.J. Souquet, E. Baldini, F.Cappuzzo, C. Chouaid, A. Dowlati, R. Sanborn, A. Lopez-Chavez, C. Grohe, R.M.Huber, C.T. Harbison, C. Baudelet, B.J. Lestini, S.S. Ramalingam, Activity andsafety of Nivolumab, an anti-PD-1 immune checkpoint inhibitor, for patientswith advanced, refractory squamous non-small-cell lung cancer (CheckMate063): a phase 2 single-arm trial, Lancet Oncol. 16 (2015) 257–265.

[8] J. Brahmer, K.L. Reckamp, P. Baas, L. Crino, W.E. Eberhardt, E. Poddubskaya, S.Antonia, A. Pluzanski, E.E. Vokes, E. Holgado, D. Waterhouse, N. Ready, J.Gainor, O. Aren Frontera, L. Havel, M. Steins, M.C. Garassino, J.G. Aerts, M.Domine, L. Paz-Ares, M. Reck, C. Baudelet, C.T. Harbison, B. Lestini, D.R. Spigel,Nivolumab versus docetaxel in advanced squamous-cell non-small-cell lungcancer, N. Engl. J. Med. 373 (2015) 123–135.

[9] R.J. Motzer, B.I. Rini, D.F. McDermott, B.G. Redman, T.M. Kuzel, M.R. Harrison,U.N. Vaishampayan, H.A. Drabkin, S. George, T.F. Logan, K.A. Margolin, E.R.Plimack, A.M. Lambert, I.M. Waxman, H.J. Hammers, Nivolumab formetastatic renal cell carcinoma: results of a randomized phase II trial, J. Clin.Oncol. 33 (2015) 1430–1437.

10] J. Hamanishi, M. Mandai, T. Ikeda, M. Minami, A. Kawaguchi, T. Murayama, M.Kanai, Y. Mori, S. Matsumoto, S. Chikuma, N. Matsumura, K. Abiko, T. Baba, K.Yamaguchi, A. Ueda, Y. Hosoe, S. Morita, M. Yokode, A. Shimizu, T. Honjo, I.Konishi, Safety and antitumor activity of anti-PD-1 antibody Nivolumab, inpatients with platinum-resistant ovarian cancer, J. Clin. Oncol. 33 (2015)4015–4022.

11] S. Slovin, Biomarkers for immunotherapy in genitourinary malignancies, Urol.Oncol. (2015), http://dx.doi.org/10.1016/j.urolonc.2015.02.007.

12] M.T. Schweizer, C.G. Drake, Immunotherapy for prostate cancer: recentdevelopments and future challenges, Cancer Metastasis Rev. 33 (2014)641–655.

13] M. Preusser, M. Lim, D.A. Hafler, D.A. Reardon, J.H. Sampson, Prospects ofimmune checkpoint modulators in the treatment of glioblastoma, Nat. Rev.Neurol. 11 (2015) 504–514.

14] S.M. Ansell, A.M. Lesokhin, I. Borrello, A. Halwani, E.C. Scott, M. Gutierrez, S.J.Schuster, M.M. Millenson, D. Cattry, G.J. Freeman, S.J. Rodig, B. Chapuy, A.H.

lymphoma, N. Engl. J. Med. 372 (2015) 311–319.15] M.A. Postow, Managing immune checkpoint-blocking antibody side effects,

in: American Society of Clinical Oncology Educational Book/ASCO, American

Page 8: Journal of Chromatography B - CORE · 2017. 2. 10. · 10 N. Iwamoto et al. / J. Chromatogr. B 1023-1024 (2016) 9–16 Fig. 1. Multiple sequence alignment of Nivolumab amino acid

1 atogr.

[

[

[

[

[

[

[

[

[

[

[

Lebbe, J. Charles, C. Mihalcioiu, V. Chiarion-Sileni, C. Mauch, F. Cognetti, A.Arance, H. Schmidt, D. Schadendorf, H. Gogas, L. Lundgren-Eriksson, C. Horak,

6 N. Iwamoto et al. / J. Chrom

Society of Clinical Oncology, 2015, pp. 76–83, http://dx.doi.org/10.14694/EdBook AM.2015.35.76.

16] R.A. Schmerling, Toxicity of checkpoint inhibitors, Chin. Clin. Oncol. 3 (2014)31.

17] F. Torino, A. Barnabei, R.M. Paragliola, P. Marchetti, R. Salvatori, S.M. Corsello,Endocrine side-effects of anti-cancer drugs: mAbs and pituitary dysfunction:clinical evidence and pathogenic hypotheses, Eur. J. Endocrinol. 169 (2013)R153–R164.

18] K.M. Mahoney, M.B. Atkins, Prognostic and predictive markers for the newimmunotherapies, Oncology (Williston Park) 28 (Suppl. 3) (2014) 39–48.

19] S. Pihl, L. Michaut, J. Hendriks, R. Loebbert, J. Ryding, M. Nemansky, L. Vermet,A. Companjen, EBF recommendation for stability testing of anti-drugantibodies; lessons learned from anti-vaccine antibody stability studies,Bioanalysis 6 (2014) 1409–1413.

20] R. Jenkins, J.X. Duggan, A.F. Aubry, J. Zeng, J.W. Lee, L. Cojocaru, D. Dufield, F.Garofolo, S. Kaur, G.A. Schultz, K. Xu, Z. Yang, J. Yu, Y.J. Zhang, F. Vazvaei,Recommendations for validation of LC–MS/MS bioanalytical methods forprotein biotherapeutics, AAPS J. 17 (2015) 1–16.

21] J.X. Duggan, F. Vazvaei, R. Jenkins, Bioanalytical method validationconsiderations for LC–MS/MS assays of therapeutic proteins, Bioanalysis 7(2015) 1389–1395.

22] N. Iwamoto, T. Shimada, Y. Umino, C. Aoki, T.A. Sato, A. Hamada, H. Nakagama,Selective detection of complementarity-determining regions of monoclonal

B 1023-1024 (2016) 9–16

antibody by limiting protease access to the substrate: nano-surface andmolecular-orientation limited proteolysis, Analyst 139 (2014) 576–580.

23] N. Iwamoto, N. Yamane, Y. Umino, A. Hamada, T. Shimada, The developmentof the validated LCMS bioanalysis of trastuzumab in human plasma using aselective detection method for complementarity-determining regions ofmonoclonal antibodies: nano-surface and molecular-orientation limited(nSMOL) proteolysis, Anal. Methods 7 (2015) 9177–9183.

24] N. Iwamoto, Y. Umino, C. Aoki, N. Yamane, A. Hamada, T. Shimada, Fullyvalidated LCMS bioanalysis of Bevacizumab in human plasma usingnano-surface and molecular-orientation limited (nSMOL) proteolysis, DrugMetab. Pharmacokinet. (2015), http://dx.doi.org/10.1016/j.dmpk.2015.11.004.

25] P.A. Ascierto, M. Kalos, D.A. Schaer, M.K. Callahan, J.D. Wolchok, Biomarkersfor immunostimulatory monoclonal antibodies in combination strategies formelanoma and other tumor types, Clin. Cancer Res. 19 (2013) 1009–1020.

26] C. Robert, G.V. Long, B. Brady, C. Dutriaux, M. Maio, L. Mortier, J.C. Hassel, P.Rutkowski, C. McNeil, E. Kalinka-Warzocha, K.J. Savage, M.M. Hernberg, C.

B. Sharkey, I.M. Waxman, V. Atkinson, P.A. Ascierto, Nivolumab in previouslyuntreated melanoma without BRAF mutation, N. Engl. J. Med. 372 (2015)320–330.