Engineering Calreticulin-Targeting Monobodies to Detect Immunogenic Cell Death in Cancer Chemotherapy

Surface-exposed calreticulin (ecto-CRT) plays a crucial role in the phagocytic removal of apoptotic cells during immunotherapy. Ecto-CRT is an immunogenic signal induced in response to treatment with chemotherapeutic agents such as doxorubicin (DOX) and mitoxantrone (MTX), and two peptides (KLGFFKR...

Full description

Bibliographic Details
Main Authors: Ying Zhang, Ramar Thangam, Sung-Hwan You, Rukhsora D. Sultonova, Akhil Venu, Jung-Joon Min, Yeongjin Hong
Format: Article
Language:English
Published: MDPI AG 2021-06-01
Series:Cancers
Subjects:
Online Access:https://www.mdpi.com/2072-6694/13/11/2801
_version_ 1797531395604611072
author Ying Zhang
Ramar Thangam
Sung-Hwan You
Rukhsora D. Sultonova
Akhil Venu
Jung-Joon Min
Yeongjin Hong
author_facet Ying Zhang
Ramar Thangam
Sung-Hwan You
Rukhsora D. Sultonova
Akhil Venu
Jung-Joon Min
Yeongjin Hong
author_sort Ying Zhang
collection DOAJ
description Surface-exposed calreticulin (ecto-CRT) plays a crucial role in the phagocytic removal of apoptotic cells during immunotherapy. Ecto-CRT is an immunogenic signal induced in response to treatment with chemotherapeutic agents such as doxorubicin (DOX) and mitoxantrone (MTX), and two peptides (KLGFFKR (Integrin-α) and GQPMYGQPMY (CRT binding peptide 1, Hep-I)) are known to specifically bind CRT. To engineer CRT-specific monobodies as agents to detect immunogenic cell death (ICD), we fused these peptide sequences at the binding loops (BC and FG) of human fibronectin domain III (FN3). CRT-specific monobodies were purified from <i>E. coli</i> by affinity chromatography. Using these monobodies, ecto-CRT was evaluated in vitro, in cultured cancer cell lines (CT-26, MC-38, HeLa, and MDA-MB-231), or in mice after anticancer drug treatment. Monobodies with both peptide sequences (CRT3 and CRT4) showed higher binding to ecto-CRT than those with a single peptide sequence. The binding affinity of the Rluc8 fusion protein–engineered monobodies (CRT3-Rluc8 and CRT4-Rluc8) to CRT was about 8 nM, and the half-life in serum and tumor tissue was about 12 h. By flow cytometry and confocal immunofluorescence of cancer cell lines, and by in vivo optical bioluminescence imaging of tumor-bearing mice, CRT3-Rluc8 and CRT4-Rluc8 bound specifically to ecto-CRT and effectively detected pre-apoptotic cells after treatment with ICD-inducing agents (DOX and MTX) but not a non-ICD-inducing agent (gemcitabine). Using CRT-specific monobodies, it is possible to detect ecto-CRT induction in cancer cells in response to drug exposure. This technique may be used to predict the therapeutic efficiency of chemo- and immuno-therapeutics early during anticancer treatment.
first_indexed 2024-03-10T10:43:11Z
format Article
id doaj.art-ad36d1e331c942e49d7f5b8cf6b0e97f
institution Directory Open Access Journal
issn 2072-6694
language English
last_indexed 2024-03-10T10:43:11Z
publishDate 2021-06-01
publisher MDPI AG
record_format Article
series Cancers
spelling doaj.art-ad36d1e331c942e49d7f5b8cf6b0e97f2023-11-21T22:47:51ZengMDPI AGCancers2072-66942021-06-011311280110.3390/cancers13112801Engineering Calreticulin-Targeting Monobodies to Detect Immunogenic Cell Death in Cancer ChemotherapyYing Zhang0Ramar Thangam1Sung-Hwan You2Rukhsora D. Sultonova3Akhil Venu4Jung-Joon Min5Yeongjin Hong6Department of Nuclear Medicine, Institute for Molecular Imaging and Theranostics, Hwasun Hospital, Chonnam National University Medical School, Hwasun 58128, KoreaDepartment of Nuclear Medicine, Institute for Molecular Imaging and Theranostics, Hwasun Hospital, Chonnam National University Medical School, Hwasun 58128, KoreaDepartment of Nuclear Medicine, Institute for Molecular Imaging and Theranostics, Hwasun Hospital, Chonnam National University Medical School, Hwasun 58128, KoreaDepartment of Nuclear Medicine, Institute for Molecular Imaging and Theranostics, Hwasun Hospital, Chonnam National University Medical School, Hwasun 58128, KoreaDepartment of Nuclear Medicine, Institute for Molecular Imaging and Theranostics, Hwasun Hospital, Chonnam National University Medical School, Hwasun 58128, KoreaDepartment of Nuclear Medicine, Institute for Molecular Imaging and Theranostics, Hwasun Hospital, Chonnam National University Medical School, Hwasun 58128, KoreaDepartment of Nuclear Medicine, Institute for Molecular Imaging and Theranostics, Hwasun Hospital, Chonnam National University Medical School, Hwasun 58128, KoreaSurface-exposed calreticulin (ecto-CRT) plays a crucial role in the phagocytic removal of apoptotic cells during immunotherapy. Ecto-CRT is an immunogenic signal induced in response to treatment with chemotherapeutic agents such as doxorubicin (DOX) and mitoxantrone (MTX), and two peptides (KLGFFKR (Integrin-α) and GQPMYGQPMY (CRT binding peptide 1, Hep-I)) are known to specifically bind CRT. To engineer CRT-specific monobodies as agents to detect immunogenic cell death (ICD), we fused these peptide sequences at the binding loops (BC and FG) of human fibronectin domain III (FN3). CRT-specific monobodies were purified from <i>E. coli</i> by affinity chromatography. Using these monobodies, ecto-CRT was evaluated in vitro, in cultured cancer cell lines (CT-26, MC-38, HeLa, and MDA-MB-231), or in mice after anticancer drug treatment. Monobodies with both peptide sequences (CRT3 and CRT4) showed higher binding to ecto-CRT than those with a single peptide sequence. The binding affinity of the Rluc8 fusion protein–engineered monobodies (CRT3-Rluc8 and CRT4-Rluc8) to CRT was about 8 nM, and the half-life in serum and tumor tissue was about 12 h. By flow cytometry and confocal immunofluorescence of cancer cell lines, and by in vivo optical bioluminescence imaging of tumor-bearing mice, CRT3-Rluc8 and CRT4-Rluc8 bound specifically to ecto-CRT and effectively detected pre-apoptotic cells after treatment with ICD-inducing agents (DOX and MTX) but not a non-ICD-inducing agent (gemcitabine). Using CRT-specific monobodies, it is possible to detect ecto-CRT induction in cancer cells in response to drug exposure. This technique may be used to predict the therapeutic efficiency of chemo- and immuno-therapeutics early during anticancer treatment.https://www.mdpi.com/2072-6694/13/11/2801monobodycalreticulinimmunogenic cell death (ICD)optical bioluminescence imagingearly prediction of chemotherapeutic response
spellingShingle Ying Zhang
Ramar Thangam
Sung-Hwan You
Rukhsora D. Sultonova
Akhil Venu
Jung-Joon Min
Yeongjin Hong
Engineering Calreticulin-Targeting Monobodies to Detect Immunogenic Cell Death in Cancer Chemotherapy
Cancers
monobody
calreticulin
immunogenic cell death (ICD)
optical bioluminescence imaging
early prediction of chemotherapeutic response
title Engineering Calreticulin-Targeting Monobodies to Detect Immunogenic Cell Death in Cancer Chemotherapy
title_full Engineering Calreticulin-Targeting Monobodies to Detect Immunogenic Cell Death in Cancer Chemotherapy
title_fullStr Engineering Calreticulin-Targeting Monobodies to Detect Immunogenic Cell Death in Cancer Chemotherapy
title_full_unstemmed Engineering Calreticulin-Targeting Monobodies to Detect Immunogenic Cell Death in Cancer Chemotherapy
title_short Engineering Calreticulin-Targeting Monobodies to Detect Immunogenic Cell Death in Cancer Chemotherapy
title_sort engineering calreticulin targeting monobodies to detect immunogenic cell death in cancer chemotherapy
topic monobody
calreticulin
immunogenic cell death (ICD)
optical bioluminescence imaging
early prediction of chemotherapeutic response
url https://www.mdpi.com/2072-6694/13/11/2801
work_keys_str_mv AT yingzhang engineeringcalreticulintargetingmonobodiestodetectimmunogeniccelldeathincancerchemotherapy
AT ramarthangam engineeringcalreticulintargetingmonobodiestodetectimmunogeniccelldeathincancerchemotherapy
AT sunghwanyou engineeringcalreticulintargetingmonobodiestodetectimmunogeniccelldeathincancerchemotherapy
AT rukhsoradsultonova engineeringcalreticulintargetingmonobodiestodetectimmunogeniccelldeathincancerchemotherapy
AT akhilvenu engineeringcalreticulintargetingmonobodiestodetectimmunogeniccelldeathincancerchemotherapy
AT jungjoonmin engineeringcalreticulintargetingmonobodiestodetectimmunogeniccelldeathincancerchemotherapy
AT yeongjinhong engineeringcalreticulintargetingmonobodiestodetectimmunogeniccelldeathincancerchemotherapy