1
|
Zeng X, Zhang H, Guo J, Yang D, Zhu Y, Liu N, Tang J, Liu T, Zhao X. A novel bispecific T-cell engager using the ligand-target csGRP78 against acute myeloid leukemia. Cell Mol Life Sci 2024; 81:371. [PMID: 39196413 PMCID: PMC11358366 DOI: 10.1007/s00018-024-05410-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 07/31/2024] [Accepted: 08/13/2024] [Indexed: 08/29/2024]
Abstract
Current medical therapies for treating acute myeloid leukemia (AML) remain unmet, and AML patients may benefit from targeted immunotherapy approaches that focus on specific tumor antigens. GRP78, which is upregulated in various malignant tumors such as AML, is partially expressed as cell surface GRP78 (csGRP78) on the cell membrane, making it an ideal target for redirecting T cells, including T-cell engagers. However, considering the conventional approach of using two scFv segments to construct a bispecific T-cell engager (BiTE), we have undertaken the development of a novel BiTE that utilizes a cyclic peptide ligand to specifically target csGRP78, which we refer to as GRP78-CD3/BiTE. We studied the effects of GRP78-CD3/BiTE on treatments for AML in vitro and in vivo and assessed the pharmacokinetics of this engager. Our findings demonstrated that GRP78-CD3/BiTE could not only effectively mediate the cytotoxicity of T cells against csGRP78-expressing AML cells but also specifically eliminate primary AML tumor cells in vitro. Furthermore, GRP78-CD3/BiTE exhibited a longer half-life despite having a lower molecular weight than CD19-CD3/BiTE. In a xenograft mouse model of AML, treatment with GRP78-CD3/BiTE prolonged the survival time of the mice. Our findings demonstrate that GRP78-CD3/BiTE is effective and selective for eliminating csGRP78-expressing AML cells and suggest that this approach to targeted immunotherapy could lead to effective new treatments for AML.
Collapse
MESH Headings
- Endoplasmic Reticulum Chaperone BiP
- Humans
- Animals
- Leukemia, Myeloid, Acute/immunology
- Leukemia, Myeloid, Acute/therapy
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/drug therapy
- Mice
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Antibodies, Bispecific/immunology
- Antibodies, Bispecific/pharmacology
- CD3 Complex/immunology
- Heat-Shock Proteins/immunology
- Heat-Shock Proteins/metabolism
- Xenograft Model Antitumor Assays
- Cell Line, Tumor
- Ligands
- Female
- Mice, SCID
- Immunotherapy/methods
- Mice, Inbred NOD
Collapse
Affiliation(s)
- Xiaozhu Zeng
- Department of Targeting Therapy & Immunology and Laboratory of Animal Tumor Models, Cancer Center and State Key Laboratory of Respiratory Health and Multimorbidity and Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Hang Zhang
- Department of Hematology, Institute of Hematology, West China Hospital of Sichuan University, Chengdu, Sichuan, 610041, China
| | - Jing Guo
- Department of Targeting Therapy & Immunology and Laboratory of Animal Tumor Models, Cancer Center and State Key Laboratory of Respiratory Health and Multimorbidity and Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Dong Yang
- Department of Targeting Therapy & Immunology and Laboratory of Animal Tumor Models, Cancer Center and State Key Laboratory of Respiratory Health and Multimorbidity and Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yongjie Zhu
- Department of Targeting Therapy & Immunology and Laboratory of Animal Tumor Models, Cancer Center and State Key Laboratory of Respiratory Health and Multimorbidity and Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Nan Liu
- Department of Targeting Therapy & Immunology and Laboratory of Animal Tumor Models, Cancer Center and State Key Laboratory of Respiratory Health and Multimorbidity and Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Jie Tang
- Department of Targeting Therapy & Immunology and Laboratory of Animal Tumor Models, Cancer Center and State Key Laboratory of Respiratory Health and Multimorbidity and Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Ting Liu
- Department of Hematology, Institute of Hematology, West China Hospital of Sichuan University, Chengdu, Sichuan, 610041, China.
| | - Xudong Zhao
- Department of Targeting Therapy & Immunology and Laboratory of Animal Tumor Models, Cancer Center and State Key Laboratory of Respiratory Health and Multimorbidity and Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China.
| |
Collapse
|
2
|
Zoine JT, Immadisetty K, Ibanez-Vega J, Moore SE, Nevitt C, Thanekar U, Tian L, Karouni A, Chockley PJ, Arthur B, Sheppard H, Klco JM, Langfitt DM, Krenciute G, Gottschalk S, Babu MM, Velasquez MP. Peptide-scFv antigen recognition domains effectively confer CAR T cell multiantigen specificity. Cell Rep Med 2024; 5:101422. [PMID: 38350450 PMCID: PMC10897625 DOI: 10.1016/j.xcrm.2024.101422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 10/06/2023] [Accepted: 01/19/2024] [Indexed: 02/15/2024]
Abstract
The emergence of immune escape is a significant roadblock to developing effective chimeric antigen receptor (CAR) T cell therapies against hematological malignancies, including acute myeloid leukemia (AML). Here, we demonstrate feasibility of targeting two antigens simultaneously by combining a GRP78-specific peptide antigen recognition domain with a CD123-specific scFv to generate a peptide-scFv bispecific antigen recognition domain (78.123). To achieve this, we test linkers with varying length and flexibility and perform immunophenotypic and functional characterization. We demonstrate that bispecific CAR T cells successfully recognize and kill tumor cells that express GRP78, CD123, or both antigens and have improved antitumor activity compared to their monospecific counterparts when both antigens are expressed. Protein structure prediction suggests that linker length and compactness influence the functionality of the generated bispecific CARs. Thus, we present a bispecific CAR design strategy to prevent immune escape in AML that can be extended to other peptide-scFv combinations.
Collapse
Affiliation(s)
- Jaquelyn T Zoine
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Kalyan Immadisetty
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Department of Structural Biology and Center of Excellence for Data Driven Discovery, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Jorge Ibanez-Vega
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Sarah E Moore
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Chris Nevitt
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Unmesha Thanekar
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Liqing Tian
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Abbas Karouni
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Peter J Chockley
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Bright Arthur
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Heather Sheppard
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Jeffery M Klco
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Deanna M Langfitt
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Giedre Krenciute
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Stephen Gottschalk
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - M Madan Babu
- Department of Structural Biology and Center of Excellence for Data Driven Discovery, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - M Paulina Velasquez
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, Memphis, TN 38105, USA.
| |
Collapse
|
3
|
Guo W, Wang M, Yang Z, Liu D, Ma B, Zhao Y, Chen Y, Hu Y. Recent advances in small molecule and peptide inhibitors of glucose-regulated protein 78 for cancer therapy. Eur J Med Chem 2023; 261:115792. [PMID: 37690265 DOI: 10.1016/j.ejmech.2023.115792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 08/18/2023] [Accepted: 09/02/2023] [Indexed: 09/12/2023]
Abstract
Glucose-regulated protein 78 (GRP78) is one of key endoplasmic reticulum (ER) chaperone proteins that regulates the unfolded protein response (UPR) to maintain ER homeostasis. As a core factor in the regulation of the UPR, GRP78 takes a critical part in the cellular processes required for tumorigenesis, such as proliferation, metastasis, anti-apoptosis, immune escape and chemoresistance. Overexpression of GRP78 is closely correlated with tumorigenesis and poor prognosis in various malignant tumors. Targeting GRP78 is regarded as a potentially promising therapeutic strategy for cancer therapy. Although none of the GRP78 inhibitors have been approved to date, there have been several studies of GRP78 inhibitors. Herein, we comprehensively review the structure, physiological functions of GRP78 and the recent progress of GRP78 inhibitors, and discuss the structures, in vitro and in vivo efficacies, and merits and demerits of these inhibitors to inspire further research. Additionally, the feasibility of GRP78-targeting proteolysis-targeting chimeras (PROTACs), disrupting GRP78 cochaperone interactions, or covalent inhibition are also discussed as novel strategies for drugs discovery targeting GRP78, with the hope that these strategies can provide new opportunities for targeted GRP78 antitumor therapy.
Collapse
Affiliation(s)
- Weikai Guo
- The Jointed National Laboratory of Antibody Drug Engineering, The First Affiliated Hospital of Henan University, Henan University, Kaifeng, 475004, China
| | - Manjie Wang
- The Jointed National Laboratory of Antibody Drug Engineering, The First Affiliated Hospital of Henan University, Henan University, Kaifeng, 475004, China
| | - Zhengfan Yang
- The Jointed National Laboratory of Antibody Drug Engineering, The First Affiliated Hospital of Henan University, Henan University, Kaifeng, 475004, China
| | - Danyang Liu
- The Jointed National Laboratory of Antibody Drug Engineering, The First Affiliated Hospital of Henan University, Henan University, Kaifeng, 475004, China
| | - Borui Ma
- The Jointed National Laboratory of Antibody Drug Engineering, The First Affiliated Hospital of Henan University, Henan University, Kaifeng, 475004, China
| | - Yanqun Zhao
- The Jointed National Laboratory of Antibody Drug Engineering, The First Affiliated Hospital of Henan University, Henan University, Kaifeng, 475004, China
| | - Yihua Chen
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, The Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China.
| | - Yanzhong Hu
- The Jointed National Laboratory of Antibody Drug Engineering, The First Affiliated Hospital of Henan University, Henan University, Kaifeng, 475004, China.
| |
Collapse
|
4
|
Mouawad N, Capasso G, Ruggeri E, Martinello L, Severin F, Visentin A, Facco M, Trentin L, Frezzato F. Is It Still Possible to Think about HSP70 as a Therapeutic Target in Onco-Hematological Diseases? Biomolecules 2023; 13:biom13040604. [PMID: 37189352 DOI: 10.3390/biom13040604] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 03/21/2023] [Accepted: 03/26/2023] [Indexed: 03/30/2023] Open
Abstract
The search for molecules to be targeted that are involved in apoptosis resistance/increased survival and pathogenesis of onco-hematological malignancies is ongoing since these diseases are still not completely understood. Over the years, a good candidate has been identified in the Heat Shock Protein of 70kDa (HSP70), a molecule defined as “the most cytoprotective protein ever been described”. HSP70 is induced in response to a wide variety of physiological and environmental insults, allowing cells to survive lethal conditions. This molecular chaperone has been detected and studied in almost all the onco-hematological diseases and is also correlated to poor prognosis and resistance to therapy. In this review, we give an overview of the discoveries that have led us to consider HSP70 as a therapeutic target for mono- or combination-therapies in acute and chronic leukemias, multiple myeloma and different types of lymphomas. In this excursus, we will also consider HSP70 partners, such as its transcription factor HSF1 or its co-chaperones whose druggability could indirectly affect HSP70. Finally, we will try to answer the question asked in the title of this review considering that, despite the effort made by research in this field, HSP70 inhibitors never reached the clinic.
Collapse
|
5
|
Yu W, Zhang H, Yuan Y, Tang J, Chen X, Liu T, Zhao X. Chimeric Antigen Receptor T Cells Targeting Cell Surface GRP78 to Eradicate Acute Myeloid Leukemia. Front Cell Dev Biol 2022; 10:928140. [PMID: 35990606 PMCID: PMC9387679 DOI: 10.3389/fcell.2022.928140] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 05/31/2022] [Indexed: 11/19/2022] Open
Abstract
Acute myeloid leukemia (AML) is a serious, life-threatening hematological malignancy. The treatment outcome of relapsed or refractory AML patients remains dismal, and new treatment options are needed. Chimeric antigen receptor (CAR) T cells have been successful in improving the prognosis for B-lineage acute lymphoblastic leukemia and lymphoma by targeting CD19. However, CAR T-cell therapy for AML is still elusive, owing to the lack of a tumor-specific cell surface antigen and spare hematopoietic stem cells (HSCs). This study generated a novel CAR construction that targets the cell surface protein glucose-regulated protein 78 (GRP78) (csGRP78). We confirmed that GRP78-CAR T cells demonstrate an anti-tumor effect against human AML cells in vitro. In xenograft models, GRP78-CAR T cells effectively eliminate AML cells and protect mice against systemic leukemia, in the meanwhile, prolonging survival. In addition, GRP78-CAR T cells also specifically eradicate the primary AML patient-derived blast. In particular, GRP78-CAR T cells spare normal HSCs, highlighting that GRP78-CAR is a promising approach for the therapy of AML.
Collapse
Affiliation(s)
- Wei Yu
- Laboratory of Animal Tumor Models, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Hang Zhang
- Department of Hematology, Institute of Hematology, West China Hospital of Sichuan University, Chengdu, China
| | - Yuncang Yuan
- Laboratory of Animal Tumor Models, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Jie Tang
- Laboratory of Animal Tumor Models, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Xinchuan Chen
- Department of Hematology, Institute of Hematology, West China Hospital of Sichuan University, Chengdu, China
| | - Ting Liu
- Department of Hematology, Institute of Hematology, West China Hospital of Sichuan University, Chengdu, China
- *Correspondence: Ting Liu, ; Xudong Zhao,
| | - Xudong Zhao
- Laboratory of Animal Tumor Models, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
- *Correspondence: Ting Liu, ; Xudong Zhao,
| |
Collapse
|
6
|
Fabarius A, Samra V, Drews O, Mörz H, Bierbaum M, Darwich A, Weiss C, Brendel S, Kleiner H, Seifarth W, Greffrath W, Hofmann WK, Schmitt CA, Popp HD. Evidence for Recombinant GRP78, CALR, PDIA3 and GPI as Mediators of Genetic Instability in Human CD34+ Cells. Cancers (Basel) 2022; 14:2883. [PMID: 35740549 PMCID: PMC9221337 DOI: 10.3390/cancers14122883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 06/03/2022] [Accepted: 06/08/2022] [Indexed: 12/10/2022] Open
Abstract
Soluble factors released from irradiated human mesenchymal stromal cells (MSC) may induce genetic instability in human CD34+ cells, potentially mediating hematologic disorders. Recently, we identified four key proteins in the secretome of X-ray-irradiated MSC, among them three endoplasmic reticulum proteins, the 78 kDa glucose-related protein (GRP78), calreticulin (CALR), and protein disulfide-isomerase A3 (PDIA3), as well as the glycolytic enzyme glucose-6-phosphate isomerase (GPI). Here, we demonstrate that exposition of CD34+ cells to recombinant GRP78, CALR, PDIA3 and GPI induces substantial genetic instability. Increased numbers of γH2AX foci (p < 0.0001), centrosome anomalies (p = 0.1000) and aberrant metaphases (p = 0.0022) were detected in CD34+ cells upon incubation with these factors. Specifically, γH2AX foci were found to be induced 4−5-fold in response to any individual of the four factors, and centrosome anomalies by 3−4 fold compared to control medium, which contained none of the recombinant proteins. Aberrant metaphases, not seen in the context of control medium, were detected to a similar extent than centrosome anomalies across the four factors. Notably, the strongest effects were observed when all four factors were collectively provided. In summary, our data suggest that specific components of the secretome from irradiated MSC act as mediators of genetic instability in CD34+ cells, thereby possibly contributing to the pathogenesis of radiation-induced hematologic disorders beyond direct radiation-evoked DNA strand breaks.
Collapse
Affiliation(s)
- Alice Fabarius
- Department of Hematology and Oncology, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany; (A.F.); (V.S.); (S.B.); (H.K.); (W.S.); (W.-K.H.)
| | - Vanessa Samra
- Department of Hematology and Oncology, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany; (A.F.); (V.S.); (S.B.); (H.K.); (W.S.); (W.-K.H.)
| | - Oliver Drews
- Biomedical Mass Spectrometry, Center for Medical Research, Johannes Kepler University, 4020 Linz, Austria;
| | - Handan Mörz
- Department of Neurophysiology, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany; (H.M.); (W.G.)
| | - Miriam Bierbaum
- Department of Radiation Oncology, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany;
| | - Ali Darwich
- Department of Orthopedics and Trauma Surgery, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany;
| | - Christel Weiss
- Department of Medical Statistics and Biomathematics, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany;
| | - Susanne Brendel
- Department of Hematology and Oncology, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany; (A.F.); (V.S.); (S.B.); (H.K.); (W.S.); (W.-K.H.)
| | - Helga Kleiner
- Department of Hematology and Oncology, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany; (A.F.); (V.S.); (S.B.); (H.K.); (W.S.); (W.-K.H.)
| | - Wolfgang Seifarth
- Department of Hematology and Oncology, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany; (A.F.); (V.S.); (S.B.); (H.K.); (W.S.); (W.-K.H.)
| | - Wolfgang Greffrath
- Department of Neurophysiology, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany; (H.M.); (W.G.)
| | - Wolf-Karsten Hofmann
- Department of Hematology and Oncology, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany; (A.F.); (V.S.); (S.B.); (H.K.); (W.S.); (W.-K.H.)
| | - Clemens A. Schmitt
- Department of Hematology and Oncology, Kepler University Hospital, Johannes Kepler University, 4020 Linz, Austria;
- Medical Department, Division of Hematology, Oncology and Tumor Immunology, Campus Virchow-Klinikum, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 13353 Berlin, Germany
- Max-Delbrück-Center for Molecular Medicine, Helmholtz Association, 13125 Berlin, Germany
| | - Henning D. Popp
- Department of Hematology and Oncology, Kepler University Hospital, Johannes Kepler University, 4020 Linz, Austria;
| |
Collapse
|
7
|
Tang M, Zheng Y, Li J, Hu Y. The X box binding protein 1/C/EBP homologous protein pathway induces apoptosis of endothelial cells under hyperglycemia. Exp Ther Med 2022; 24:454. [PMID: 35720621 DOI: 10.3892/etm.2022.11381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Accepted: 06/16/2021] [Indexed: 11/06/2022] Open
Affiliation(s)
- Maoshun Tang
- Department of Neurosurgery, Shenzhen Hospital, University of Chinese Academy of Sciences, Shenzhen, Guangdong 518106, P.R. China
| | - Yi Zheng
- Department of Science and Education, Shenzhen Hospital, University of Chinese Academy of Sciences, Shenzhen, Guangdong 518106, P.R. China
| | - Jianping Li
- Department of Cardiology, Shenzhen Hospital, University of Chinese Academy of Sciences, Shenzhen, Guangdong 518106, P.R. China
| | - Yuanlang Hu
- Department of Obstetrics and Gynecology, Shenzhen Hospital, University of Chinese Academy of Sciences, Shenzhen, Guangdong 518106, P.R. China
| |
Collapse
|
8
|
Chen J, Lynn EG, Yousof TR, Sharma H, MacDonald ME, Byun JH, Shayegan B, Austin RC. Scratching the Surface—An Overview of the Roles of Cell Surface GRP78 in Cancer. Biomedicines 2022; 10:biomedicines10051098. [PMID: 35625836 PMCID: PMC9138746 DOI: 10.3390/biomedicines10051098] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 05/01/2022] [Accepted: 05/05/2022] [Indexed: 02/04/2023] Open
Abstract
The 78 kDa glucose-regulated protein (GRP78) is considered an endoplasmic reticulum (ER)-resident molecular chaperone that plays a crucial role in protein folding homeostasis by regulating the unfolded protein response (UPR) and inducing numerous proapoptotic and autophagic pathways within the eukaryotic cell. However, in cancer cells, GRP78 has also been shown to migrate from the ER lumen to the cell surface, playing a role in several cellular pathways that promote tumor growth and cancer cell progression. There is another insidious consequence elicited by cell surface GRP78 (csGRP78) on cancer cells: the accumulation of csGRP78 represents a novel neoantigen leading to the production of anti-GRP78 autoantibodies that can bind csGRP78 and further amplify these cellular pathways to enhance cell growth and mitigate apoptotic cell death. This review examines the current body of literature that delineates the mechanisms by which ER-resident GRP78 localizes to the cell surface and its consequences, as well as potential therapeutics that target csGRP78 and block its interaction with anti-GRP78 autoantibodies, thereby inhibiting further amplification of cancer cell progression.
Collapse
Affiliation(s)
- Jack Chen
- Department of Medicine, Division of Nephrology, St. Joseph′s Healthcare Hamilton, Hamilton Center for Kidney Research, McMaster University, Hamilton, ON L8N 4A6, Canada; (J.C.); (E.G.L.); (T.R.Y.); (H.S.); (M.E.M.); (J.H.B.)
| | - Edward G. Lynn
- Department of Medicine, Division of Nephrology, St. Joseph′s Healthcare Hamilton, Hamilton Center for Kidney Research, McMaster University, Hamilton, ON L8N 4A6, Canada; (J.C.); (E.G.L.); (T.R.Y.); (H.S.); (M.E.M.); (J.H.B.)
| | - Tamana R. Yousof
- Department of Medicine, Division of Nephrology, St. Joseph′s Healthcare Hamilton, Hamilton Center for Kidney Research, McMaster University, Hamilton, ON L8N 4A6, Canada; (J.C.); (E.G.L.); (T.R.Y.); (H.S.); (M.E.M.); (J.H.B.)
| | - Hitesh Sharma
- Department of Medicine, Division of Nephrology, St. Joseph′s Healthcare Hamilton, Hamilton Center for Kidney Research, McMaster University, Hamilton, ON L8N 4A6, Canada; (J.C.); (E.G.L.); (T.R.Y.); (H.S.); (M.E.M.); (J.H.B.)
| | - Melissa E. MacDonald
- Department of Medicine, Division of Nephrology, St. Joseph′s Healthcare Hamilton, Hamilton Center for Kidney Research, McMaster University, Hamilton, ON L8N 4A6, Canada; (J.C.); (E.G.L.); (T.R.Y.); (H.S.); (M.E.M.); (J.H.B.)
| | - Jae Hyun Byun
- Department of Medicine, Division of Nephrology, St. Joseph′s Healthcare Hamilton, Hamilton Center for Kidney Research, McMaster University, Hamilton, ON L8N 4A6, Canada; (J.C.); (E.G.L.); (T.R.Y.); (H.S.); (M.E.M.); (J.H.B.)
| | - Bobby Shayegan
- Department of Surgery, Division of Urology, The Research Institute of St. Joe′s Hamilton, McMaster University, ON L8N 4A6, Canada;
| | - Richard C. Austin
- Department of Medicine, Division of Nephrology, St. Joseph′s Healthcare Hamilton, Hamilton Center for Kidney Research, McMaster University, Hamilton, ON L8N 4A6, Canada; (J.C.); (E.G.L.); (T.R.Y.); (H.S.); (M.E.M.); (J.H.B.)
- Correspondence: ; Tel.: +1-905-522-1155 (ext. 35175)
| |
Collapse
|
9
|
Angeles-Floriano T, Rivera-Torruco G, García-Maldonado P, Juárez E, Gonzalez Y, Parra-Ortega I, Vilchis-Ordoñez A, Lopez-Martinez B, Arriaga-Pizano L, Orozco-Ruíz D, Torres-Nava JR, Licona-Limón P, López-Sosa F, Bremer A, Alvarez-Arellano L, Valle-Rios R. Cell surface expression of GRP78 and CXCR4 is associated with childhood high-risk acute lymphoblastic leukemia at diagnostics. Sci Rep 2022; 12:2322. [PMID: 35149705 PMCID: PMC8837614 DOI: 10.1038/s41598-022-05857-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 01/12/2022] [Indexed: 12/11/2022] Open
Abstract
Acute lymphocytic leukemia is the most common type of cancer in pediatric individuals. Glucose regulated protein (GRP78) is an endoplasmic reticulum chaperone that facilitates the folding and assembly of proteins and regulates the unfolded protein response pathway. GRP78 has a role in survival of cancer and metastasis and cell-surface associated GRP78 (sGRP78) is expressed on cancer cells but not in normal cells. Here, we explored the presence of sGRP78 in pediatric B-ALL at diagnosis and investigated the correlation with bona fide markers of leukemia. By using a combination of flow cytometry and high multidimensional analysis, we found a distinctive cluster containing high levels of sGRP78, CD10, CD19, and CXCR4 in bone marrow samples obtained from High-risk leukemia patients, which was absent in the compartment of Standard-risk leukemia. We confirmed that sGRP78+CXCR4+ blood-derived cells were more frequent in High-risk leukemia patients. Finally, we analyzed the dissemination capacity of sGRP78 leukemia cells in a model of xenotransplantation. sGRP78+ cells emigrated to the bone marrow and lymph nodes, maintaining the expression of CXCR4. Testing the presence of sGRP78 and CXCR4 together with conventional markers may help to achieve a better categorization of High and Standard-risk pediatric leukemia at diagnosis.
Collapse
Affiliation(s)
- Tania Angeles-Floriano
- Unidad Universitaria de Investigación, División de Investigación, Facultad de Medicina, UNAM-Hospital Infantil de México Federico Gómez, Universidad 3000, CP 04510, Mexico City, Mexico
- Programa de Maestría y Doctorado en Ciencias Médicas Odontológicas y de la Salud, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
- Unidad de Investigación en Inmunología y Proteómica, Hospital Infantil de México Federico Gómez, Mexico City, Mexico
| | - Guadalupe Rivera-Torruco
- Unidad de Investigación en Inmunología y Proteómica, Hospital Infantil de México Federico Gómez, Mexico City, Mexico
- Departamento de Fisiología y Neurociencias, Centro de Investigación y de Estudios Avanzados (CINVESTAV), Mexico City, Mexico
| | - Paulina García-Maldonado
- Unidad Universitaria de Investigación, División de Investigación, Facultad de Medicina, UNAM-Hospital Infantil de México Federico Gómez, Universidad 3000, CP 04510, Mexico City, Mexico
| | - Esmeralda Juárez
- Departamento de Investigación en Microbiología, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
| | - Yolanda Gonzalez
- Departamento de Investigación en Microbiología, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
| | - Israel Parra-Ortega
- Subdirección de Diagnóstico clínico y Departamento de Laboratorio Clínico, Hospital Infantil de México Federico Gómez, Mexico City, Mexico
| | - Armando Vilchis-Ordoñez
- Subdirección de Diagnóstico clínico y Departamento de Laboratorio Clínico, Hospital Infantil de México Federico Gómez, Mexico City, Mexico
| | - Briceida Lopez-Martinez
- Subdirección de Diagnóstico clínico y Departamento de Laboratorio Clínico, Hospital Infantil de México Federico Gómez, Mexico City, Mexico
| | - Lourdes Arriaga-Pizano
- Unidad de Investigación Médica en Inmunoquímica, CMN Siglo XXI, IMSS, Mexico City, Mexico
| | | | | | - Paula Licona-Limón
- Departamento de Biología Celular y del Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
| | - Francisco López-Sosa
- Departamento de Ortopedia, Hospital Infantil de México Federico Gómez, Mexico City, Mexico
| | - Alhelí Bremer
- Departamento de Ortopedia, Hospital Infantil de México Federico Gómez, Mexico City, Mexico
| | | | - Ricardo Valle-Rios
- Unidad Universitaria de Investigación, División de Investigación, Facultad de Medicina, UNAM-Hospital Infantil de México Federico Gómez, Universidad 3000, CP 04510, Mexico City, Mexico.
- Unidad de Investigación en Inmunología y Proteómica, Hospital Infantil de México Federico Gómez, Mexico City, Mexico.
| |
Collapse
|
10
|
Hebbar N, Epperly R, Vaidya A, Thanekar U, Moore SE, Umeda M, Ma J, Patil SL, Langfitt D, Huang S, Cheng C, Klco JM, Gottschalk S, Velasquez MP. CAR T cells redirected to cell surface GRP78 display robust anti-acute myeloid leukemia activity and do not target hematopoietic progenitor cells. Nat Commun 2022; 13:587. [PMID: 35102167 PMCID: PMC8803836 DOI: 10.1038/s41467-022-28243-6] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 01/12/2022] [Indexed: 02/06/2023] Open
Abstract
Developing CAR T cells for acute myeloid leukemia (AML) has been hampered by a paucity of targets that are expressed on AML blasts and not on hematopoietic progenitor cells (HPCs). Here we demonstrate that GRP78 is expressed on the cell surface of primary AML blasts but not HPCs. To target GRP78, we generate T cell expressing a GRP78-specific peptide-based CAR, which show evidence of minimal fratricide post activation/transduction and antigen-dependent T cell differentiation. GRP78-CAR T cells recognize and kill GRP78-positive AML cells without toxicity to HPCs. In vivo, GRP78-CAR T cells have significant anti-AML activity. To prevent antigen-dependent T cell differentiation, we block CAR signaling and GRP78 cell surface expression post activation by using dasatinib during GRP78-CAR T cell manufacturing. This significantly improves their effector function in vitro and in vivo. Thus, targeting cell surface GRP78-positive AML with CAR T cells is feasible, and warrants further active exploration.
Collapse
MESH Headings
- Animals
- Cell Line, Tumor
- Cell Membrane/drug effects
- Cell Membrane/metabolism
- Cell Survival/drug effects
- Cytokines/metabolism
- Cytotoxicity, Immunologic/drug effects
- Dasatinib/pharmacology
- Endoplasmic Reticulum Chaperone BiP/immunology
- Gene Expression Regulation, Leukemic/drug effects
- Hematopoietic Stem Cells/immunology
- Humans
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/immunology
- Mice, Inbred NOD
- Mice, SCID
- Receptors, Chimeric Antigen/immunology
- T-Lymphocytes/drug effects
- T-Lymphocytes/immunology
- Xenograft Model Antitumor Assays
- Mice
Collapse
Affiliation(s)
- Nikhil Hebbar
- Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Rebecca Epperly
- Department of Oncology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Abishek Vaidya
- Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Unmesha Thanekar
- Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Sarah E Moore
- Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Masayuki Umeda
- Department of Pathology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Jing Ma
- Department of Pathology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Sagar L Patil
- Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Deanna Langfitt
- Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Sujuan Huang
- Department of Biostatistics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Cheng Cheng
- Department of Biostatistics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Jeffery M Klco
- Department of Pathology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Stephen Gottschalk
- Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - M Paulina Velasquez
- Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA.
| |
Collapse
|
11
|
Kabakov AE, Gabai VL. HSP70s in Breast Cancer: Promoters of Tumorigenesis and Potential Targets/Tools for Therapy. Cells 2021; 10:cells10123446. [PMID: 34943954 PMCID: PMC8700403 DOI: 10.3390/cells10123446] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 11/25/2021] [Accepted: 12/03/2021] [Indexed: 12/20/2022] Open
Abstract
The high frequency of breast cancer worldwide and the high mortality among women with this malignancy are a serious challenge for modern medicine. A deeper understanding of the mechanisms of carcinogenesis and emergence of metastatic, therapy-resistant breast cancers would help development of novel approaches to better treatment of this disease. The review is dedicated to the role of members of the heat shock protein 70 subfamily (HSP70s or HSPA), mainly inducible HSP70, glucose-regulated protein 78 (GRP78 or HSPA5) and GRP75 (HSPA9 or mortalin), in the development and pathogenesis of breast cancer. Various HSP70-mediated cellular mechanisms and pathways which contribute to the oncogenic transformation of mammary gland epithelium are reviewed, as well as their role in the development of human breast carcinomas with invasive, metastatic traits along with the resistance to host immunity and conventional therapeutics. Additionally, intracellular and cell surface HSP70s are considered as potential targets for therapy or sensitization of breast cancer. We also discuss a clinical implication of Hsp70s and approaches to targeting breast cancer with gene vectors or nanoparticles downregulating HSP70s, natural or synthetic (small molecule) inhibitors of HSP70s, HSP70-binding antibodies, HSP70-derived peptides, and HSP70-based vaccines.
Collapse
Affiliation(s)
- Alexander E. Kabakov
- Department of Radiation Biochemistry, A. Tsyb Medical Radiological Research Center—Branch of the National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, Koroleva 4, 249036 Obninsk, Russia;
| | - Vladimir L. Gabai
- CureLab Oncology Inc., Dedham, MA 02026, USA
- Correspondence: ; Tel.: +1-617-319-7314
| |
Collapse
|
12
|
Elfiky AA, Baghdady AM, Ali SA, Ahmed MI. GRP78 targeting: Hitting two birds with a stone. Life Sci 2020; 260:118317. [PMID: 32841659 PMCID: PMC7442953 DOI: 10.1016/j.lfs.2020.118317] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 07/22/2020] [Accepted: 08/19/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND Glucose regulating protein 78 (GRP78) is one member of the Heat Shock Protein family of chaperone proteins (HSPA5) found in eukaryotes. It acts as the master of the Unfolded Protein Response (UPR) process in the lumen of the Endoplasmic Reticulum (ER). SCOPE Under the stress of unfolded proteins, GRP78 binds to the unfolded proteins to prevent misfolding, while under the load of the unfolded protein, it drives the cell to autophagy or apoptosis. Several attempts reported the overexpression of GRP78 on the cell membrane of cancer cells and cells infected with viruses or fungi. MAJOR CONCLUSIONS Cell-surface GRP78 is used as a cancer cell target in previous studies. Additionally, GRP78 is used as a drug target to stop the progression of cancer cells by different compounds, including peptides, antibodies, and some natural compounds. Additionally, it can be used as a protein target to reduce the infectivity of different viruses, including the pandemic SARS-CoV-2. Besides, GRP78 targeting is used in diagnosis and imaging modalities using radionuclides. GENERAL SIGNIFICANCE This review summarizes the various attempts that used GRP78 both in therapy (fighting cancer, viral and fungal infections) and diagnosis (imaging).
Collapse
|
13
|
Ninkovic S, Harrison SJ, Quach H. Glucose-regulated protein 78 (GRP78) as a potential novel biomarker and therapeutic target in multiple myeloma. Expert Rev Hematol 2020; 13:1201-1210. [PMID: 32990063 DOI: 10.1080/17474086.2020.1830372] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
INTRODUCTION Glucose-regulated protein 78 (GRP78) is a stress-inducible molecular chaperone expressed within the endoplasmic reticulum where it acts as a master regulator of the unfolded protein response (UPR) pathway. At times of ER stress, activation of the UPR, a multimolecular pathway, limits proteotoxicity induced by misfolded proteins. In malignancies, including multiple myeloma which is characterized by an accumulation of misfolded immunoglobulins, GRP78 expression is increased, with notable translocation of GRP78 to the cell surface. Studies suggest cell-surface GRP78 (csGRP78) to be of prognostic significance with emerging evidence that it interacts with a myriad of co-ligands to activate signaling pathways promoting cell proliferation and survival or apoptosis. AREAS COVERED This review focuses on the role of ER and csGRP78 in physiology and oncogenesis in multiple myeloma, addressing factors that shift the balance in GRP78 signaling from survival to apoptosis. The role of GRP78 as a potential prognostic biomarker is explored and current therapeutics in development aimed at targeting csGRP78 are addressed. We conducted a PubMed literature search using the keywords 'GRP78,' 'multiple myeloma' reviewing studies prior to 2020. EXPERT OPINION Cell-surface GRP78 expression is a potential novel prognostic biomarker in myeloma and targeting of csGRP78 is promising and requires further investigation.
Collapse
Affiliation(s)
- Slavisa Ninkovic
- Department of Haematology, St. Vincent's Hospital Melbourne , Fitzroy, Australia.,Department of Medicine, University of Melbourne , Fitzroy, Australia
| | - Simon J Harrison
- Clinical Haematology, Peter MacCallum Cancer Centre and Royal Melbourne Hospital , Melbourne, Australia.,Sir Peter MacCallum Dept of Oncology, University of Melbourne , Parkville, Australia
| | - Hang Quach
- Department of Haematology, St. Vincent's Hospital Melbourne , Fitzroy, Australia.,Department of Medicine, University of Melbourne , Fitzroy, Australia
| |
Collapse
|
14
|
Gopal U, Pizzo SV. Cell surface GRP78 signaling: An emerging role as a transcriptional modulator in cancer. J Cell Physiol 2020; 236:2352-2363. [PMID: 32864780 DOI: 10.1002/jcp.30030] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 08/07/2020] [Accepted: 08/17/2020] [Indexed: 12/14/2022]
Abstract
Cancer cells acquire dysregulated gene expression to establish specific transcriptional dependencies and their underlying mechanisms that are ultimately responsible for this addictions have not been fully elucidated. Glucose-regulated protein 78 (GRP78) is a stress-inducible, multifunctional, prosurvival, endoplasmic reticulum chaperone in the heat shock protein 70 family. Expression of cell surface GRP78 (CS-GRP78) is associated with increased malignant behavior and resistance to chemotherapy and radiotherapy by endowing various cancer cells with increased proliferative ability, altered metabolism, improved survival, and augmented invasive and metastatic potential. Emerging evidence has highlighted an unusual role of CS-GRP78 in regulating transcription factors (TFs) by mediating various signaling pathways involved in malignant transformation, metabolic reprogramming, and tumor progression. During the last decade, we targeted CS-GRP78 with C38 monoclonal antibody (C38 Mab) in numerous studies, which have highlighted the epigenetic interplay between CS-GRP78 and various TFs including c-MYC, Yes-associated protein/transcriptional coactivator with PDZ-binding motif, c-Fos, and histone acetylation to potentiate subsequent modulation of tumorigenesis, invasion, and metastasis. Here, we summarize the current state of knowledge about the role of CS-GRP78 in cancer development and progression, including epigenetic regulation and sheds light on CS-GRP78 as vulnerable target for cancer therapy. Overall, this review focuses on the mechanisms of TFs that are behind the transcriptional dysregulation in cancer and lays the groundwork for rational therapeutic use of C38 Mab based on CS-GRP78 biology.
Collapse
Affiliation(s)
- Udhayakumar Gopal
- Department of Pathology, Duke University Medical Center, Durham, North Carolina
| | - Salvatore V Pizzo
- Department of Pathology, Duke University Medical Center, Durham, North Carolina
| |
Collapse
|
15
|
Khateb A, Ronai ZA. Unfolded Protein Response in Leukemia: From Basic Understanding to Therapeutic Opportunities. Trends Cancer 2020; 6:960-973. [PMID: 32540455 DOI: 10.1016/j.trecan.2020.05.012] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 05/03/2020] [Accepted: 05/19/2020] [Indexed: 12/11/2022]
Abstract
Understanding genetic and epigenetic changes that underlie abnormal proliferation of hematopoietic stem and progenitor cells is critical for development of new approaches to monitor and treat leukemia. The unfolded protein response (UPR) is a conserved adaptive signaling pathway that governs protein folding, secretion, and energy production and serves to maintain protein homeostasis in various cellular compartments. Deregulated UPR signaling, which often occurs in hematopoietic stem cells and leukemia, defines the degree of cellular toxicity and perturbs protein homeostasis, and at the same time, offers a novel therapeutic target. Here, we review current knowledge related to altered UPR signaling in leukemia and highlight possible strategies for exploiting the UPR as treatment for this disease.
Collapse
Affiliation(s)
- Ali Khateb
- Tumor Initiation and Maintenance Program, National Cancer Institute (NCI) Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Ze'ev A Ronai
- Tumor Initiation and Maintenance Program, National Cancer Institute (NCI) Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA.
| |
Collapse
|
16
|
Cell Surface GRP78 as a Death Receptor and an Anticancer Drug Target. Cancers (Basel) 2019; 11:cancers11111787. [PMID: 31766302 PMCID: PMC6896222 DOI: 10.3390/cancers11111787] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 10/31/2019] [Accepted: 11/08/2019] [Indexed: 12/16/2022] Open
Abstract
Cell surface GRP78 (csGRP78, glucose-regulated protein 78 kDa) is preferentially overexpressed in aggressive, metastatic, and chemo-resistant cancers. GRP78 is best studied as a chaperone protein in the lumen of endoplasmic reticulum (ER), facilitating folding and secretion of the newly synthesized proteins and regulating protein degradation as an ER stress sensor in the unfolded protein pathway. As a cell surface signal receptor, multiple csGRP78 ligands have been discovered to date, and they trigger various downstream cell signaling pathways including pro-proliferative, pro-survival, and pro-apoptotic pathways. In this perspective, we evaluate csGRP78 as a cell surface death receptor and its prospect as an anticancer drug target. The pro-apoptotic ligands of csGRP78 discovered so far include natural proteins, monoclonal antibodies, and synthetic peptides. Even the secreted GRP78 itself was recently found to function as a pro-apoptotic ligand for csGRP78, mediating pancreatic β-cell death. As csGRP78 is found to mainly configur as an external peripheral protein on cancer cell surface, how it can transmit death signals to the cytoplasmic environment remains enigmatic. With the recent encouraging results from the natural csGRP78 targeting pro-apoptotic monoclonal antibody PAT-SM6 in early-stage cancer clinical trials, the potential to develop a novel class of anticancer therapeutics targeting csGRP78 is becoming more compelling.
Collapse
|
17
|
Dual role of Endoplasmic Reticulum Stress-Mediated Unfolded Protein Response Signaling Pathway in Carcinogenesis. Int J Mol Sci 2019; 20:ijms20184354. [PMID: 31491919 PMCID: PMC6770252 DOI: 10.3390/ijms20184354] [Citation(s) in RCA: 109] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 08/30/2019] [Accepted: 09/03/2019] [Indexed: 12/24/2022] Open
Abstract
Cancer constitutes a grave problem nowadays in view of the fact that it has become one of the main causes of death worldwide. Poor clinical prognosis is presumably due to cancer cells metabolism as tumor microenvironment is affected by oxidative stress. This event triggers adequate cellular response and thereby creates appropriate conditions for further cancer progression. Endoplasmic reticulum (ER) stress occurs when the balance between an ability of the ER to fold and transfer proteins and the degradation of the misfolded ones become distorted. Since ER is an organelle relatively sensitive to oxidative damage, aforementioned conditions swiftly cause the activation of the unfolded protein response (UPR) signaling pathway. The output of the UPR, depending on numerous factors, may vary and switch between the pro-survival and the pro-apoptotic branch, and hence it displays opposing effects in deciding the fate of the cancer cell. The role of UPR-related proteins in tumorigenesis, such as binding the immunoglobulin protein (BiP) and inositol-requiring enzyme-1α (IRE1α), activating transcription factor 6 (ATF6) or the protein kinase R (PKR)-like endoplasmic reticulum kinase (PERK), has already been specifically described so far. Nevertheless, due to the paradoxical outcomes of the UPR activation as well as gaps in current knowledge, it still needs to be further investigated. Herein we would like to elicit the actual link between neoplastic diseases and the UPR signaling pathway, considering its major branches and discussing its potential use in the development of a novel, anti-cancer, targeted therapy.
Collapse
|
18
|
Bailly C, Waring MJ. Pharmacological effectors of GRP78 chaperone in cancers. Biochem Pharmacol 2019; 163:269-278. [PMID: 30831072 DOI: 10.1016/j.bcp.2019.02.038] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Accepted: 02/28/2019] [Indexed: 12/21/2022]
Abstract
The protein chaperone GRP78 is a master regulator of endoplasmic reticulum (ER) functions and is frequently over-expressed at the surface of cancer cells where it contributes to chemo-resistance. It represents a well-studied ER stress marker but an under-explored target for new drug development. This review aims to untangle the structural and functional diversity of GRP78 modulators, covering over 130 natural products, synthetic molecules, specific peptides and monoclonal antibodies that target GRP78. Several approaches to promote or to incapacitate GRP78 are presented, including the use of oligonucleotides and specific cell-delivery peptides often conjugated to cytotoxic payloads to design GRP78-targeted therapeutics. A repertoire of drugs that turn on/off GRP78 is exposed, including molecules which bind directly to GRP78, principally to its ATP site. There exist many options to regulate positively or negatively the expression of the chaperone, or to interfere with its cellular trafficking. This review provides a molecular cartography of GRP78 pharmacological effectors and adds weight to the notion that GRP78 repressors could represent promising anticancer therapeutics, notably as regards limiting chemo-resistance of cancer cells. The potential of GRP78-targeting drugs in other therapeutic modalities is also evoked.
Collapse
Affiliation(s)
- Christian Bailly
- UMR-S 1172, Centre de Recherche Jean-Pierre Aubert, INSERM, University of Lille, CHU Lille, 59045 Lille, France.
| | - Michael J Waring
- Department of Biochemistry, University of Cambridge, Cambridge CB2 1GA, UK
| |
Collapse
|
19
|
Araujo N, Hebbar N, Rangnekar VM. GRP78 Is a Targetable Receptor on Cancer and Stromal Cells. EBioMedicine 2018; 33:2-3. [PMID: 29997052 PMCID: PMC6085536 DOI: 10.1016/j.ebiom.2018.06.030] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 06/25/2018] [Indexed: 01/09/2023] Open
Affiliation(s)
- Nathalia Araujo
- Departments of Toxicology and Cancer Biology, University of Kentucky, United States
| | - Nikhil Hebbar
- St. Jude Children's Research Hospital, Memphis, TN, United States
| | - Vivek M Rangnekar
- Departments of Toxicology and Cancer Biology, University of Kentucky, United States; Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky, United States; Department of Radiation Medicine, University of Kentucky, United States; Markey Cancer Center, University of Kentucky, United States.
| |
Collapse
|
20
|
Kao C, Chandna R, Ghode A, Dsouza C, Chen M, Larsson A, Lim SH, Wang M, Cao Z, Zhu Y, Anand GS, Ge R. Proapoptotic Cyclic Peptide BC71 Targets Cell-Surface GRP78 and Functions as an Anticancer Therapeutic in Mice. EBioMedicine 2018; 33:22-32. [PMID: 29907328 PMCID: PMC6085501 DOI: 10.1016/j.ebiom.2018.06.004] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Revised: 05/25/2018] [Accepted: 06/06/2018] [Indexed: 02/08/2023] Open
Abstract
Glucose regulated protein 78 kDa (GRP78) is a recently emerged target for cancer therapy and a biomarker for cancer prognosis. Overexpression of GRP78 is observed in many types of cancers, with the cell-surface GRP78 being preferentially present in cancer cells and cancer blood vessel endothelial cells. Isthmin (ISM) is a secreted high-affinity proapoptotic protein ligand of cell-surface GRP78 that suppresses angiogenesis and tumor growth in mice. The C-terminal AMOP (adhesion-associated domain in MUC4 and other proteins) domain of ISM is critical in mediating its interaction with human umbilical vein endothelial cells (HUVECs). In this work, we report novel cyclic peptides harboring the RKD motif in the ISM AMOP domain that function as proapoptotic ligands of cell-surface GRP78. The most potent peptide, BC71, binds to GRP78 and converge to tumor in mice. Intravenous administration of BC71 suppressed xenograft tumor growth in mice as a single agent, with significant reduction in tumor angiogenesis and upsurge in apoptosis. Fluorescent-labeled BC71 accumulates in tumor in mice by targeting cell-surface GRP78. We show that BC71 triggers apoptosis via cell-surface GRP78 and activates caspase-8 and p53 signaling pathways in HUVECs. Using amide hydrogen-deuterium exchange mass spectrometry (HDXMS), we identified that BC71 preferentially binds to ATP-bound GRP78 via amino acid residues 244-257 of GRP78. Hence, BC71 serves as a valuable prototype for further development of peptidomimetic anticancer drugs targeting cell-surface GRP78 as well as PET imaging agents for cancer prognosis.
Collapse
Affiliation(s)
- Chieh Kao
- Department of Biological Sciences, National University of Singapore, 16 Science Drive 4, 117558, Singapore
| | - Ritu Chandna
- Department of Biological Sciences, National University of Singapore, 16 Science Drive 4, 117558, Singapore
| | - Abhijeet Ghode
- Department of Biological Sciences, National University of Singapore, 16 Science Drive 4, 117558, Singapore
| | - Charlotte Dsouza
- Department of Biological Sciences, National University of Singapore, 16 Science Drive 4, 117558, Singapore
| | - Mo Chen
- Department of Biological Sciences, National University of Singapore, 16 Science Drive 4, 117558, Singapore
| | - Andreas Larsson
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, 639798, Singapore
| | - Siau Hoi Lim
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, 639798, Singapore
| | - Minjun Wang
- School of Pharmacy, Fudan University, 826 Zhangheng Rd, Shanghai 201203, China
| | - Zhonglian Cao
- School of Pharmacy, Fudan University, 826 Zhangheng Rd, Shanghai 201203, China
| | - Yizhun Zhu
- School of Pharmacy, Fudan University, 826 Zhangheng Rd, Shanghai 201203, China
| | - Ganesh S Anand
- Department of Biological Sciences, National University of Singapore, 16 Science Drive 4, 117558, Singapore
| | - Ruowen Ge
- Department of Biological Sciences, National University of Singapore, 16 Science Drive 4, 117558, Singapore.
| |
Collapse
|
21
|
Al-Hashimi AA, Lebeau P, Majeed F, Polena E, Lhotak Š, Collins CAF, Pinthus JH, Gonzalez-Gronow M, Hoogenes J, Pizzo SV, Crowther M, Kapoor A, Rak J, Gyulay G, D'Angelo S, Marchiò S, Pasqualini R, Arap W, Shayegan B, Austin RC. Autoantibodies against the cell surface-associated chaperone GRP78 stimulate tumor growth via tissue factor. J Biol Chem 2017; 292:21180-21192. [PMID: 29066620 PMCID: PMC5743090 DOI: 10.1074/jbc.m117.799908] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 09/05/2017] [Indexed: 12/24/2022] Open
Abstract
Tumor cells display on their surface several molecular chaperones that normally reside in the endoplasmic reticulum. Because this display is unique to cancer cells, these chaperones are attractive targets for drug development. Previous epitope-mapping of autoantibodies (AutoAbs) from prostate cancer patients identified the 78-kDa glucose-regulated protein (GRP78) as one such target. Although we previously showed that anti-GRP78 AutoAbs increase tissue factor (TF) procoagulant activity on the surface of tumor cells, the direct effect of TF activation on tumor growth was not examined. In this study, we explore the interplay between the AutoAbs against cell surface-associated GRP78, TF expression/activity, and prostate cancer progression. First, we show that tumor GRP78 expression correlates with disease stage and that anti-GRP78 AutoAb levels parallel prostate-specific antigen concentrations in patient-derived serum samples. Second, we demonstrate that these anti-GRP78 AutoAbs target cell-surface GRP78, activating the unfolded protein response and inducing tumor cell proliferation through a TF-dependent mechanism, a specific effect reversed by neutralization or immunodepletion of the AutoAb pool. Finally, these AutoAbs enhance tumor growth in mice bearing human prostate cancer xenografts, and heparin derivatives specifically abrogate this effect by blocking AutoAb binding to cell-surface GRP78 and decreasing TF expression/activity. Together, these results establish a molecular mechanism in which AutoAbs against cell-surface GRP78 drive TF-mediated tumor progression in an experimental model of prostate cancer. Heparin derivatives counteract this mechanism and, as such, represent potentially appealing compounds to be evaluated in well-designed translational clinical trials.
Collapse
Affiliation(s)
- Ali A Al-Hashimi
- From the Department of Medicine, McMaster University and St. Joseph's Healthcare Hamilton, Hamilton, Ontario L8N 4A6, Canada
- the Department of Surgery, McMaster University and St. Joseph's Healthcare Hamilton, Hamilton, Ontario L8N 4A6, Canada
| | - Paul Lebeau
- From the Department of Medicine, McMaster University and St. Joseph's Healthcare Hamilton, Hamilton, Ontario L8N 4A6, Canada
| | - Fadwa Majeed
- From the Department of Medicine, McMaster University and St. Joseph's Healthcare Hamilton, Hamilton, Ontario L8N 4A6, Canada
| | - Enio Polena
- From the Department of Medicine, McMaster University and St. Joseph's Healthcare Hamilton, Hamilton, Ontario L8N 4A6, Canada
| | - Šárka Lhotak
- From the Department of Medicine, McMaster University and St. Joseph's Healthcare Hamilton, Hamilton, Ontario L8N 4A6, Canada
| | - Celeste A F Collins
- From the Department of Medicine, McMaster University and St. Joseph's Healthcare Hamilton, Hamilton, Ontario L8N 4A6, Canada
| | - Jehonathan H Pinthus
- From the Department of Medicine, McMaster University and St. Joseph's Healthcare Hamilton, Hamilton, Ontario L8N 4A6, Canada
- the Department of Surgery, McMaster University and St. Joseph's Healthcare Hamilton, Hamilton, Ontario L8N 4A6, Canada
| | - Mario Gonzalez-Gronow
- the Department of Pathology, Duke University Medical Center, Durham, North Carolina 27710
| | - Jen Hoogenes
- From the Department of Medicine, McMaster University and St. Joseph's Healthcare Hamilton, Hamilton, Ontario L8N 4A6, Canada
- the Department of Surgery, McMaster University and St. Joseph's Healthcare Hamilton, Hamilton, Ontario L8N 4A6, Canada
| | - Salvatore V Pizzo
- the Department of Pathology, Duke University Medical Center, Durham, North Carolina 27710
| | - Mark Crowther
- From the Department of Medicine, McMaster University and St. Joseph's Healthcare Hamilton, Hamilton, Ontario L8N 4A6, Canada
| | - Anil Kapoor
- From the Department of Medicine, McMaster University and St. Joseph's Healthcare Hamilton, Hamilton, Ontario L8N 4A6, Canada
- the Department of Surgery, McMaster University and St. Joseph's Healthcare Hamilton, Hamilton, Ontario L8N 4A6, Canada
| | - Janusz Rak
- the Department of Pediatrics, Division of Experimental Medicine, Faculty of Medicine, McGill University, Montreal, Quebec H3A 0G4, Canada
| | - Gabriel Gyulay
- From the Department of Medicine, McMaster University and St. Joseph's Healthcare Hamilton, Hamilton, Ontario L8N 4A6, Canada
| | - Sara D'Angelo
- the University of New Mexico Comprehensive Cancer Center, Albuquerque, New Mexico 87106
- the Divisions of Molecular Medicine and
| | - Serena Marchiò
- the University of New Mexico Comprehensive Cancer Center, Albuquerque, New Mexico 87106
- the Divisions of Molecular Medicine and
- the Department of Oncology, University of Turin, 10124 Turin, Italy, and
- the Candiolo Cancer Institute-Fondazione del Piemonte per l'Oncologia (FPO)-Istituto di Ricerca e Cura a Carattere Scientifico (IRCCS), 10060 Candiolo, Italy
| | - Renata Pasqualini
- the University of New Mexico Comprehensive Cancer Center, Albuquerque, New Mexico 87106
- the Divisions of Molecular Medicine and
| | - Wadih Arap
- the University of New Mexico Comprehensive Cancer Center, Albuquerque, New Mexico 87106
- Hematology/Oncology, Department of Internal Medicine, University of New Mexico School of Medicine, Albuquerque, New Mexico 87131
| | - Bobby Shayegan
- From the Department of Medicine, McMaster University and St. Joseph's Healthcare Hamilton, Hamilton, Ontario L8N 4A6, Canada
- the Department of Surgery, McMaster University and St. Joseph's Healthcare Hamilton, Hamilton, Ontario L8N 4A6, Canada
| | - Richard C Austin
- From the Department of Medicine, McMaster University and St. Joseph's Healthcare Hamilton, Hamilton, Ontario L8N 4A6, Canada,
| |
Collapse
|