Skip to main content
Erschienen in: Journal of Hematology & Oncology 1/2022

Open Access 01.12.2022 | Correspondence

SIRPα-Fc fusion protein IMM01 exhibits dual anti-tumor activities by targeting CD47/SIRPα signal pathway via blocking the “don’t eat me” signal and activating the “eat me” signal

verfasst von: Jifeng Yu, Song Li, Dianze Chen, Dandan Liu, Huiqin Guo, Chunmei Yang, Wei Zhang, Li Zhang, Gui Zhao, Xiaoping Tu, Liang Peng, Sijin Liu, Xing Bai, Yongping Song, Zhongxing Jiang, Ruliang Zhang, Wenzhi Tian

Erschienen in: Journal of Hematology & Oncology | Ausgabe 1/2022

download
DOWNLOAD
print
DRUCKEN
insite
SUCHEN

Abstract

A novel recombinant SIRPα-Fc fusion protein, IMM01, was constructed and produced using an in-house developed CHO-K1 cell expression system, and the anti-tumor mechanism of IMM01 targeting the CD47-SIRPα pathway was explored. The phagocytosis and in vitro anti-tumor activity of IMM01 were evaluated by antibody-dependent cellular phagocytosis (ADCP), antibody-dependent cell-mediated cytotoxicity (ADCC), and complement-dependent cytotoxicity (CDC) assays. In vivo mouse tumor model studies were used to explore therapeutic efficacy as well as the mechanism of action. An in vitro binding assay revealed that IMM01 has a strong binding affinity to CD47 with an EC50 of 0.4967 nM. IMM01 can induce strong ADCP and moderate ADCC, but not CDC. IMM01-induced strong phagocytosis against tumor cells was attributed to dual activities of blocking the "don’t eat me" signal and activating the "eat me" signal, and IMM01 exhibits strong and robust in vivo anti-tumor activities either as monotherapy on hematological malignancies, or in combination therapy with PD-L1 monoclonal antibody (mAb), PD-1 mAb, and HER-2 mAb on solid tumors. Finally, IMM01 demonstrated a favorable safety profile with no human RBC binding activity or hemagglutination induction. IMM01 inhibits the growth of tumor cells by the following three possible mechanisms: (1) directly activating macrophages to phagocytize tumor cells; (2) activated macrophages degrade phagocytized tumor cells and present tumor antigens to T cells through MHC molecules to activate T cells; (3) activated macrophages can convert "cold tumors" into "hot tumors" and increase the infiltration of immune cells through chemotaxis by secreting some cytokines and chemokines.
Hinweise

Supplementary Information

The online version contains supplementary material available at https://​doi.​org/​10.​1186/​s13045-022-01385-2.
Jifeng Yu and Song Li contributed equally to this work

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
ADCC
Antibody-dependent cell-mediated cytotoxicity
ADCP
Antibody-dependent cellular phagocytosis
CDC
Complement-dependent cytotoxicity
CR
Complete remission
mAb
Monoclonal antibody
RBC
Red blood cells
TGI
Tumor growth inhibition value
To the Editor,
Recently, we reported the crystal structure of human CD47 in a complex with engineered SIRPα.D1 (N80A) [1]. The CD47 surface was buried by IMM01, a novel established new generation recombinant SIRPα-Fc fusion protein targeting CD47. Here, we report the anti-tumor therapeutic potential of IMM01.
IMM01 can significantly block the binding of a chimeric SIRPα receptor expressing cell (Jurkat-CSR) to CD47 (Additional file 1: Fig. S1A, B, C), inhibits the apoptosis of Jurkat-CSR cells via CD47-Fc (Additional file 1: Fig. S1D) and induces antibody-dependent cellular phagocytosis (ADCP) with an EC50 of 0.1389 nM (Additional file 1: Fig. S2A). The maximum antibody-dependent cell-mediated cytotoxicity (ADCC) levels were 1 nM and 5 nM for rituximab and IMM01 (Additional file 1: Fig. S2B), respectively. Rituximab induced strong complement-dependent cytotoxicity (CDC) activity in a dose saturation manner. IMM01 and Herceptin, however, showed no CDC induction activity (Additional file 1: Fig. S2C), implying that IMM01 induces strong ADCP and ADCC but not the CDC.
IMM01 has strong binding activity on all seventeen cancer cell lines, including Raji, Daudi, SU-DHL-10, Jurkat, HL60, MV-4-11, Reh, HCC827, NCI-H1299, NCI-H1975, A549, BT474, SK-BR-3, SK-OV-3, Hela, AGS and HT-29 (Additional file 1: Fig. S3), limited binding activities on human T, B, NK, and monocyte cells (Additional file 1: Fig. S4), and importantly, no binding activity on red blood cells (RBC). This indicates that IMM01 has a superior safety profile and will not have the so-called antigen sink phenomenon [26]. IMM01 reacts with cynomolgus CD47 but not with mouse or rat CD47 (Additional file 1: Fig. S5). IMM01 does not bind to human RBCs (Additional file 1: Fig. S6) to induce hemagglutination (Additional file 1: Fig. S7) and phagocytosis, but it binds to cynomolgus RBCs and induces phagocytosis (Additional file 1: Fig. S7). The N-linked glycosylation of CD47 protein contributes to the RBC non-binding attributes of IMM01 (Additional file 1: Fig. S8). Furthermore, IMM01 stimulates IL-10 and TNF secretion but not IL-1β, IL-2, IL-4, IL-5, IL-6, GM-CSF, and IFN-γ (Additional file 1: Fig. S9), indicating that IMM01 will not cause the cytokine release storm [7, 8].
IMM01 induced strong phagocytosis by binding to FcγRIIA and FcγRIIIA (Additional file 1: Fig. S10A, B), while IMM01M (D265A mutant) diminished phagocytosis due to the reduced binding activity to FcγRs. This suggests that the blocking axis of the CD47/SIRPα pathway not only needs to block the “don’t eat me” signal from CD47/SIRPα interaction but also needs to activate the “eat me” signal by the effective engagement of Fc with activating FcγRs in macrophages[2, 46].
The HL-60 xenograft model (Additional file 1: Fig. S11) demonstrated that 100% of the mice achieved complete remission (CR) after administration of IMM01, whereas 0% of the mice achieved CR after administration of IMM01M-inactive Fc for 2 weeks. Furthermore, 100% of the mice achieved CR after administration of IMM01 with the macrophages intact, whereas 0% of the mice achieved CR after macrophage depletion, indicating that IMM01 performs the therapeutic function through effective Fc function. The Daudi xenograft tumor model (Fig. 1A,B) revealed that the tumor growth inhibition (TGI) value was 97.48% on the day of 24 after administration of IMM01. A Raji orthotopic model showed that IMM01 has better effects than rituximab, and the combination of IMM01 and rituximab has significantly better results than any single drug alone (Fig. 1C).
In CT26-hPDL1/hCD47 syngeneic tumor model in hPD-1/hSIRPα Tg Balb/c nude mice, the combination of IMM01 with PD-1 or PD-L1 antibody resulted in significantly stronger antitumor effects than PD-1 or PD-L1 alone (Fig. 2A,B). The efficacy of IMM01 alone and in combination with CD33 mAb and HER-2 mAb in the SNU-1 and HL-60 xenograft models (Fig. 2C, D), respectively, revealed great therapeutic potential in combination with these mAbs [912], as well as synergistic efficacy with pomalidomide and dexamethasone (Additional file 1: Fig. S12).
In summary, IMM01 exhibits strong anti-tumor activities with dual anti-tumor activities by blocking the CD47 “don’t eat me” signal and activating the phagocytosis “eat me” signal (Additional file 1: Fig. S13). It has good synergistic effects with different immunotherapeutic agents and has no human RBC binding activity and no hemagglutination induction. IMM01 inhibits anti-tumor activity via three possible mechanisms (Additional file 1: Fig. S14): (1) directly activating macrophages to phagocytize tumor cells; (2) presenting tumor antigens through MHC molecules to T cells; (3) activated macrophages can increase the infiltration of immune cells through chemotaxis by secreting some cytokines and chemokines. A phase I/II clinical trial of IMM01 combined with azacytidine for acute myeloid leukemia and myelodysplastic syndrome has been initiated (NCT05140811).

Acknowledgments

Authors would like to thank Medicilon Biology, Pharma Legacy, WuXi AppTec and GemPharmaTech for the wonderful services and technical support.

Declarations

Murine studies were conducted after IACUC approved protocol. Blood samples were collected from healthy donors with written consent.
All authors critically reviewed and approved the final manuscript.

Competing interests

Song Li, Dianze Chen, Dandan Liu, Huiqin Guo, Chunmei Yang, Wei Zhang, Li Zhang, Gui Zhao, Xiaoping Tu, Liang Peng, Sijin Liu, Xing Bai, and Ruliang Zhang are employees in ImmuneOnco Biopharmaceuticals (Shanghai) Co., Ltd. Wenzhi Tian is the founder of ImmuneOnco Biopharmaceuticals (Shanghai) Co., Ltd. Jifeng Yu, Yongping Song, Zhongxing Jiang declared no conflicts of interest.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

Literatur
1.
Zurück zum Zitat Yu J, Li S, Chen D, Liu D, Guo H, Yang C, et al. Crystal structure of human CD47 in complex with engineered SIRPα D1(N80A). Molecules. 2022;27(17):5574.CrossRefPubMedPubMedCentral Yu J, Li S, Chen D, Liu D, Guo H, Yang C, et al. Crystal structure of human CD47 in complex with engineered SIRPα D1(N80A). Molecules. 2022;27(17):5574.CrossRefPubMedPubMedCentral
3.
Zurück zum Zitat Pinho SS, Reis CA. Glycosylation in cancer: mechanisms and clinical implications. Nat Rev Cancer. 2015;15(9):540–55.CrossRefPubMed Pinho SS, Reis CA. Glycosylation in cancer: mechanisms and clinical implications. Nat Rev Cancer. 2015;15(9):540–55.CrossRefPubMed
4.
Zurück zum Zitat Li W, Wang F, Guo R, Bian Z, Song Y. Targeting macrophages in hematological malignancies: recent advances and future directions. J Hematol Oncol. 2022;15(1):110.CrossRefPubMedPubMedCentral Li W, Wang F, Guo R, Bian Z, Song Y. Targeting macrophages in hematological malignancies: recent advances and future directions. J Hematol Oncol. 2022;15(1):110.CrossRefPubMedPubMedCentral
5.
Zurück zum Zitat Zhang A, Ren Z, Tseng KF, Liu X, Li H, Lu C, et al. Dual targeting of CTLA-4 and CD47 on T(reg) cells promotes immunity against solid tumors. Sci Transl Med. 2021;13(605):eabg8693.CrossRefPubMed Zhang A, Ren Z, Tseng KF, Liu X, Li H, Lu C, et al. Dual targeting of CTLA-4 and CD47 on T(reg) cells promotes immunity against solid tumors. Sci Transl Med. 2021;13(605):eabg8693.CrossRefPubMed
6.
Zurück zum Zitat Upton R, Banuelos A, Feng D, Biswas T, Kao K, McKenna K, et al. Combining CD47 blockade with trastuzumab eliminates HER2-positive breast cancer cells and overcomes trastuzumab tolerance. Proc Natl Acad Sci United States Am. 2021;118(29):e2026849118.CrossRef Upton R, Banuelos A, Feng D, Biswas T, Kao K, McKenna K, et al. Combining CD47 blockade with trastuzumab eliminates HER2-positive breast cancer cells and overcomes trastuzumab tolerance. Proc Natl Acad Sci United States Am. 2021;118(29):e2026849118.CrossRef
7.
Zurück zum Zitat Yuen KC, Liu LF, Gupta V, Madireddi S, Keerthivasan S, Li C, et al. High systemic and tumor-associated IL-8 correlates with reduced clinical benefit of PD-L1 blockade. Nat Med. 2020;26(5):693–8.CrossRefPubMedPubMedCentral Yuen KC, Liu LF, Gupta V, Madireddi S, Keerthivasan S, Li C, et al. High systemic and tumor-associated IL-8 correlates with reduced clinical benefit of PD-L1 blockade. Nat Med. 2020;26(5):693–8.CrossRefPubMedPubMedCentral
8.
Zurück zum Zitat Schalper KA, Carleton M, Zhou M, Chen T, Feng Y, Huang SP, et al. Elevated serum interleukin-8 is associated with enhanced intratumor neutrophils and reduced clinical benefit of immune-checkpoint inhibitors. Nat Med. 2020;26(5):688–92.CrossRefPubMedPubMedCentral Schalper KA, Carleton M, Zhou M, Chen T, Feng Y, Huang SP, et al. Elevated serum interleukin-8 is associated with enhanced intratumor neutrophils and reduced clinical benefit of immune-checkpoint inhibitors. Nat Med. 2020;26(5):688–92.CrossRefPubMedPubMedCentral
9.
Zurück zum Zitat Andrechak JC, Dooling LJ, Tobin MP, Zhang W, Hayes BH, Lee JY, et al. CD47-SIRPα checkpoint disruption in metastases requires tumor-targeting antibody for molecular and engineered macrophage therapies. Cancers. 2022;14(8):1930.CrossRefPubMedPubMedCentral Andrechak JC, Dooling LJ, Tobin MP, Zhang W, Hayes BH, Lee JY, et al. CD47-SIRPα checkpoint disruption in metastases requires tumor-targeting antibody for molecular and engineered macrophage therapies. Cancers. 2022;14(8):1930.CrossRefPubMedPubMedCentral
11.
Zurück zum Zitat Müller K, Vogiatzi F, Winterberg D, Rösner T, Lenk L, Bastian L, et al. Combining daratumumab with CD47 blockade prolongs survival in preclinical models of pediatric T-ALL. Blood. 2022;140(1):45–57.CrossRefPubMed Müller K, Vogiatzi F, Winterberg D, Rösner T, Lenk L, Bastian L, et al. Combining daratumumab with CD47 blockade prolongs survival in preclinical models of pediatric T-ALL. Blood. 2022;140(1):45–57.CrossRefPubMed
Metadaten
Titel
SIRPα-Fc fusion protein IMM01 exhibits dual anti-tumor activities by targeting CD47/SIRPα signal pathway via blocking the “don’t eat me” signal and activating the “eat me” signal
verfasst von
Jifeng Yu
Song Li
Dianze Chen
Dandan Liu
Huiqin Guo
Chunmei Yang
Wei Zhang
Li Zhang
Gui Zhao
Xiaoping Tu
Liang Peng
Sijin Liu
Xing Bai
Yongping Song
Zhongxing Jiang
Ruliang Zhang
Wenzhi Tian
Publikationsdatum
01.12.2022
Verlag
BioMed Central
Erschienen in
Journal of Hematology & Oncology / Ausgabe 1/2022
Elektronische ISSN: 1756-8722
DOI
https://doi.org/10.1186/s13045-022-01385-2

Weitere Artikel der Ausgabe 1/2022

Journal of Hematology & Oncology 1/2022 Zur Ausgabe

Mehr Lebenszeit mit Abemaciclib bei fortgeschrittenem Brustkrebs?

24.05.2024 Mammakarzinom Nachrichten

In der MONARCHE-3-Studie lebten Frauen mit fortgeschrittenem Hormonrezeptor-positivem, HER2-negativem Brustkrebs länger, wenn sie zusätzlich zu einem nicht steroidalen Aromatasehemmer mit Abemaciclib behandelt wurden; allerdings verfehlte der numerische Zugewinn die statistische Signifikanz.

ADT zur Radiatio nach Prostatektomie: Wenn, dann wohl länger

24.05.2024 Prostatakarzinom Nachrichten

Welchen Nutzen es trägt, wenn die Strahlentherapie nach radikaler Prostatektomie um eine Androgendeprivation ergänzt wird, hat die RADICALS-HD-Studie untersucht. Nun liegen die Ergebnisse vor. Sie sprechen für länger dauernden Hormonentzug.

Das sind die führenden Symptome junger Darmkrebspatienten

Darmkrebserkrankungen in jüngeren Jahren sind ein zunehmendes Problem, das häufig längere Zeit übersehen wird, gerade weil die Patienten noch nicht alt sind. Welche Anzeichen Ärzte stutzig machen sollten, hat eine Metaanalyse herausgearbeitet.

„Überwältigende“ Evidenz für Tripeltherapie beim metastasierten Prostata-Ca.

22.05.2024 Prostatakarzinom Nachrichten

Patienten mit metastasiertem hormonsensitivem Prostatakarzinom sollten nicht mehr mit einer alleinigen Androgendeprivationstherapie (ADT) behandelt werden, mahnt ein US-Team nach Sichtung der aktuellen Datenlage. Mit einer Tripeltherapie haben die Betroffenen offenbar die besten Überlebenschancen.

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.