3 16

Cited 0 times in

GNUV201, a novel human/mouse cross-reactive and low pH-selective anti-PD-1 monoclonal antibody for cancer immunotherapy

DC Field Value Language
dc.contributor.author나혜영-
dc.contributor.author박채규-
dc.date.accessioned2024-07-18T05:19:35Z-
dc.date.available2024-07-18T05:19:35Z-
dc.date.issued2024-05-
dc.identifier.urihttps://ir.ymlib.yonsei.ac.kr/handle/22282913/200061-
dc.description.abstractBackground: Several PD-1 antibodies approved as anti-cancer therapies work by blocking the interaction of PD-1 with its ligand PD-L1, thus restoring anti-cancer T cell activities. These PD-1 antibodies lack inter-species cross-reactivity, necessitating surrogate antibodies for preclinical studies, which may limit the predictability and translatability of the studies. Results: To overcome this limitation, we have developed an inter-species cross-reactive PD-1 antibody, GNUV201, by utilizing an enhanced diversity mouse platform (SHINE MOUSE™). GNUV201 equally binds to human PD-1 and mouse PD-1, equally inhibits the binding of human PD-1/PD-L1 and mouse PD-1/PD-L1, and effectively suppresses tumor growth in syngeneic mouse models. The epitope of GNUV201 mapped to the "FG loop" of hPD-1, distinct from those of Keytruda® ("C'D loop") and Opdivo® (N-term). Notably, the structural feature where the protruding epitope loop fits into GNUV201's binding pocket supports the enhanced binding affinity due to slower dissociation (8.7 times slower than Keytruda®). Furthermore, GNUV201 shows a stronger binding affinity at pH 6.0 (5.6 times strong than at pH 7.4), which mimics the hypoxic and acidic tumor microenvironment (TME). This phenomenon is not observed with marketed antibodies (Keytruda®, Opdivo®), implying that GNUV201 achieves more selective binding to and better occupancy on PD-1 in the TME. Conclusions: In summary, GNUV201 exhibited enhanced affinity for PD-1 with slow dissociation and preferential binding in TME-mimicking low pH. Human/monkey/mouse inter-species cross-reactivity of GNUV201 could enable more predictable and translatable efficacy and toxicity preclinical studies. These results suggest that GNUV201 could be an ideal antibody candidate for anti-cancer drug development.-
dc.description.statementOfResponsibilityopen-
dc.languageEnglish-
dc.publisherBioMed Central-
dc.relation.isPartOfBMC IMMUNOLOGY-
dc.rightsCC BY-NC-ND 2.0 KR-
dc.subject.MESHAnimals-
dc.subject.MESHAntibodies, Monoclonal / immunology-
dc.subject.MESHAntibodies, Monoclonal / pharmacology-
dc.subject.MESHAntibodies, Monoclonal / therapeutic use-
dc.subject.MESHAntibodies, Monoclonal, Humanized / immunology-
dc.subject.MESHAntibodies, Monoclonal, Humanized / pharmacology-
dc.subject.MESHAntibodies, Monoclonal, Humanized / therapeutic use-
dc.subject.MESHB7-H1 Antigen / antagonists & inhibitors-
dc.subject.MESHB7-H1 Antigen / immunology-
dc.subject.MESHB7-H1 Antigen / metabolism-
dc.subject.MESHCell Line, Tumor-
dc.subject.MESHCross Reactions* / immunology-
dc.subject.MESHEpitopes / immunology-
dc.subject.MESHFemale-
dc.subject.MESHHumans-
dc.subject.MESHHydrogen-Ion Concentration-
dc.subject.MESHImmune Checkpoint Inhibitors / pharmacology-
dc.subject.MESHImmunotherapy* / methods-
dc.subject.MESHMice-
dc.subject.MESHMice, Inbred C57BL-
dc.subject.MESHNeoplasms / immunology-
dc.subject.MESHNeoplasms / therapy-
dc.subject.MESHProgrammed Cell Death 1 Receptor* / antagonists & inhibitors-
dc.subject.MESHProgrammed Cell Death 1 Receptor* / immunology-
dc.subject.MESHProgrammed Cell Death 1 Receptor* / metabolism-
dc.titleGNUV201, a novel human/mouse cross-reactive and low pH-selective anti-PD-1 monoclonal antibody for cancer immunotherapy-
dc.typeArticle-
dc.contributor.collegeCollege of Medicine (의과대학)-
dc.contributor.departmentDept. of Neurology (신경과학교실)-
dc.contributor.googleauthorHae-Mi Kim-
dc.contributor.googleauthorKyoung-Jin Kim-
dc.contributor.googleauthorKwanghyun Lee-
dc.contributor.googleauthorMyeong Jin Yoon-
dc.contributor.googleauthorJenny Choih-
dc.contributor.googleauthorTae-Joon Hong-
dc.contributor.googleauthorEun Ji Cho-
dc.contributor.googleauthorHak-Jun Jung-
dc.contributor.googleauthorJayoung Kim-
dc.contributor.googleauthorJi Soo Park-
dc.contributor.googleauthorHye Young Na-
dc.contributor.googleauthorYong-Seok Heo-
dc.contributor.googleauthorChae Gyu Park-
dc.contributor.googleauthorHeungrok Park-
dc.contributor.googleauthorSungho Han-
dc.contributor.googleauthorDonggoo Bae-
dc.identifier.doi10.1186/s12865-024-00609-z-
dc.contributor.localIdA04556-
dc.contributor.localIdA01718-
dc.relation.journalcodeJ03324-
dc.identifier.eissn1471-2172-
dc.identifier.pmid38730320-
dc.subject.keywordAnti-PD-1 antibody-
dc.subject.keywordCancer immunotherapy-
dc.subject.keywordCross-reactive-
dc.subject.keywordMonoclonal antibody-
dc.subject.keywordTME selective-
dc.contributor.alternativeNameNa, Hye Young-
dc.contributor.affiliatedAuthor나혜영-
dc.contributor.affiliatedAuthor박채규-
dc.citation.volume25-
dc.citation.number1-
dc.citation.startPage29-
dc.identifier.bibliographicCitationBMC IMMUNOLOGY, Vol.25(1) : 29, 2024-05-
Appears in Collections:
1. College of Medicine (의과대학) > BioMedical Science Institute (의생명과학부) > 1. Journal Papers
1. College of Medicine (의과대학) > Dept. of Neurology (신경과학교실) > 1. Journal Papers

qrcode

Items in DSpace are protected by copyright, with all rights reserved, unless otherwise indicated.