1059 ON203: A new antibody targeting the oxidized form of macrophage migration inhibitory factor (oxMIF) exerts antitumorigenic activity and modulates the tumor microenvironment

BackgroundImmunotherapy success in solid cancers is largely dependent on the tumor microenvironment (TME) and its modulation is central to improve clinical outcomes. One of the key players regulating the TME is the macrophage migration inhibitory factor (MIF), which contributes to an immunosuppressi...

Full description

Saved in:
Bibliographic Details
Published inJournal for immunotherapy of cancer Vol. 10; no. Suppl 2; p. A1101
Main Authors Maurer, Barbara, Mirkina, Irina, Mayer, Julia, Landlinger, Christine, Schinagl, Alexander, Thiele, Michael, Kerschbaumer, Randolf
Format Journal Article
LanguageEnglish
Published London BMJ Publishing Group LTD 01.11.2022
Subjects
Online AccessGet full text
ISSN2051-1426
DOI10.1136/jitc-2022-SITC2022.1059

Cover

Abstract BackgroundImmunotherapy success in solid cancers is largely dependent on the tumor microenvironment (TME) and its modulation is central to improve clinical outcomes. One of the key players regulating the TME is the macrophage migration inhibitory factor (MIF), which contributes to an immunosuppressive environment. MIF induces polarization of macrophages to the M2 subtype, suppresses cytotoxic T cells and correlates with poor response to immune checkpoint therapy. MIF expression is associated with tumor aggressiveness, metastasis, and disease progression, but due to its ubiquitous nature is considered an elusive target for therapeutic intervention. In contrast, the disease-related structural isoform of MIF, termed oxMIF, is specifically present in solid tumor tissue. We now determined the antitumorigenic and TME-modifying potential of the new oxMIF-specific antibody ON203.MethodsIn 3D tumoroids retaining an intact TME, which were isolated from colorectal carcinoma patients, tumor cell killing (high-content 3D computational bioimaging) and TME modulation (immune cell composition and activation, secretome analysis) induced by ON203 were analyzed. In vivo tumor penetration was assessed by infrared-labeled ON203 injected in tumor-bearing mice. Efficacy was assessed in human cancer cell-line (PC3) xenografted mice and ON203’s effects on tumor cell proliferation, vascularization and infiltrating immune cells were evaluated by immunohistochemistry.ResultsFour out of five ON203-treated, freshly isolated tumoroids from colorectal carcinoma patients responded with significant tumor cell death. In the responding tumoroids a clear immunomodulatory effect on the tumor-associated immune cells was detected: ON203 activated NK and NKT cells (upregulation of Granzyme B and CD107a) and supported M1 polarization, correlating with reduced IL-10 levels in the secretome of ON203-responding tumoroids.ON203 accumulated and retained in the tumor tissue in vivo and treatment of immunocompromised mice xenografted with human PC3 tumors led to significantly reduced tumor volumes. Tumor cell proliferation (assessed by Ki67 staining quantification) and tumor vessel density (CD31 staining quantification) were strongly decreased and currently ongoing analysis of tumor-infiltrating immune cells by immunohistochemistry and flow cytometry will provide further insights on the immunomodulatory therapeutic effects of ON203.ConclusionsThe anti-oxMIF antibody ON203 demonstrated antitumorigenic effects by (i) reducing tumor cell proliferation, (ii) reducing angiogenesis and intravasation and (iii) by modulating the TME towards immunosupportive functions. In the upcoming clinical Phase 1 trial ON203’s safety, tolerability, pharmacokinetics, and pharmacodynamics in patients with solid tumors will be analyzed to evaluate its potential as a standalone or combinatorial therapy with immune checkpoint inhibitors, kinase inhibitors or antiangiogenic agents.
AbstractList BackgroundImmunotherapy success in solid cancers is largely dependent on the tumor microenvironment (TME) and its modulation is central to improve clinical outcomes. One of the key players regulating the TME is the macrophage migration inhibitory factor (MIF), which contributes to an immunosuppressive environment. MIF induces polarization of macrophages to the M2 subtype, suppresses cytotoxic T cells and correlates with poor response to immune checkpoint therapy. MIF expression is associated with tumor aggressiveness, metastasis, and disease progression, but due to its ubiquitous nature is considered an elusive target for therapeutic intervention. In contrast, the disease-related structural isoform of MIF, termed oxMIF, is specifically present in solid tumor tissue. We now determined the antitumorigenic and TME-modifying potential of the new oxMIF-specific antibody ON203.MethodsIn 3D tumoroids retaining an intact TME, which were isolated from colorectal carcinoma patients, tumor cell killing (high-content 3D computational bioimaging) and TME modulation (immune cell composition and activation, secretome analysis) induced by ON203 were analyzed. In vivo tumor penetration was assessed by infrared-labeled ON203 injected in tumor-bearing mice. Efficacy was assessed in human cancer cell-line (PC3) xenografted mice and ON203’s effects on tumor cell proliferation, vascularization and infiltrating immune cells were evaluated by immunohistochemistry.ResultsFour out of five ON203-treated, freshly isolated tumoroids from colorectal carcinoma patients responded with significant tumor cell death. In the responding tumoroids a clear immunomodulatory effect on the tumor-associated immune cells was detected: ON203 activated NK and NKT cells (upregulation of Granzyme B and CD107a) and supported M1 polarization, correlating with reduced IL-10 levels in the secretome of ON203-responding tumoroids.ON203 accumulated and retained in the tumor tissue in vivo and treatment of immunocompromised mice xenografted with human PC3 tumors led to significantly reduced tumor volumes. Tumor cell proliferation (assessed by Ki67 staining quantification) and tumor vessel density (CD31 staining quantification) were strongly decreased and currently ongoing analysis of tumor-infiltrating immune cells by immunohistochemistry and flow cytometry will provide further insights on the immunomodulatory therapeutic effects of ON203.ConclusionsThe anti-oxMIF antibody ON203 demonstrated antitumorigenic effects by (i) reducing tumor cell proliferation, (ii) reducing angiogenesis and intravasation and (iii) by modulating the TME towards immunosupportive functions. In the upcoming clinical Phase 1 trial ON203’s safety, tolerability, pharmacokinetics, and pharmacodynamics in patients with solid tumors will be analyzed to evaluate its potential as a standalone or combinatorial therapy with immune checkpoint inhibitors, kinase inhibitors or antiangiogenic agents.
Author Thiele, Michael
Schinagl, Alexander
Kerschbaumer, Randolf
Mayer, Julia
Landlinger, Christine
Maurer, Barbara
Mirkina, Irina
Author_xml – sequence: 1
  givenname: Barbara
  surname: Maurer
  fullname: Maurer, Barbara
– sequence: 2
  givenname: Irina
  surname: Mirkina
  fullname: Mirkina, Irina
– sequence: 3
  givenname: Julia
  surname: Mayer
  fullname: Mayer, Julia
– sequence: 4
  givenname: Christine
  surname: Landlinger
  fullname: Landlinger, Christine
– sequence: 5
  givenname: Alexander
  surname: Schinagl
  fullname: Schinagl, Alexander
– sequence: 6
  givenname: Michael
  surname: Thiele
  fullname: Thiele, Michael
– sequence: 7
  givenname: Randolf
  surname: Kerschbaumer
  fullname: Kerschbaumer, Randolf
BookMark eNotkMFOAjEURRujiYh8g03c6GKw005nOu4IESVBWciedNo3QwnTYqcguHLjF_lHfokDuro3eSfnJu8CnVpnAaGrmPTjmKV3SxNURAml0et4NjyUfkx4foI6lPA4ihOanqNe0ywJITFhTAjRQd8H5Ofza_pCCbvHA2zhHUsbTOH0HgfpKwjGVjgsALud0eYDNC6dr7ErcS2Vd-uFrADXpvIyGGexsQtTmOD8HpdStYlv3O55PLrFsAMfmqM9bGrnTQXWKNxCZmvCvj1oXDu9WckAzXHxiLXudgbs1nhna7DhEp2VctVA7z-7aDZ6mA2fosn0cTwcTKJ1luVRUmQ5FUmpCiBSFEwWCde8oCVQlQETBWQpTUTGIS2JyoGXVCsNQnEQecyBddH1n3bt3dsGmjBfuo237eKcCsZ5ItL2Z792y3oR
ContentType Journal Article
Copyright 2022 Author(s) (or their employer(s)) 2022. No commercial re-use. See rights and permissions. Published by BMJ.
Copyright_xml – notice: 2022 Author(s) (or their employer(s)) 2022. No commercial re-use. See rights and permissions. Published by BMJ.
DBID K9.
DOI 10.1136/jitc-2022-SITC2022.1059
DatabaseName ProQuest Health & Medical Complete (Alumni)
DatabaseTitle ProQuest Health & Medical Complete (Alumni)
DatabaseTitleList ProQuest Health & Medical Complete (Alumni)
DeliveryMethod fulltext_linktorsrc
Discipline Medicine
EISSN 2051-1426
EndPage A1101
GroupedDBID 4.4
53G
5VS
7X7
88E
8FI
8FJ
9YT
ABUWG
ACGFS
ADBBV
ADRAZ
AFKRA
AHBYD
AHYZX
ALIPV
ALMA_UNASSIGNED_HOLDINGS
AMKLP
AOIJS
ASPBG
AVWKF
BAWUL
BCNDV
BENPR
BFQNJ
BMC
BPHCQ
BVXVI
C6C
CCPQU
DIK
EBS
FYUFA
GROUPED_DOAJ
HMCUK
HYE
IAO
IHR
IHW
INH
INR
K9.
KQ8
M1P
M48
M~E
OK1
PHGZM
PHGZT
PIMPY
PJZUB
PPXIY
PQQKQ
PROAC
PSQYO
RBZ
RMJ
RPM
RSV
SOJ
UKHRP
ID FETCH-LOGICAL-p779-4b79284fcbe0a8b3ab45d5b2fe2c7e38be7624875e6f0c9e5f2dcde8c5e8915e3
IEDL.DBID M48
IngestDate Fri Jul 25 22:36:03 EDT 2025
IsDoiOpenAccess false
IsOpenAccess true
IsPeerReviewed true
IsScholarly true
Issue Suppl 2
Language English
LinkModel DirectLink
MergedId FETCHMERGED-LOGICAL-p779-4b79284fcbe0a8b3ab45d5b2fe2c7e38be7624875e6f0c9e5f2dcde8c5e8915e3
Notes ObjectType-Article-1
SourceType-Scholarly Journals-1
ObjectType-Feature-2
content type line 14
OpenAccessLink https://jitc.bmj.com/content/jitc/10/Suppl_2/A1101.full.pdf
PQID 2835548620
PQPubID 2040222
ParticipantIDs proquest_journals_2835548620
PublicationCentury 2000
PublicationDate 20221101
PublicationDateYYYYMMDD 2022-11-01
PublicationDate_xml – month: 11
  year: 2022
  text: 20221101
  day: 01
PublicationDecade 2020
PublicationPlace London
PublicationPlace_xml – name: London
PublicationTitle Journal for immunotherapy of cancer
PublicationYear 2022
Publisher BMJ Publishing Group LTD
Publisher_xml – name: BMJ Publishing Group LTD
SSID ssj0001033888
Score 2.20359
Snippet BackgroundImmunotherapy success in solid cancers is largely dependent on the tumor microenvironment (TME) and its modulation is central to improve clinical...
SourceID proquest
SourceType Aggregation Database
StartPage A1101
SubjectTerms Antibodies
Cell growth
Colorectal cancer
Immunotherapy
Tumors
Title 1059 ON203: A new antibody targeting the oxidized form of macrophage migration inhibitory factor (oxMIF) exerts antitumorigenic activity and modulates the tumor microenvironment
URI https://www.proquest.com/docview/2835548620
Volume 10
hasFullText 1
inHoldings 1
isFullTextHit
isPrint
link http://utb.summon.serialssolutions.com/2.0.0/link/0/eLvHCXMwfV1LSwMxEA5aQbyIT3zUMgcPeljdbveRFURULCq0ilboTTbJrI3YXbUrVE9e_EX-I3-Jk3RFQfG2kE0CyWTm-5JMPsbW4zBJZBrGjoyiwPEToWjNBaEj6gl3uY9K2kd9Wu3w-Mo_7Qbd78y6cgAHf1I7oyd19Xi3NXx43qMFv1sqkmzf6kLSbBOrujzpHJoPo14bj7MJe2hk7vOVmN9uvLjEyjgv73r9U_-Xd7YhpznDpkusCPujyZ1lY5jNsclWeRo-z95N_Y_Xt7O25zZ2YB8IIgONlBa5eobRHW-KTEAYD_KhVvoFFRiQCnkK_cSId_XInUBf34zsAHTW00Kbc3cYCfHARj5snTQ3wUgzFQPbevHUN3JamGkJJi3CqE9QgYJ-rowYGA5sj_Y3apu6-ZFNt8A6zaPO4bFTijA491EUO76IYopgqRToJlw0EuEHKhBeip6MsMEFkjc1pAfD1JUxBqmnpEIuA-RxPcDGIqtkeYZLDOpc-Ygy9YUvyQgEJyNBgmfE0HmYhPVlVv0a7-svO7g2z8ERqQo9d-X_4lU2ZafQpglWWaV4fMI1wguFqLHxqBvV2MTBUfv8omZZd83axCd52cgo
linkProvider Scholars Portal
openUrl ctx_ver=Z39.88-2004&ctx_enc=info%3Aofi%2Fenc%3AUTF-8&rfr_id=info%3Asid%2Fsummon.serialssolutions.com&rft_val_fmt=info%3Aofi%2Ffmt%3Akev%3Amtx%3Ajournal&rft.genre=article&rft.atitle=1059%E2%80%85ON203%3A+A+new+antibody+targeting+the+oxidized+form+of+macrophage+migration+inhibitory+factor+%28oxMIF%29+exerts+antitumorigenic+activity+and+modulates+the+tumor+microenvironment&rft.jtitle=Journal+for+immunotherapy+of+cancer&rft.au=Maurer%2C+Barbara&rft.au=Mirkina%2C+Irina&rft.au=Mayer%2C+Julia&rft.au=Landlinger%2C+Christine&rft.date=2022-11-01&rft.pub=BMJ+Publishing+Group+LTD&rft.eissn=2051-1426&rft.volume=10&rft.issue=Suppl+2&rft.spage=A1101&rft.epage=A1101&rft_id=info:doi/10.1136%2Fjitc-2022-SITC2022.1059&rft.externalDBID=HAS_PDF_LINK