1
|
Almawash S. Revolutionary Cancer Therapy for Personalization and Improved Efficacy: Strategies to Overcome Resistance to Immune Checkpoint Inhibitor Therapy. Cancers (Basel) 2025; 17:880. [PMID: 40075727 PMCID: PMC11899125 DOI: 10.3390/cancers17050880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 02/09/2025] [Accepted: 02/12/2025] [Indexed: 03/14/2025] Open
Abstract
Cancer remains a significant public health issue worldwide, standing as a primary contributor to global mortality, accounting for approximately 10 million fatalities in 2020 [...].
Collapse
Affiliation(s)
- Saud Almawash
- Department of Pharmaceutics, College of Pharmacy, Shaqra University, Shaqra 11961, Saudi Arabia
| |
Collapse
|
2
|
Song Z, Tao Y, You J. The potential applications of peptide-loading complex in cancer treatment. Front Immunol 2025; 16:1526137. [PMID: 40098955 PMCID: PMC11911339 DOI: 10.3389/fimmu.2025.1526137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Accepted: 02/05/2025] [Indexed: 03/19/2025] Open
Abstract
Immunotherapy for cancer has made significant strides in the last several years. The prognosis for cancer patients has significantly improved as a result, particularly in hematological diseases. However, it was discovered that translating these achievements to solid tumors proved challenging. The peptide-loading complex (PLC), a temporary multisubunit membrane assembly in the endoplasmic reticulum (ER), is crucial for initiating a hierarchical immune response. Chaperones calreticulin and tapasin make up the PLC, unique to class I glycoproteins, thiooxido-reductase ERp57, and a transporter associated with antigen processing. The loading and editing of major histocompatibility complex class I (MHC-I) molecules with peptide translocation into the ER are synchronized by the PLC. One of the immune escape strategies revealed for tumors so far is changes in the expression of MHC molecules. This is because MHC antigens are crucial in presenting antigens to T-lymphocytes and controlling NK cell activity. Furthermore, decreased MHC-I expression has been linked to malignancies resistant to T-cell-based cancer immunotherapies (adoptive transfer of antitumor CD8 T-cells or checkpoint inhibition). The PLC is essential for T-cell priming, differentiation, and tumor growth control because it can bind to a wide range of MHC-I allomorphs. In this review, we have looked into PLC's function and effects in all forms of cancer to improve cancer therapy techniques.
Collapse
Affiliation(s)
- Zhidu Song
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, China
| | - Ying Tao
- Department of Anesthesiology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Jiaxin You
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
3
|
Okura GC, Bharadwaj AG, Waisman DM. Calreticulin-From the Endoplasmic Reticulum to the Plasma Membrane-Adventures of a Wandering Protein. Cancers (Basel) 2025; 17:288. [PMID: 39858072 PMCID: PMC11764459 DOI: 10.3390/cancers17020288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 01/11/2025] [Accepted: 01/14/2025] [Indexed: 01/27/2025] Open
Abstract
Calreticulin (CRT) is a 46 kDa highly conserved protein initially identified as calregulin, a prominent Ca2+-binding protein of the endoplasmic reticulum (ER). Subsequent studies have established that CRT functions in the ER's protein folding response and Ca2+ homeostatic mechanisms. An ER retention signal on the carboxyl terminus of CRT suggested that CRT was restricted to the ER. However, the identification of CRT in the nucleus and cytosol has established that CRT is a multi-compartmental, multifunctional protein. CRT also plays an important role in cancer progression. Most recently, CRT was identified on the cell surface and shown to be a potent 'eat-me' signal that plays a key role in the uptake of apoptotic and viable cancer cells by phagocytes. Elevated CRT exposure on the outer leaflet of cancer cells has been linked with anticancer immunity and superior therapeutic outcomes in patients with non-small cell lung carcinoma, colorectal carcinoma, acute myeloid leukemia, ovarian cancer, and high-grade serous carcinomas. Mutations in the CRT gene have been identified in a subset of patients with myeloproliferative neoplasms. The most recent studies from our laboratory have revealed a new and significant function for extracellular CRT as a plasminogen receptor. This discovery has profound implications for our understanding of the role of CRT in myeloproliferative neoplasms, specifically, essential thrombocythemia.
Collapse
Affiliation(s)
- Gillian C. Okura
- Department of Pathology, Dalhousie University, Halifax, NS B3H 1X5, Canada; (G.C.O.); (A.G.B.)
| | - Alamelu G. Bharadwaj
- Department of Pathology, Dalhousie University, Halifax, NS B3H 1X5, Canada; (G.C.O.); (A.G.B.)
| | - David M. Waisman
- Department of Pathology, Dalhousie University, Halifax, NS B3H 1X5, Canada; (G.C.O.); (A.G.B.)
- Department of Biochemistry and Molecular Biology, Dalhousie University, Halifax, NS B3H 1X5, Canada
| |
Collapse
|
4
|
Wang CY, Lin SC, Chang KJ, Cheong HP, Wu SR, Lee CH, Chuang MW, Chiou SH, Hsu CH, Ko PS. Immunoediting in acute myeloid leukemia: Reappraising T cell exhaustion and the aberrant antigen processing machinery in leukemogenesis. Heliyon 2024; 10:e39731. [PMID: 39568858 PMCID: PMC11577197 DOI: 10.1016/j.heliyon.2024.e39731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Revised: 10/16/2024] [Accepted: 10/22/2024] [Indexed: 11/22/2024] Open
Abstract
Acute myeloid leukemia (AML) establishes an immunosuppressive microenvironment that favors leukemic proliferation. The immune-suppressive cytokines altered antigen processing, and presentation collectively assist AML cells in escaping cytotoxic T-cell surveillance. These CD8+ T cell dysfunction features are emerging therapeutic targets in relapsed/refractory AML patients. Besides, CD8+ T cell exhaustion is a hotspot in recent clinical oncology studies, but its pathophysiology has yet to be elucidated in AML. In this review, we summarize high-quality original studies encompassing the phenotypic and genomic characteristics of T cell exhaustion events in the leukemia progression, emphasize the surface immuno-peptidome that dynamically tunes the fate of T cells to function or dysfunction states, and revisit the biochemical and biophysical properties of type 1 MHC antigen processing mechanism (APM) that pivots in the phenomenon of leukemia antigen dampening.
Collapse
Affiliation(s)
- Ching-Yun Wang
- Department of Medical Education, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Shiuan-Chen Lin
- School of Medicine, National Yang-Ming Chiao Tung University, Taipei, Taiwan
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Kao-Jung Chang
- School of Medicine, National Yang-Ming Chiao Tung University, Taipei, Taiwan
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan
- Institute of Clinical Medicine, National Yang-Ming Chiao Tung University, Taipei, Taiwan
- Department of Ophthalmology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Han-Ping Cheong
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan
- Institute of Pharmacology, National Yang-Ming Chiao Tung University, Taipei, Taiwan
| | - Sin-Rong Wu
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Cheng-Hao Lee
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Ming-Wei Chuang
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Shih-Hwa Chiou
- School of Medicine, National Yang-Ming Chiao Tung University, Taipei, Taiwan
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan
- Institute of Clinical Medicine, National Yang-Ming Chiao Tung University, Taipei, Taiwan
- Department of Ophthalmology, Taipei Veterans General Hospital, Taipei, Taiwan
- Institute of Pharmacology, National Yang-Ming Chiao Tung University, Taipei, Taiwan
- Genomic Research Center, Academia Sinica, Taipei, Taiwan
| | - Chih-Hung Hsu
- Department of Environmental Medicine, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Po-Shen Ko
- School of Medicine, National Yang-Ming Chiao Tung University, Taipei, Taiwan
- Division of Hematology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| |
Collapse
|
5
|
Guijarro-Hernández A, Hurtado C, Urizar-Compains E, Ezcurra B, Galiana-Sáenz A, Baquero E, Cabello J, Vizmanos JL. Myeloproliferative Neoplasm-like Mutations of Calreticulin Induce Phenotypes Associated with Calreticulin Dysfunction in C. elegans. Int J Mol Sci 2024; 25:11606. [PMID: 39519157 PMCID: PMC11546369 DOI: 10.3390/ijms252111606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 10/22/2024] [Accepted: 10/25/2024] [Indexed: 11/16/2024] Open
Abstract
In previous research, we created a C. elegans model with homozygous mutations in calreticulin similar to those found in patients with essential thrombocythemia (ET) and primary myelofibrosis (PMF), two myeloproliferative neoplasms (MPNs). This model, lacking JAK orthologs, enabled us to examine the transcriptomic effects caused by mutant calreticulin without the influence of JAK/STAT activation, the primary pathogenic mechanism associated with calreticulin mutations known to date. Most of the gene expression changes observed seemed to be due to a partial loss of protein function, with the alteration of the extracellular matrix being particularly notable. In this study, our aim was to determine whether this model exhibited any phenotype related to these transcriptomic alterations. The results demonstrate that these strains exhibit multiple phenotypes related to the alteration of the extracellular matrix, fat levels, and fertility, which could be a possible consequence of a partial loss of calreticulin function. These phenotypes resemble some of the clinical and molecular characteristics described in patients with MPNs, but they had never before been linked to a loss of protein function in humans. Thus, these results collectively suggest that CALR mutations could have significant effects on MPNs due to loss of protein function. Delving deeper into these effects to develop innovative therapies for these patients offers considerable potential and interest, given that targeted therapies for these patients have not yielded very promising results so far.
Collapse
Affiliation(s)
- Ana Guijarro-Hernández
- Department of Biochemistry and Genetics, School of Sciences, University of Navarra, 31008 Pamplona, Spain; (A.G.-H.); (C.H.); (E.U.-C.); (A.G.-S.)
| | - Cristina Hurtado
- Department of Biochemistry and Genetics, School of Sciences, University of Navarra, 31008 Pamplona, Spain; (A.G.-H.); (C.H.); (E.U.-C.); (A.G.-S.)
| | - Estibaliz Urizar-Compains
- Department of Biochemistry and Genetics, School of Sciences, University of Navarra, 31008 Pamplona, Spain; (A.G.-H.); (C.H.); (E.U.-C.); (A.G.-S.)
| | - Begoña Ezcurra
- Center for Biomedical Research of La Rioja (CIBIR), 26006 Logroño, Spain; (B.E.); (J.C.)
| | - Alberto Galiana-Sáenz
- Department of Biochemistry and Genetics, School of Sciences, University of Navarra, 31008 Pamplona, Spain; (A.G.-H.); (C.H.); (E.U.-C.); (A.G.-S.)
- Center for Biomedical Research of La Rioja (CIBIR), 26006 Logroño, Spain; (B.E.); (J.C.)
| | - Enrique Baquero
- Department of Environmental Biology, School of Sciences, University of Navarra, 31008 Pamplona, Spain;
- Institute for Biodiversity and Environment BIOMA, University of Navarra, 31008 Pamplona, Spain
| | - Juan Cabello
- Center for Biomedical Research of La Rioja (CIBIR), 26006 Logroño, Spain; (B.E.); (J.C.)
| | - José Luis Vizmanos
- Department of Biochemistry and Genetics, School of Sciences, University of Navarra, 31008 Pamplona, Spain; (A.G.-H.); (C.H.); (E.U.-C.); (A.G.-S.)
| |
Collapse
|
6
|
Shivarov V, Tsvetkova G, Micheva I, Hadjiev E, Petrova J, Ivanova A, Madjarova G, Ivanova M. Differential modulation of mutant CALR and JAK2 V617F-driven oncogenesis by HLA genotype in myeloproliferative neoplasms. Front Immunol 2024; 15:1427810. [PMID: 39351227 PMCID: PMC11439724 DOI: 10.3389/fimmu.2024.1427810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 08/26/2024] [Indexed: 10/04/2024] Open
Abstract
It has been demonstrated previously that human leukocyte antigen class I (HLA-I) and class II (HLA-II) alleles may modulate JAK2 V617F and CALR mutation (CALRmut)-associated oncogenesis in myeloproliferative neoplasms (MPNs). However, the role of immunogenetic factors in MPNs remains underexplored. We aimed to investigate the potential involvement of HLA genes in CALRmut+ MPNs. High-resolution genotyping of HLA-I and -II loci was conducted in 42 CALRmut+ and 158 JAK2 V617F+ MPN patients and 1,083 healthy controls. A global analysis of the diversity of HLA-I genotypes revealed no significant differences between CALRmut+ patients and controls. However, one HLA-I allele (C*06:02) showed an inverse correlation with presence of CALR mutation. A meta-analysis across independent cohorts and healthy individuals from the 1000 Genomes Project confirmed an inverse correlation between the presentation capabilities of the HLA-I loci for JAK2 V617F and CALRmut-derived peptides in both patients and healthy individuals. scRNA-Seq analysis revealed low expression of TAP1 and CIITA genes in CALRmut+ hematopoietic stem and progenitor cells. In conclusion, the HLA-I genotype differentially restricts JAK2 V617F and CALRmut-driven oncogenesis potentially explaining the mutual exclusivity of the two mutations and differences in their presentation latency. These findings have practical implications for the development of neoantigen-based vaccines in MPNs.
Collapse
Affiliation(s)
- Velizar Shivarov
- Department of Experimental Research, Medical University Pleven, Pleven, Bulgaria
| | - Gergana Tsvetkova
- Department of Clinical Hematology, Alexandrovska University Hospital, Medical University Sofia, Sofia, Bulgaria
| | - Ilina Micheva
- Department of Clinical Hematology, Saint Marina University Hospital, Medical University Varna, Varna, Bulgaria
| | - Evgueniy Hadjiev
- Department of Clinical Hematology, Alexandrovska University Hospital, Medical University Sofia, Sofia, Bulgaria
| | - Jasmina Petrova
- Department of Physical Chemistry, Faculty of Chemistry and Pharmacy, Sofia University “St. Kl. Ohridski”, Sofia, Bulgaria
| | - Anela Ivanova
- Department of Physical Chemistry, Faculty of Chemistry and Pharmacy, Sofia University “St. Kl. Ohridski”, Sofia, Bulgaria
| | - Galia Madjarova
- Department of Physical Chemistry, Faculty of Chemistry and Pharmacy, Sofia University “St. Kl. Ohridski”, Sofia, Bulgaria
| | - Milena Ivanova
- Department of Clinical Immunology, Alexandrovska University Hospital, Medical University Sofia, Sofia, Bulgaria
| |
Collapse
|
7
|
Naskar S, Sriraman N, Sarkar A, Mahajan N, Sarkar K. Tumor antigen presentation and the associated signal transduction during carcinogenesis. Pathol Res Pract 2024; 261:155485. [PMID: 39088877 DOI: 10.1016/j.prp.2024.155485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Revised: 07/17/2024] [Accepted: 07/22/2024] [Indexed: 08/03/2024]
Abstract
Numerous developments have been achieved in the study and treatment of cancer throughout the decades that it has been common. After decades of research, about 100 different kinds of cancer have been found, each with unique subgroups within certain organs. This has significantly expanded our understanding of the illness. A mix of genetic, environmental, and behavioral variables contribute to the complicated and diverse process of cancer formation. Mutations, or changes in the DNA sequence, are crucial to the development of cancer. These mutations have the ability to downregulate the expression and function of Major Histocompatibility Complex class I (MHC I) and MHCII receptors, as well as activate oncogenes and inactivate tumor suppressor genes. Cancer cells use this tactic to avoid being recognized by cytotoxic CD8+T lymphocytes, which causes issues with antigen presentation and processing. This review goes into great length into the PI3K pathway, changes to MHC I, and positive impacts of tsMHC-II on disease-free survival and overall survival and the involvement of dendritic cells (DCs) in different tumor microenvironments. The vital functions that the PI3K pathway and its link to the mTOR pathway are highlighted and difficulties in developing effective cancer targeted therapies and feedback systems has also been mentioned, where resistance mechanisms include RAS-mediated oncogenic changes and active PI3K signalling.
Collapse
Affiliation(s)
- Sohom Naskar
- Department of Biotechnology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu 603203, India
| | - Nawaneetan Sriraman
- Department of Biotechnology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu 603203, India
| | - Ankita Sarkar
- Department of Biotechnology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu 603203, India
| | - Nitika Mahajan
- Department of Biotechnology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu 603203, India
| | - Koustav Sarkar
- Department of Biotechnology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu 603203, India.
| |
Collapse
|
8
|
Kaur A, Venkatesan A, Kandarpa M, Talpaz M, Raghavan M. Lysosomal degradation targets mutant calreticulin and the thrombopoietin receptor in myeloproliferative neoplasms. Blood Adv 2024; 8:3372-3387. [PMID: 38640435 PMCID: PMC11255115 DOI: 10.1182/bloodadvances.2023011432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 03/24/2024] [Accepted: 04/15/2024] [Indexed: 04/21/2024] Open
Abstract
ABSTRACT Somatic mutants of calreticulin (CRT) drive myeloproliferative neoplasms (MPNs) via binding to the thrombopoietin receptor (MPL) and aberrant activation of the JAK/STAT pathway. Compared with healthy donors, platelets from mutant CRT-expressing patients with MPN display low cell surface MPL. Additionally, coexpression of MPL with an MPN-linked CRT mutant (CRTDel52) reduces cell surface MPL, suggesting that CRTDel52 may induce MPL degradation. We show that lysosomal degradation is relevant to the turnover of CRTDel52 and MPL. Furthermore, CRTDel52 increases the lysosomal localization and degradation of MPL. Mammalian target of rapamycin (mTOR) inhibitors reduce cellular CRTDel52 and MPL, secreted CRTDel52 levels, and impair CRTDel52-mediated cell proliferation. mTOR inhibition also reduces colony formation and differentiation of CD34+ cells from patients with MPN but not from healthy donors. Together, these findings indicate that low-surface MPL is a biomarker of mutant CRT-mediated MPN and that induced degradation of CRTDel52 and MPL is an avenue for therapeutic intervention.
Collapse
Affiliation(s)
- Amanpreet Kaur
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI
| | - Arunkumar Venkatesan
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI
| | - Malathi Kandarpa
- Division of Hematology/Oncology, Department of Internal Medicine, University of Michigan Rogel Cancer Center, Ann Arbor, MI
| | - Moshe Talpaz
- Division of Hematology/Oncology, Department of Internal Medicine, University of Michigan Rogel Cancer Center, Ann Arbor, MI
| | - Malini Raghavan
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI
| |
Collapse
|
9
|
Brunnberg J, Barends M, Frühschulz S, Winter C, Battin C, de Wet B, Cole DK, Steinberger P, Tampé R. Dual role of the peptide-loading complex as proofreader and limiter of MHC-I presentation. Proc Natl Acad Sci U S A 2024; 121:e2321600121. [PMID: 38771881 PMCID: PMC11145271 DOI: 10.1073/pnas.2321600121] [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: 12/08/2023] [Accepted: 04/17/2024] [Indexed: 05/23/2024] Open
Abstract
Antigen presentation via major histocompatibility complex class I (MHC-I) molecules is essential for surveillance by the adaptive immune system. Central to this process is the peptide-loading complex (PLC), which translocates peptides from the cytosol to the endoplasmic reticulum and catalyzes peptide loading and proofreading of peptide-MHC-I (pMHC-I) complexes. Despite its importance, the impact of individual PLC components on the presented pMHC-I complexes is still insufficiently understood. Here, we used stoichiometrically defined antibody-nanobody complexes and engineered soluble T cell receptors (sTCRs) to quantify different MHC-I allomorphs and defined pMHC-I complexes, respectively. Thereby, we uncovered distinct effects of individual PLC components on the pMHC-I surface pool. Knockouts of components of the PLC editing modules, namely tapasin, ERp57, or calreticulin, changed the MHC-I surface composition to a reduced proportion of HLA-A*02:01 presentation compensated by a higher ratio of HLA-B*40:01 molecules. Intriguingly, these knockouts not only increased the presentation of suboptimally loaded HLA-A*02:01 complexes but also elevated the presentation of high-affinity peptides overexpressed in the cytosol. Our findings suggest that the components of the PLC editing module serve a dual role, acting not only as peptide proofreaders but also as limiters for abundant peptides. This dual function ensures the presentation of a broad spectrum of antigenic peptides.
Collapse
Affiliation(s)
- Jamina Brunnberg
- Institute of Biochemistry, Biocenter, Goethe University Frankfurt, Frankfurt am Main60438, Germany
| | - Martina Barends
- Institute of Biochemistry, Biocenter, Goethe University Frankfurt, Frankfurt am Main60438, Germany
| | - Stefan Frühschulz
- Institute of Biochemistry, Biocenter, Goethe University Frankfurt, Frankfurt am Main60438, Germany
| | - Christian Winter
- Institute of Biochemistry, Biocenter, Goethe University Frankfurt, Frankfurt am Main60438, Germany
| | - Claire Battin
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna1090, Austria
| | - Ben de Wet
- Immunocore Ltd., AbingdonOX14 4RY, United Kingdom
| | | | - Peter Steinberger
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna1090, Austria
| | - Robert Tampé
- Institute of Biochemistry, Biocenter, Goethe University Frankfurt, Frankfurt am Main60438, Germany
| |
Collapse
|
10
|
Michalak M. Calreticulin: Endoplasmic reticulum Ca 2+ gatekeeper. J Cell Mol Med 2024; 28:e17839. [PMID: 37424156 PMCID: PMC10902585 DOI: 10.1111/jcmm.17839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 06/21/2023] [Accepted: 06/27/2023] [Indexed: 07/11/2023] Open
Abstract
Endoplasmic reticulum (ER) luminal Ca2+ is vital for the function of the ER and regulates many cellular processes. Calreticulin is a highly conserved, ER-resident Ca2+ binding protein and lectin-like chaperone. Over four decades of studying calreticulin demonstrate that this protein plays a crucial role in maintaining Ca2+ supply under different physiological conditions, in managing access to Ca2+ and how Ca2+ is used depending on the environmental events and in making sure that Ca2+ is not misused. Calreticulin plays a role of ER luminal Ca2+ sensor to manage Ca2+-dependent ER luminal events including maintaining interaction with its partners, Ca2+ handling molecules, substrates and stress sensors. The protein is strategically positioned in the lumen of the ER from where the protein manages access to and distribution of Ca2+ for many cellular Ca2+-signalling events. The importance of calreticulin Ca2+ pool extends beyond the ER and includes influence of cellular processes involved in many aspects of cellular pathophysiology. Abnormal handling of the ER Ca2+ contributes to many pathologies from heart failure to neurodegeneration and metabolic diseases.
Collapse
Affiliation(s)
- Marek Michalak
- Department of BiochemistryUniversity of AlbertaEdmontonAlbertaCanada
| |
Collapse
|
11
|
Palacon MP, Ferrisse TM, Barbeiro CDO, Massucato EMS, Bufalino A. Calreticulin Expression in Human Carcinomas: A Systematic Review and Meta-Analysis. Asian Pac J Cancer Prev 2023; 24:2929-2940. [PMID: 37774043 PMCID: PMC10762747 DOI: 10.31557/apjcp.2023.24.9.2929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 09/11/2023] [Indexed: 10/01/2023] Open
Abstract
OBJECTIVE The present study performed a systematic review and meta-analysis of observational studies on whether calreticulin levels could represent a prognostic factor in carcinoma patients. Calreticulin (CRT) is a multifunctional protein in the endoplasmic reticulum that can play distinct roles in different cancers. METHODS The search was performed in PubMed, Scopus, the Cochrane Library, Web of Science, Lilacs, Science Direct, Embase, Bireme, and SciELO databases. After a full-text evaluation, only 14 articles remained. The RoBANS tool assessed the risk of bias. The meta-analysis was performed with R software, and the odds ratio (OR) was the effect measure. The random effects model was chosen, and the quality of evidence was evaluated according to GRADE. RESULT The most frequent carcinomas were in the breasts and the colon. CRT expression varied according to carcinoma origin and type, but these diseases had a prevalence of high CRT levels, indicating tumor progression. The high CRT levels were associated with lymph node metastasis (OR = 3.06 [1.71; 5.48]/p = 0.0002/I2 = 0%). All included articles had a blinding bias. CONCLUSION High CRT levels may represent a prognostic factor for metastatic lymph nodes in carcinoma patients.
Collapse
Affiliation(s)
- Mariana Paravani Palacon
- Department of Diagnosis and Surgery, São Paulo State University (UNESP), School of Dentistry, Araraquara, São Paulo, Brazil.
| | - Tulio Morandin Ferrisse
- Department of Dental Prothesis, São Paulo State University (UNESP), School of Dentistry, Araraquara, São Paulo, Brazil.
| | - Camila de Oliveira Barbeiro
- Department of Diagnosis and Surgery, São Paulo State University (UNESP), School of Dentistry, Araraquara, São Paulo, Brazil.
| | - Elaine Maria Sgavioli Massucato
- Department of Diagnosis and Surgery, São Paulo State University (UNESP), School of Dentistry, Araraquara, São Paulo, Brazil.
| | - Andreia Bufalino
- Department of Diagnosis and Surgery, São Paulo State University (UNESP), School of Dentistry, Araraquara, São Paulo, Brazil.
| |
Collapse
|
12
|
Yang K, Halima A, Chan TA. Antigen presentation in cancer - mechanisms and clinical implications for immunotherapy. Nat Rev Clin Oncol 2023; 20:604-623. [PMID: 37328642 DOI: 10.1038/s41571-023-00789-4] [Citation(s) in RCA: 84] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/25/2023] [Indexed: 06/18/2023]
Abstract
Over the past decade, the emergence of effective immunotherapies has revolutionized the clinical management of many types of cancers. However, long-term durable tumour control is only achieved in a fraction of patients who receive these therapies. Understanding the mechanisms underlying clinical response and resistance to treatment is therefore essential to expanding the level of clinical benefit obtained from immunotherapies. In this Review, we describe the molecular mechanisms of antigen processing and presentation in tumours and their clinical consequences. We examine how various aspects of the antigen-presentation machinery (APM) shape tumour immunity. In particular, we discuss genomic variants in HLA alleles and other APM components, highlighting their influence on the immunopeptidomes of both malignant cells and immune cells. Understanding the APM, how it is regulated and how it changes in tumour cells is crucial for determining which patients will respond to immunotherapy and why some patients develop resistance. We focus on recently discovered molecular and genomic alterations that drive the clinical outcomes of patients receiving immune-checkpoint inhibitors. An improved understanding of how these variables mediate tumour-immune interactions is expected to guide the more precise administration of immunotherapies and reveal potentially promising directions for the development of new immunotherapeutic approaches.
Collapse
Affiliation(s)
- Kailin Yang
- Department of Radiation Oncology, Taussig Cancer Center, Cleveland Clinic, Cleveland, OH, USA
| | - Ahmed Halima
- Department of Radiation Oncology, Taussig Cancer Center, Cleveland Clinic, Cleveland, OH, USA
| | - Timothy A Chan
- Department of Radiation Oncology, Taussig Cancer Center, Cleveland Clinic, Cleveland, OH, USA.
- Center for Immunotherapy and Precision Immuno-Oncology, Cleveland Clinic, Cleveland, OH, USA.
- National Center for Regenerative Medicine, Cleveland, OH, USA.
- Case Comprehensive Cancer Center, Cleveland, OH, USA.
| |
Collapse
|
13
|
Chen X, Lu Q, Zhou H, Liu J, Nadorp B, Lasry A, Sun Z, Lai B, Rona G, Zhang J, Cammer M, Wang K, Al-Santli W, Ciantra Z, Guo Q, You J, Sengupta D, Boukhris A, Zhang H, Liu C, Cresswell P, Dahia PLM, Pagano M, Aifantis I, Wang J. A membrane-associated MHC-I inhibitory axis for cancer immune evasion. Cell 2023; 186:3903-3920.e21. [PMID: 37557169 PMCID: PMC10961051 DOI: 10.1016/j.cell.2023.07.016] [Citation(s) in RCA: 75] [Impact Index Per Article: 37.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 05/30/2023] [Accepted: 07/11/2023] [Indexed: 08/11/2023]
Abstract
Immune-checkpoint blockade has revolutionized cancer treatment, but some cancers, such as acute myeloid leukemia (AML), do not respond or develop resistance. A potential mode of resistance is immune evasion of T cell immunity involving aberrant major histocompatibility complex class I (MHC-I) antigen presentation (AP). To map such mechanisms of resistance, we identified key MHC-I regulators using specific peptide-MHC-I-guided CRISPR-Cas9 screens in AML. The top-ranked negative regulators were surface protein sushi domain containing 6 (SUSD6), transmembrane protein 127 (TMEM127), and the E3 ubiquitin ligase WWP2. SUSD6 is abundantly expressed in AML and multiple solid cancers, and its ablation enhanced MHC-I AP and reduced tumor growth in a CD8+ T cell-dependent manner. Mechanistically, SUSD6 forms a trimolecular complex with TMEM127 and MHC-I, which recruits WWP2 for MHC-I ubiquitination and lysosomal degradation. Together with the SUSD6/TMEM127/WWP2 gene signature, which negatively correlates with cancer survival, our findings define a membrane-associated MHC-I inhibitory axis as a potential therapeutic target for both leukemia and solid cancers.
Collapse
Affiliation(s)
- Xufeng Chen
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA; The Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY 10016, USA
| | - Qiao Lu
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA; The Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY 10016, USA
| | - Hua Zhou
- Applied Bioinformatics Laboratories, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Jia Liu
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA; The Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY 10016, USA
| | - Bettina Nadorp
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA; The Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY 10016, USA
| | - Audrey Lasry
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA; The Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY 10016, USA
| | - Zhengxi Sun
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA; The Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY 10016, USA
| | - Baoling Lai
- Department of Neuroscience and Physiology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Gergely Rona
- The Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY 10016, USA; Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY 10016, USA; Howard Hughes Medical Institute, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Jiangyan Zhang
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA; The Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY 10016, USA
| | - Michael Cammer
- Microscopy Core, Division of Advanced Research Technologies, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Kun Wang
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA; The Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY 10016, USA
| | - Wafa Al-Santli
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA; The Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY 10016, USA
| | - Zoe Ciantra
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Qianjin Guo
- Department of Medicine, Division of Hematology and Medical Oncology, Mays Cancer Center, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Jia You
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA; The Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY 10016, USA
| | - Debrup Sengupta
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06519, USA
| | - Ahmad Boukhris
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA; The Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY 10016, USA
| | | | - Cheng Liu
- Eureka Therapeutics Inc., Emeryville, CA 94608, USA
| | - Peter Cresswell
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06519, USA
| | - Patricia L M Dahia
- Department of Medicine, Division of Hematology and Medical Oncology, Mays Cancer Center, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Michele Pagano
- The Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY 10016, USA; Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY 10016, USA; Howard Hughes Medical Institute, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Iannis Aifantis
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA; The Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY 10016, USA.
| | - Jun Wang
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA; The Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY 10016, USA.
| |
Collapse
|
14
|
Holmström MO, Andersen M, Traynor S, Ahmad SM, Lisle TL, Handlos Grauslund J, Skov V, Kjær L, Ottesen JT, Gjerstorff MF, Hasselbalch HC, Andersen MH. Therapeutic cancer vaccination against mutant calreticulin in myeloproliferative neoplasms induces expansion of specific T cells in the periphery but specific T cells fail to enrich in the bone marrow. Front Immunol 2023; 14:1240678. [PMID: 37662956 PMCID: PMC10470021 DOI: 10.3389/fimmu.2023.1240678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 07/12/2023] [Indexed: 09/05/2023] Open
Abstract
Background Therapeutic cancer vaccination against mutant calreticulin (CALR) in patients with CALR-mutant (CALRmut) myeloproliferative neoplasms (MPN) induces strong T-cell responses against mutant CALR yet fails to demonstrate clinical activity. Infiltration of tumor specific T cells into the tumor microenvironment is needed to attain a clinical response to therapeutic cancer vaccination. Aim Determine if CALRmut specific T cells isolated from vaccinated patients enrich in the bone marrow upon completion of vaccination and explore possible explanations for the lack of enrichment. Methods CALRmut specific T cells from four of ten vaccinated patients were expanded, enriched, and analyzed by T-cell receptor sequencing (TCRSeq). The TCRs identified were used as fingerprints of CALRmut specific T cells. Bone marrow aspirations from the four patients were acquired at baseline and at the end of trial. T cells were enriched from the bone marrow aspirations and analyzed by TCRSeq to identify the presence and fraction of CALRmut specific T cells at the two different time points. In silico calculations were performed to calculate the ratio between transformed cells and effector cells in patients with CALRmut MPN. Results The fraction of CALRmut specific T cells in the bone marrow did not increase upon completion of the vaccination trial. In general, the T cell repertoire in the bone marrow remains relatively constant through the vaccination trial. The enriched and expanded CALRmut specific T cells recognize peripheral blood autologous CALRmut cells. In silico analyses demonstrate a high imbalance in the fraction of CALRmut cells and CALRmut specific effector T-cells in peripheral blood. Conclusion CALRmut specific T cells do not enrich in the bone marrow after therapeutic cancer peptide vaccination against mutant CALR. The specific T cells recognize autologous peripheral blood derived CALRmut cells. In silico analyses demonstrate a high imbalance between the number of transformed cells and CALRmut specific effector T-cells in the periphery. We suggest that the high burden of transformed cells in the periphery compared to the number of effector cells could impact the ability of specific T cells to enrich in the bone marrow.
Collapse
Affiliation(s)
- Morten Orebo Holmström
- National Center for Cancer Immune Therapy, Department of Oncology, Herlev University Hospital, Herlev, Denmark
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Morten Andersen
- Centre for Mathematical Modeling – Human Health and Disease, IMFUFA, Department of Science and Environment, Roskilde University, Roskilde, Denmark
| | - Sofie Traynor
- Department of Cancer and Inflammation Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Shamaila Munir Ahmad
- National Center for Cancer Immune Therapy, Department of Oncology, Herlev University Hospital, Herlev, Denmark
| | - Thomas Landkildehus Lisle
- National Center for Cancer Immune Therapy, Department of Oncology, Herlev University Hospital, Herlev, Denmark
| | - Jacob Handlos Grauslund
- National Center for Cancer Immune Therapy, Department of Oncology, Herlev University Hospital, Herlev, Denmark
| | - Vibe Skov
- Department of Hematology, Zealand University Hospital, Roskilde, Denmark
| | - Lasse Kjær
- Department of Hematology, Zealand University Hospital, Roskilde, Denmark
| | - Johnny T. Ottesen
- Centre for Mathematical Modeling – Human Health and Disease, IMFUFA, Department of Science and Environment, Roskilde University, Roskilde, Denmark
| | - Morten Frier Gjerstorff
- Department of Cancer and Inflammation Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
- Department of Oncology, Odense University Hospital, Odense, Denmark
| | | | - Mads Hald Andersen
- National Center for Cancer Immune Therapy, Department of Oncology, Herlev University Hospital, Herlev, Denmark
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
15
|
Luque Paz D, Kralovics R, Skoda RC. Genetic basis and molecular profiling in myeloproliferative neoplasms. Blood 2023; 141:1909-1921. [PMID: 36347013 PMCID: PMC10646774 DOI: 10.1182/blood.2022017578] [Citation(s) in RCA: 63] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 11/03/2022] [Accepted: 11/03/2022] [Indexed: 11/11/2022] Open
Abstract
BCR::ABL1-negative myeloproliferative neoplasms (MPNs) are clonal diseases originating from a single hematopoietic stem cell that cause excessive production of mature blood cells. The 3 subtypes, that is, polycythemia vera (PV), essential thrombocythemia (ET), and primary myelofibrosis (PMF), are diagnosed according to the World Health Organization (WHO) and international consensus classification (ICC) criteria. Acquired gain-of-function mutations in 1 of 3 disease driver genes (JAK2, CALR, and MPL) are the causative events that can alone initiate and promote MPN disease without requiring additional cooperating mutations. JAK2-p.V617F is present in >95% of PV patients, and also in about half of the patients with ET or PMF. ET and PMF are also caused by mutations in CALR or MPL. In ∼10% of MPN patients, those referred to as being "triple negative," none of the known driver gene mutations can be detected. The common theme between the 3 driver gene mutations and triple-negative MPN is that the Janus kinase-signal transducer and activator of transcription (JAK/STAT) signaling pathway is constitutively activated. We review the recent advances in our understanding of the early events after the acquisition of a driver gene mutation. The limiting factor that determines the frequency at which MPN disease develops with a long latency is not the acquisition of driver gene mutations, but rather the expansion of the clone. Factors that control the conversion from clonal hematopoiesis to MPN disease include inherited predisposition, presence of additional mutations, and inflammation. The full extent of knowledge of the mutational landscape in individual MPN patients is now increasingly being used to predict outcome and chose the optimal therapy.
Collapse
Affiliation(s)
- Damien Luque Paz
- Univ Angers, Nantes Université, CHU Angers, Inserm, CNRS, CRCI2NA, Angers, France
| | - Robert Kralovics
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Radek C. Skoda
- Department of Biomedicine, Experimental Hematology, University Hospital Basel and University of Basel, Basel, Switzerland
| |
Collapse
|
16
|
Desikan H, Kaur A, Pogozheva ID, Raghavan M. Effects of calreticulin mutations on cell transformation and immunity. J Cell Mol Med 2023; 27:1032-1044. [PMID: 36916035 PMCID: PMC10098294 DOI: 10.1111/jcmm.17713] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 02/22/2023] [Accepted: 02/24/2023] [Indexed: 03/16/2023] Open
Abstract
Myeloproliferative neoplasms (MPNs) are cancers involving dysregulated production and function of myeloid lineage hematopoietic cells. Among MPNs, Essential thrombocythemia (ET), Polycythemia Vera (PV) and Myelofibrosis (MF), are driven by mutations that activate the JAK-STAT signalling pathway. Somatic mutations of calreticulin (CRT), an endoplasmic reticulum (ER)-localized lectin chaperone, are driver mutations in approximately 25% of ET and 35% of MF patients. The MPN-linked mutant CRT proteins have novel frameshifted carboxy-domain sequences and lack an ER retention motif, resulting in their secretion. Wild type CRT is a regulator of ER calcium homeostasis and plays a key role in the assembly of major histocompatibility complex (MHC) class I molecules, which are the ligands for antigen receptors of CD8+ T cells. Mutant CRT-linked oncogenesis results from the dysregulation of calcium signalling in cells and the formation of stable complexes of mutant CRT with myeloproliferative leukemia (MPL) protein, followed by downstream activation of the JAK-STAT signalling pathway. The intricate participation of CRT in ER protein folding, calcium homeostasis and immunity suggests the involvement of multiple mechanisms of mutant CRT-linked oncogenesis. In this review, we highlight recent findings related to the role of MPN-linked CRT mutations in the dysregulation of calcium homeostasis, MPL activation and immunity.
Collapse
Affiliation(s)
- Harini Desikan
- Department of Microbiology and ImmunologyUniversity of Michigan Medical SchoolAnn ArborMichiganUSA
| | - Amanpreet Kaur
- Department of Microbiology and ImmunologyUniversity of Michigan Medical SchoolAnn ArborMichiganUSA
| | - Irina D. Pogozheva
- Department of Medicinal ChemistryCollege of Pharmacy, University of MichiganAnn ArborMichiganUSA
| | - Malini Raghavan
- Department of Microbiology and ImmunologyUniversity of Michigan Medical SchoolAnn ArborMichiganUSA
| |
Collapse
|
17
|
Foßelteder J, Pabst G, Sconocchia T, Schlacher A, Auinger L, Kashofer K, Beham-Schmid C, Trajanoski S, Waskow C, Schöll W, Sill H, Zebisch A, Wölfler A, Thomas D, Reinisch A. Human gene-engineered calreticulin mutant stem cells recapitulate MPN hallmarks and identify targetable vulnerabilities. Leukemia 2023; 37:843-853. [PMID: 36813992 PMCID: PMC10079532 DOI: 10.1038/s41375-023-01848-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 02/07/2023] [Accepted: 02/08/2023] [Indexed: 02/24/2023]
Abstract
Calreticulin (CALR) mutations present the main oncogenic drivers in JAK2 wildtype (WT) myeloproliferative neoplasms (MPN), including essential thrombocythemia and myelofibrosis, where mutant (MUT) CALR is increasingly recognized as a suitable mutation-specific drug target. However, our current understanding of its mechanism-of-action is derived from mouse models or immortalized cell lines, where cross-species differences, ectopic over-expression and lack of disease penetrance are hampering translational research. Here, we describe the first human gene-engineered model of CALR MUT MPN using a CRISPR/Cas9 and adeno-associated viral vector-mediated knock-in strategy in primary human hematopoietic stem and progenitor cells (HSPCs) to establish a reproducible and trackable phenotype in vitro and in xenografted mice. Our humanized model recapitulates many disease hallmarks: thrombopoietin-independent megakaryopoiesis, myeloid-lineage skewing, splenomegaly, bone marrow fibrosis, and expansion of megakaryocyte-primed CD41+ progenitors. Strikingly, introduction of CALR mutations enforced early reprogramming of human HSPCs and the induction of an endoplasmic reticulum stress response. The observed compensatory upregulation of chaperones revealed novel mutation-specific vulnerabilities with preferential sensitivity of CALR mutant cells to inhibition of the BiP chaperone and the proteasome. Overall, our humanized model improves purely murine models and provides a readily usable basis for testing of novel therapeutic strategies in a human setting.
Collapse
Affiliation(s)
- Johannes Foßelteder
- Department of Internal Medicine, Division of Hematology, Medical University of Graz, Graz, Austria
| | - Gabriel Pabst
- Department of Internal Medicine, Division of Hematology, Medical University of Graz, Graz, Austria.,Research Institute of Molecular Pathology (IMP), Vienna BioCenter (VBC), Vienna, Austria.,Vienna BioCenter PhD Program, Doctoral School of the University of Vienna and Medical University of Vienna, Vienna BioCenter (VBC), Vienna, Austria
| | - Tommaso Sconocchia
- Department of Internal Medicine, Division of Hematology, Medical University of Graz, Graz, Austria
| | - Angelika Schlacher
- Department of Internal Medicine, Division of Hematology, Medical University of Graz, Graz, Austria
| | - Lisa Auinger
- Department of Internal Medicine, Division of Hematology, Medical University of Graz, Graz, Austria
| | - Karl Kashofer
- Diagnostic & Research Institute of Pathology, Medical University of Graz, Graz, Austria
| | | | - Slave Trajanoski
- Core Facility Computational Bioanalytics, Medical University of Graz, Graz, Austria
| | - Claudia Waskow
- Leibniz Institute on Aging, Fritz Lipmann Institute, Jena, Germany.,Institute of Biochemistry and Biophysics, Faculty of Biological Sciences, Friedrich-Schiller-University, Jena, Germany
| | - Wolfgang Schöll
- Department of Obstetrics and Gynecology, Medical University of Graz, Graz, Austria
| | - Heinz Sill
- Department of Internal Medicine, Division of Hematology, Medical University of Graz, Graz, Austria
| | - Armin Zebisch
- Department of Internal Medicine, Division of Hematology, Medical University of Graz, Graz, Austria.,Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Division of Pharmacology, Medical University of Graz, Graz, Austria
| | - Albert Wölfler
- Department of Internal Medicine, Division of Hematology, Medical University of Graz, Graz, Austria
| | - Daniel Thomas
- Cancer Program, Precision Medicine Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, Australia.,Adelaide Medical School, The University of Adelaide, Adelaide, Australia
| | - Andreas Reinisch
- Department of Internal Medicine, Division of Hematology, Medical University of Graz, Graz, Austria. .,Department of Blood Group Serology and Transfusion Medicine, Medical University of Graz, Graz, Austria.
| |
Collapse
|
18
|
Calreticulin mutations affect its chaperone function and perturb the glycoproteome. Cell Rep 2022; 41:111689. [DOI: 10.1016/j.celrep.2022.111689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 07/17/2022] [Accepted: 10/27/2022] [Indexed: 11/23/2022] Open
|
19
|
Gigoux M, Holmström MO, Zappasodi R, Park JJ, Pourpe S, Bozkus CC, Mangarin LMB, Redmond D, Verma S, Schad S, George MM, Venkatesh D, Ghosh A, Hoyos D, Molvi Z, Kamaz B, Marneth AE, Duke W, Leventhal MJ, Jan M, Ho VT, Hobbs GS, Knudsen TA, Skov V, Kjær L, Larsen TS, Hansen DL, Lindsley RC, Hasselbalch H, Grauslund JH, Lisle TL, Met Ö, Wilkinson P, Greenbaum B, Sepulveda MA, Chan T, Rampal R, Andersen MH, Abdel-Wahab O, Bhardwaj N, Wolchok JD, Mullally A, Merghoub T. Calreticulin mutant myeloproliferative neoplasms induce MHC-I skewing, which can be overcome by an optimized peptide cancer vaccine. Sci Transl Med 2022; 14:eaba4380. [PMID: 35704596 PMCID: PMC11182673 DOI: 10.1126/scitranslmed.aba4380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
The majority of JAK2V617F-negative myeloproliferative neoplasms (MPNs) have disease-initiating frameshift mutations in calreticulin (CALR), resulting in a common carboxyl-terminal mutant fragment (CALRMUT), representing an attractive source of neoantigens for cancer vaccines. However, studies have shown that CALRMUT-specific T cells are rare in patients with CALRMUT MPN for unknown reasons. We examined class I major histocompatibility complex (MHC-I) allele frequencies in patients with CALRMUT MPN from two independent cohorts. We observed that MHC-I alleles that present CALRMUT neoepitopes with high affinity are underrepresented in patients with CALRMUT MPN. We speculated that this was due to an increased chance of immune-mediated tumor rejection by individuals expressing one of these MHC-I alleles such that the disease never clinically manifested. As a consequence of this MHC-I allele restriction, we reasoned that patients with CALRMUT MPN would not efficiently respond to a CALRMUT fragment cancer vaccine but would when immunized with a modified CALRMUT heteroclitic peptide vaccine approach. We found that heteroclitic CALRMUT peptides specifically designed for the MHC-I alleles of patients with CALRMUT MPN efficiently elicited a CALRMUT cross-reactive CD8+ T cell response in human peripheral blood samples but not to the matched weakly immunogenic CALRMUT native peptides. We corroborated this effect in vivo in mice and observed that C57BL/6J mice can mount a CD8+ T cell response to the CALRMUT fragment upon immunization with a CALRMUT heteroclitic, but not native, peptide. Together, our data emphasize the therapeutic potential of heteroclitic peptide-based cancer vaccines in patients with CALRMUT MPN.
Collapse
Affiliation(s)
- Mathieu Gigoux
- Ludwig Collaborative and Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Human Oncology and Pathogenesis Program and Immuno-Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Morten O. Holmström
- Department of Oncology, National Center for Cancer Immune Therapy, Herlev Hospital, Herlev 2730, Denmark
- Department of Immunology and Microbiology, Copenhagen University Hospital, Herlev 2730, Denmark
| | - Roberta Zappasodi
- Ludwig Collaborative and Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Human Oncology and Pathogenesis Program and Immuno-Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY 10021, USA
| | - Joseph J. Park
- Ludwig Collaborative and Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Weill Cornell Medical College, New York, NY 10065, USA
| | - Stephane Pourpe
- Ludwig Collaborative and Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | | | - Levi M. B. Mangarin
- Ludwig Collaborative and Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Human Oncology and Pathogenesis Program and Immuno-Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - David Redmond
- Ludwig Collaborative and Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Division of Regenerative Medicine, Ansary Stem Cell Institute, Weill Cornell Medicine, New York, NY 10065, USA
| | - Svena Verma
- Ludwig Collaborative and Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Human Oncology and Pathogenesis Program and Immuno-Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Weill Cornell Medical College, New York, NY 10065, USA
| | - Sara Schad
- Ludwig Collaborative and Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Human Oncology and Pathogenesis Program and Immuno-Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Weill Cornell Medical College, New York, NY 10065, USA
| | - Mariam M. George
- Ludwig Collaborative and Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Human Oncology and Pathogenesis Program and Immuno-Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Divya Venkatesh
- Ludwig Collaborative and Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Human Oncology and Pathogenesis Program and Immuno-Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Arnab Ghosh
- Ludwig Collaborative and Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Human Oncology and Pathogenesis Program and Immuno-Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Adult Bone Marrow Transplantation Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - David Hoyos
- Computational Oncology, Department of Epidemiology & Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Zaki Molvi
- Weill Cornell Medicine, New York, NY 10065, USA
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Baransel Kamaz
- Department of Medicine, Division of Hematology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Anna E. Marneth
- Department of Medicine, Division of Hematology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - William Duke
- Department of Medicine, Division of Hematology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | | | - Max Jan
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Vincent T. Ho
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Gabriela S. Hobbs
- Department of Medical Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Trine Alma Knudsen
- Department of Hematology, Zealand University Hospital, Roskilde 4000, Denmark
| | - Vibe Skov
- Department of Hematology, Zealand University Hospital, Roskilde 4000, Denmark
| | - Lasse Kjær
- Department of Hematology, Zealand University Hospital, Roskilde 4000, Denmark
| | | | - Dennis Lund Hansen
- Department of Hematology, Odense University Hospital, Odense 5000, Denmark
| | - R. Coleman Lindsley
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Hans Hasselbalch
- Department of Hematology, Zealand University Hospital, Roskilde 4000, Denmark
| | - Jacob H. Grauslund
- Department of Oncology, National Center for Cancer Immune Therapy, Herlev Hospital, Herlev 2730, Denmark
- Department of Immunology and Microbiology, Copenhagen University Hospital, Herlev 2730, Denmark
| | - Thomas L. Lisle
- Department of Oncology, National Center for Cancer Immune Therapy, Herlev Hospital, Herlev 2730, Denmark
- Department of Immunology and Microbiology, Copenhagen University Hospital, Herlev 2730, Denmark
| | - Özcan Met
- Department of Oncology, National Center for Cancer Immune Therapy, Herlev Hospital, Herlev 2730, Denmark
- Department of Immunology and Microbiology, Copenhagen University Hospital, Herlev 2730, Denmark
| | - Patrick Wilkinson
- Janssen Oncology Therapeutic Area, Janssen Research and Development, LLC, Pharmaceutical Companies of Johnson & Johnson, Spring House, PA 19002, USA
| | - Benjamin Greenbaum
- Computational Oncology, Department of Epidemiology & Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Weill Cornell Medicine, Physiology, Biophysics and Systems Biology, Weill Cornell Medical College, New York, NY 10065, USA
| | - Manuel A. Sepulveda
- Janssen Oncology Therapeutic Area, Janssen Research and Development, LLC, Pharmaceutical Companies of Johnson & Johnson, Spring House, PA 19002, USA
| | - Timothy Chan
- Weill Cornell Medical College, New York, NY 10065, USA
- Immunogenomics and Precision Oncology Platform, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Raajit Rampal
- Human Oncology and Pathogenesis Program and Leukemia Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Mads H. Andersen
- Department of Oncology, National Center for Cancer Immune Therapy, Herlev Hospital, Herlev 2730, Denmark
- Department of Immunology and Microbiology, Copenhagen University Hospital, Herlev 2730, Denmark
| | - Omar Abdel-Wahab
- Human Oncology and Pathogenesis Program and Leukemia Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Nina Bhardwaj
- Parker Institute for Cancer Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Jedd D. Wolchok
- Ludwig Collaborative and Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Human Oncology and Pathogenesis Program and Immuno-Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Weill Cornell Medical College, New York, NY 10065, USA
| | - Ann Mullally
- Department of Medicine, Division of Hematology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Broad Institute, Cambridge, MA 02142, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Taha Merghoub
- Ludwig Collaborative and Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Human Oncology and Pathogenesis Program and Immuno-Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Weill Cornell Medical College, New York, NY 10065, USA
| |
Collapse
|
20
|
Mercier R, LaPointe P. The role of cellular proteostasis in anti-tumor immunity. J Biol Chem 2022; 298:101930. [PMID: 35421375 PMCID: PMC9108985 DOI: 10.1016/j.jbc.2022.101930] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 03/21/2022] [Accepted: 03/31/2022] [Indexed: 12/25/2022] Open
Abstract
Immune checkpoint blockade therapy is perhaps the most important development in cancer treatment in recent memory. It is based on decades of investigation into the biology of immune cells and the role of the immune system in controlling cancer growth. While the molecular circuitry that governs the immune system in general - and anti-tumor immunity in particular - is intensely studied, far less attention has been paid to the role of cellular stress in this process. Proteostasis, intimately linked to cell stress responses, refers to the dynamic regulation of the cellular proteome and is maintained through a complex network of systems that govern the synthesis, folding, and degradation of proteins in the cell. Disruption of these systems can result in the loss of protein function, altered protein function, the formation of toxic aggregates, or pathologies associated with cell stress. However, the importance of proteostasis extends beyond its role in maintaining proper protein function; proteostasis governs how tolerant cells may be to mutations in protein coding genes and the overall half-life of proteins. Such gene expression changes may be associated with human diseases including neurodegenerative diseases, metabolic disease, and cancer and manifest at the protein level against the backdrop of the proteostasis network in any given cellular environment. In this review, we focus on the role of proteostasis in regulating immune responses against cancer as well the role of proteostasis in determining immunogenicity of cancer cells.
Collapse
Affiliation(s)
- Rebecca Mercier
- Department of Cell Biology, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Paul LaPointe
- Department of Cell Biology, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
21
|
Bartalucci N, Galluzzi L. Philadelphia-negative myeloproliferative neoplasms: From origins to new perspectives. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2022; 366:ix-xx. [PMID: 35153008 DOI: 10.1016/s1937-6448(22)00019-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Affiliation(s)
- Niccolò Bartalucci
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy; DENOThe Excellence Center, University of Florence, Florence, Italy.
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, United States; Sandra and Edward Meyer Cancer Center, New York, NY, United States; Caryl and Israel Englander Institute for Precision Medicine, New York, NY, United States; Department of Dermatology, Yale School of Medicine, New Haven, CT, United States; Université de Paris, Paris, France.
| |
Collapse
|
22
|
Levy G, Mambet C, Pecquet C, Bailly S, Havelange V, Diaconu CC, Constantinescu SN. Targets in MPNs and potential therapeutics. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2021; 366:41-81. [PMID: 35153006 DOI: 10.1016/bs.ircmb.2021.06.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Philadelphia-negative classical Myeloproliferative Neoplasms (MPNs), including Polycythemia Vera (PV), Essential Thrombocythemia (ET) and Primary Myelofibrosis (PMF), are clonal hemopathies that emerge in the hematopoietic stem cell (HSC) compartment. MPN driver mutations are restricted to specific exons (14 and 12) of Janus kinase 2 (JAK2), thrombopoietin receptor (MPL/TPOR) and calreticulin (CALR) genes, are involved directly in clonal myeloproliferation and generate the MPN phenotype. As a result, an increased number of fully functional erythrocytes, platelets and leukocytes is observed in the peripheral blood. Nevertheless, the complexity and heterogeneity of MPN clinical phenotypes cannot be solely explained by the type of driver mutation. Other factors, such as additional somatic mutations affecting epigenetic regulators or spliceosomes components, mutant allele burdens and modifiers of signaling by driver mutants, clonal architecture and the order of mutation acquisition, signaling events that occur downstream of a driver mutation, the presence of specific germ-line variants, the interaction of the neoplastic clone with bone marrow microenvironment and chronic inflammation, all can modulate the disease phenotype, influence the MPN clinical course and therefore, might be useful therapeutic targets.
Collapse
Affiliation(s)
- Gabriel Levy
- Ludwig Institute for Cancer Research, Brussels, Belgium; SIGN Unit, de Duve Institute, Université Catholique de Louvain, Brussels, Belgium; Department of Pediatric Hematology and Oncology, Cliniques Universitaires Saint-Luc, Université Catholique de Louvain, Brussels, Belgium
| | - Cristina Mambet
- Department of Cellular and Molecular Pathology, Stefan S. Nicolau Institute of Virology, Bucharest, Romania; Department of Hematology, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Christian Pecquet
- Ludwig Institute for Cancer Research, Brussels, Belgium; SIGN Unit, de Duve Institute, Université Catholique de Louvain, Brussels, Belgium; WELBIO (Walloon Excellence in Life Sciences and Biotechnology), Brussels, Belgium
| | - Sarah Bailly
- Ludwig Institute for Cancer Research, Brussels, Belgium; SIGN Unit, de Duve Institute, Université Catholique de Louvain, Brussels, Belgium; Department of Hematology, Cliniques Universitaires Saint Luc, Université Catholique de Louvain, Brussels, Belgium
| | - Violaine Havelange
- SIGN Unit, de Duve Institute, Université Catholique de Louvain, Brussels, Belgium; Department of Hematology, Cliniques Universitaires Saint Luc, Université Catholique de Louvain, Brussels, Belgium
| | - Carmen C Diaconu
- Department of Cellular and Molecular Pathology, Stefan S. Nicolau Institute of Virology, Bucharest, Romania
| | - Stefan N Constantinescu
- Ludwig Institute for Cancer Research, Brussels, Belgium; SIGN Unit, de Duve Institute, Université Catholique de Louvain, Brussels, Belgium; WELBIO (Walloon Excellence in Life Sciences and Biotechnology), Brussels, Belgium; Ludwig Institute for Cancer Research, Nuffield Department of Medicine, Oxford University, Oxford, United Kingdom.
| |
Collapse
|
23
|
Roy A, Shrivastva S, Naseer S. In and out: Traffic and dynamics of thrombopoietin receptor. J Cell Mol Med 2021; 25:9073-9083. [PMID: 34448528 PMCID: PMC8500957 DOI: 10.1111/jcmm.16878] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 07/27/2021] [Accepted: 08/04/2021] [Indexed: 12/17/2022] Open
Abstract
Thrombopoiesis had long been a challenging area of study due to the rarity of megakaryocyte precursors in the bone marrow and the incomplete understanding of its regulatory cytokines. A breakthrough was achieved in the early 1990s with the discovery of the thrombopoietin receptor (TpoR) and its ligand thrombopoietin (TPO). This accelerated research in thrombopoiesis, including the uncovering of the molecular basis of myeloproliferative neoplasms (MPN) and the advent of drugs to treat thrombocytopenic purpura. TpoR mutations affecting its membrane dynamics or transport were increasingly associated with pathologies such as MPN and thrombocytosis. It also became apparent that TpoR affected hematopoietic stem cell (HSC) quiescence while priming hematopoietic stem cells (HSCs) towards the megakaryocyte lineage. Thorough knowledge of TpoR surface localization, dimerization, dynamics and stability is therefore crucial to understanding thrombopoiesis and related pathologies. In this review, we will discuss the mechanisms of TpoR traffic. We will focus on the recent progress in TpoR membrane dynamics and highlight the areas that remain unexplored.
Collapse
Affiliation(s)
- Anita Roy
- Kusuma School of Biological Sciences, Indian Institute of Technology, New Delhi, India
| | - Saurabh Shrivastva
- Kusuma School of Biological Sciences, Indian Institute of Technology, New Delhi, India
| | - Saadia Naseer
- Kusuma School of Biological Sciences, Indian Institute of Technology, New Delhi, India
| |
Collapse
|
24
|
Impact of Calreticulin and Its Mutants on Endoplasmic Reticulum Function in Health and Disease. PROGRESS IN MOLECULAR AND SUBCELLULAR BIOLOGY 2021. [PMID: 34050866 DOI: 10.1007/978-3-030-67696-4_8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/03/2024]
Abstract
The endoplasmic reticulum (ER) performs key cellular functions including protein synthesis, lipid metabolism and signaling. While these functions are spatially isolated in structurally distinct regions of the ER, there is cross-talk between the pathways. One vital player that is involved in ER function is the ER-resident protein calreticulin (CALR). It is a calcium ion-dependent lectin chaperone that primarily assists in glycoprotein synthesis in the ER as part of the protein quality control machinery. CALR also buffers calcium ion release and mediates other glycan-independent protein interactions. Mutations in CALR have been reported in a subset of chronic blood tumors called myeloproliferative neoplasms. The mutations consist of insertions or deletions in the CALR gene that all cause a + 1 bp shift in the reading frame and lead to a dramatic alteration of the amino acid sequence of the C-terminal domain of CALR. This alters CALR function and affects cell homeostasis. This chapter will discuss how CALR and mutant CALR affect ER health and disease.
Collapse
|
25
|
Mapping human calreticulin regions important for structural stability. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2021; 1869:140710. [PMID: 34358706 DOI: 10.1016/j.bbapap.2021.140710] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 07/24/2021] [Accepted: 08/02/2021] [Indexed: 12/28/2022]
Abstract
Calreticulin (CALR) is a highly conserved multifunctional chaperone protein primarily present in the endoplasmic reticulum, where it regulates Ca2+ homeostasis. Recently, CALR has gained special interest for its diverse functions outside the endoplasmic reticulum, including the cell surface and extracellular space. Although high-resolution structures of CALR exist, it has not yet been established how different regions and individual amino acid residues contribute to structural stability of the protein. In the present study, we have identified key residues determining the structural stability of CALR. We used a Saccharomyces cerevisiae expression system to express and purify 50 human CALR mutants, which were analysed for several parameters including secretion titer, melting temperature (Tm), stability and oligomeric state. Our results revealed the importance of a previously identified small patch of conserved surface residues, amino acids 166-187 ("cluster 2") for structural stability of the human CALR protein. Two residues, Tyr172 and Asp187, were critical for maintaining the native structure of the protein. Mutant D187A revealed a severe drop in secretion titer, it was thermally unstable, prone to degradation, and oligomer formation. Tyr172 was critical for thermal stability of CALR and interacted with the third free Cys163 residue. This illustrates an unusual thermal stability of CALR dominated by Asp187, Tyr172 and Cys163, which may interact as part of a conserved structural unit. Besides structural clusters, we found a correlation of some measured parameter values in groups of CALR mutants that cause myeloproliferative neoplasms (MPN) and in mutants that may be associated with sudden unexpected death (SUD).
Collapse
|
26
|
Bergmann AC, Kyllesbech C, Slibinskas R, Ciplys E, Højrup P, Trier NH, Houen G. Epitope Mapping of Monoclonal Antibodies to Calreticulin Reveals That Charged Amino Acids Are Essential for Antibody Binding. Antibodies (Basel) 2021; 10:antib10030031. [PMID: 34449535 PMCID: PMC8395503 DOI: 10.3390/antib10030031] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 06/25/2021] [Accepted: 07/30/2021] [Indexed: 02/01/2023] Open
Abstract
Calreticulin is a chaperone protein, which is associated with myeloproliferative diseases. In this study, we used resin-bound peptides to characterize two monoclonal antibodies (mAbs) directed to calreticulin, mAb FMC 75 and mAb 16, which both have significantly contributed to understanding the biological function of calreticulin. The antigenicity of the resin-bound peptides was determined by modified enzyme-linked immunosorbent assay. Specific binding was determined to an 8-mer epitope located in the N-terminal (amino acids 34–41) and to a 12-mer peptide located in the C-terminal (amino acids 362–373). Using truncated peptides, the epitopes were identified as TSRWIESK and DEEQRLKEEED for mAb FMC 75 and mAb 16, respectively, where, especially the charged amino acids, were found to have a central role for a stable binding. Further studies indicated that the epitope of mAb FMC 75 is assessable in the oligomeric structure of calreticulin, making this epitope a potential therapeutic target.
Collapse
Affiliation(s)
| | - Cecilie Kyllesbech
- Department of Neurology, Rigshospitalet Glostrup, 2600 Glostrup, Denmark;
| | - Rimantas Slibinskas
- Institute of Biotechnology, University of Vilnius, 01513 Vilnius, Lithuania; (R.S.); (E.C.)
| | - Evaldas Ciplys
- Institute of Biotechnology, University of Vilnius, 01513 Vilnius, Lithuania; (R.S.); (E.C.)
| | - Peter Højrup
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense, Denmark;
| | - Nicole Hartwig Trier
- Department of Neurology, Rigshospitalet Glostrup, 2600 Glostrup, Denmark;
- Correspondence: (N.H.T.); (G.H.)
| | - Gunnar Houen
- Department of Neurology, Rigshospitalet Glostrup, 2600 Glostrup, Denmark;
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense, Denmark;
- Correspondence: (N.H.T.); (G.H.)
| |
Collapse
|
27
|
Shide K. Calreticulin mutations in myeloproliferative neoplasms. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2021; 365:179-226. [PMID: 34756244 DOI: 10.1016/bs.ircmb.2021.05.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Calreticulin (CALR) is a chaperone present in the endoplasmic reticulum, which is involved in the quality control of N-glycosylated proteins and storage of calcium ions. In 2013, the C-terminal mutation in CALR was identified in half of the patients with essential thrombocythemia and primary myelofibrosis who did not have a JAK2 or MPL mutation. The results of 8 years of intensive research are changing the clinical practice associated with treating myeloproliferative neoplasms (MPNs). The presence or absence of CALR mutations and their mutation types already provide important information for diagnosis and treatment decision making. In addition, the interaction with the thrombopoietin receptor MPL, which is the main mechanism of transformation by CALR mutation, and the expression of the mutant protein on the cell surface have a great potential as targets for molecular-targeted drugs and immunotherapy. This chapter presents recent findings on the clinical significance of the CALR mutation and the molecular basis by which this mutation drives MPNs.
Collapse
Affiliation(s)
- Kotaro Shide
- Division of Haematology, Diabetes, and Endocrinology, Department of Internal Medicine, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan.
| |
Collapse
|
28
|
Aflalo A, Boyle LH. Polymorphisms in MHC class I molecules influence their interactions with components of the antigen processing and presentation pathway. Int J Immunogenet 2021; 48:317-325. [PMID: 34176210 DOI: 10.1111/iji.12546] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 05/12/2021] [Accepted: 05/24/2021] [Indexed: 10/21/2022]
Abstract
MHC class I (MHC-I) molecules undergo an intricate folding process in order to pick up antigenic peptide to present to the immune system. In recent years, the discovery of a new peptide editor for MHC-I has added an extra level of complexity in our understanding of how peptide presentation is regulated. On top of this, the incredible diversity in MHC-I molecules leads to significant variation in the interaction between MHC-I and components of the antigen processing and presentation pathway. Here, we review our current understanding regarding how polymorphisms in human leukocyte antigen class I molecules influence their interactions with key components of the antigen processing and presentation pathway. A deeper understanding of this may offer new insights regarding how apparently subtle variation in MHC-I can have a significant impact on susceptibility to disease.
Collapse
Affiliation(s)
- Aure Aflalo
- Department of Pathology, University of Cambridge, Cambridge, UK
| | - Louise H Boyle
- Department of Pathology, University of Cambridge, Cambridge, UK
| |
Collapse
|
29
|
Functional Consequences of Mutations in Myeloproliferative Neoplasms. Hemasphere 2021; 5:e578. [PMID: 34095761 PMCID: PMC8171364 DOI: 10.1097/hs9.0000000000000578] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 04/13/2021] [Indexed: 01/14/2023] Open
Abstract
Driver mutations occur in Janus kinase 2 (JAK2), thrombopoietin receptor (MPL), and calreticulin (CALR) in BCR-ABL1 negative myeloproliferative neoplasms (MPNs). From mutations leading to one amino acid substitution in JAK2 or MPL, to frameshift mutations in CALR resulting in a protein with a different C-terminus, all the mutated proteins lead to pathologic and persistent JAK2-STAT5 activation. The most prevalent mutation, JAK2 V617F, is associated with the 3 entities polycythemia vera (PV), essential thrombocythemia (ET), and myelofibrosis (MF), while CALR and MPL mutations are associated only with ET and MF. Triple negative ET and MF patients may harbor noncanonical mutations in JAK2 or MPL. One major fundamental question is whether the conformations of JAK2 V617F, MPL W515K/L/A, or CALR mutants differ from those of their wild type counterparts so that a specific treatment could target the clone carrying the mutated driver and spare physiological hematopoiesis. Of great interest, a set of epigenetic mutations can co-exist with the phenotypic driver mutations in 35%–40% of MPNs. These epigenetic mutations, such as TET2, EZH2, ASXL1, or DNMT3A mutations, promote clonal hematopoiesis and increased fitness of aged hematopoietic stem cells in both clonal hematopoiesis of indeterminate potential (CHIP) and MPNs. Importantly, the main MPN driver mutation JAK2 V617F is also associated with CHIP. Accumulation of several epigenetic and splicing mutations favors progression of MPNs to secondary acute myeloid leukemia. Another major fundamental question is how epigenetic rewiring due to these mutations interacts with persistent JAK2-STAT5 signaling. Answers to these questions are required for better therapeutic interventions aimed at preventing progression of ET and PV to MF, and transformation of these MPNs in secondary acute myeloid leukemia.
Collapse
|
30
|
Jongsma MLM, Neefjes J, Spaapen RM. Playing hide and seek: Tumor cells in control of MHC class I antigen presentation. Mol Immunol 2021; 136:36-44. [PMID: 34082257 DOI: 10.1016/j.molimm.2021.05.009] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 04/07/2021] [Accepted: 05/18/2021] [Indexed: 12/15/2022]
Abstract
MHC class I (MHC-I) molecules present a blueprint of the intracellular proteome to T cells allowing them to control infection or malignant transformation. As a response, pathogens and tumor cells often downmodulate MHC-I mediated antigen presentation to escape from immune surveillance. Although the fundamental rules of antigen presentation are known in detail, the players in this system are not saturated and new modules of regulation have recently been uncovered. Here, we update the understanding of antigen presentation by MHC-I molecules and how this can be exploited by tumors to prevent exposure of the intracellular proteome. This knowledge can provide new ways to improve immune responses against tumors and pathogens.
Collapse
Affiliation(s)
- M L M Jongsma
- Department of Cell and Chemical Biology, Oncode Institute, Leiden University Medical Center, Leiden, the Netherlands
| | - J Neefjes
- Department of Cell and Chemical Biology, Oncode Institute, Leiden University Medical Center, Leiden, the Netherlands
| | - R M Spaapen
- Department of Immunopathology, Sanquin Research, Amsterdam, the Netherlands; Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands; Cancer Center Amsterdam, Amsterdam, the Netherlands.
| |
Collapse
|
31
|
Kanduła Z, Lewandowski K. Calreticulin – a multifaced protein. POSTEP HIG MED DOSW 2021. [DOI: 10.5604/01.3001.0014.8892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Calreticulin (CALR) is a highly conserved multi-function protein that primarily localizes within
the lumen of the endoplasmic reticulum (ER). It participates in various processes in the cells,
including glycoprotein chaperoning, regulation of Ca2+ homeostasis, antigen processing and
presentation for adaptive immune response, cell adhesion/migration, cell proliferation, immunogenic
cell death, gene expression and RNA stability. The role of CALR in the assembly,
retrieval and cell surface expression of MHC class I molecules is well known. A fraction of
the total cellular CALR is localized in the cytosol, following its retro-translocation from the
ER. In the cell stress conditions, CALR is also expressed on the cell surface via an interaction
with phosphatidylserine localized on the inner leaflet of the plasma membrane. The abovementioned
mechanism is relevant for the recognition of the cells, as well as immunogenicity
and phagocytic uptake of proapoptotic and apoptotic cells.
Lastly, the presence of CALR exon 9 gene mutations was confirmed in patients with myeloproliferative
neoplasms. Their presence results in an abnormal CALR structure due to the
loss of its ER-retention sequence, CALR extra-ER localisation, the formation of a complex
with thrombopoietin receptor, and oncogenic transformation of hematopoietic stem cells. It
is also known that CALR exon 9 mutants are highly immunogenic and induce T cell response.
Despite this fact, CALR mutant positive hematopoietic cells emerge. The last phenomenon is
probably the result of the inhibition of phagocytosis of the cancer cells exposing CALR mutant
protein by dendritic cells.
Collapse
Affiliation(s)
- Zuzanna Kanduła
- Department of Hematology and Bone Marrow Transplantation, Poznań University of Medical Sciences, Poland
| | - Krzysztof Lewandowski
- Department of Hematology and Bone Marrow Transplantation, Poznań University of Medical Sciences, Poland
| |
Collapse
|
32
|
Jhunjhunwala S, Hammer C, Delamarre L. Antigen presentation in cancer: insights into tumour immunogenicity and immune evasion. Nat Rev Cancer 2021; 21:298-312. [PMID: 33750922 DOI: 10.1038/s41568-021-00339-z] [Citation(s) in RCA: 782] [Impact Index Per Article: 195.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/01/2021] [Indexed: 01/31/2023]
Abstract
Immune checkpoint blockade, which blocks inhibitory signals of T cell activation, has shown tremendous success in treating cancer, although success still remains limited to a fraction of patients. To date, clinically effective CD8+ T cell responses appear to target predominantly antigens derived from tumour-specific mutations that accumulate in cancer, also called neoantigens. Tumour antigens are displayed on the surface of cells by class I human leukocyte antigens (HLA-I). To elicit an effective antitumour response, antigen presentation has to be successful at two distinct events: first, cancer antigens have to be taken up by dendritic cells (DCs) and cross-presented for CD8+ T cell priming. Second, the antigens have to be directly presented by the tumour for recognition by primed CD8+ T cells and killing. Tumours exploit multiple escape mechanisms to evade immune recognition at both of these steps. Here, we review the tumour-derived factors modulating DC function, and we summarize evidence of immune evasion by means of quantitative modulation or qualitative alteration of the antigen repertoire presented on tumours. These mechanisms include modulation of antigen expression, HLA-I surface levels, alterations in the antigen processing and presentation machinery in tumour cells. Lastly, as complete abrogation of antigen presentation can lead to natural killer (NK) cell-mediated tumour killing, we also discuss how tumours can harbour antigen presentation defects and still evade NK cell recognition.
Collapse
|
33
|
Venkatesan A, Geng J, Kandarpa M, Wijeyesakere SJ, Bhide A, Talpaz M, Pogozheva ID, Raghavan M. Mechanism of mutant calreticulin-mediated activation of the thrombopoietin receptor in cancers. J Cell Biol 2021; 220:212031. [PMID: 33909030 PMCID: PMC8085772 DOI: 10.1083/jcb.202009179] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 02/10/2021] [Accepted: 03/17/2021] [Indexed: 12/21/2022] Open
Abstract
Myeloproliferative neoplasms (MPNs) are frequently driven by mutations within the C-terminal domain (C-domain) of calreticulin (CRT). CRTDel52 and CRTIns5 are recurrent mutations. Oncogenic transformation requires both mutated CRT and the thrombopoietin receptor (Mpl), but the molecular mechanism of CRT-mediated constitutive activation of Mpl is unknown. We show that the acquired C-domain of CRTDel52 mediates both Mpl binding and disulfide-linked CRTDel52 dimerization. Cysteine mutations within the novel C-domain (C400A and C404A) and the conserved N-terminal domain (N-domain; C163A) of CRTDel52 are required to reduce disulfide-mediated dimers and multimers of CRTDel52. Based on these data and published structures of CRT oligomers, we identify an N-domain dimerization interface relevant to both WT CRT and CRTDel52. Elimination of disulfide bonds and ionic interactions at both N-domain and C-domain dimerization interfaces is required to abrogate the ability of CRTDel52 to mediate cell proliferation via Mpl. Thus, MPNs exploit a natural dimerization interface of CRT combined with C-domain gain of function to achieve cell transformation.
Collapse
Affiliation(s)
- Arunkumar Venkatesan
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI
| | - Jie Geng
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI
| | - Malathi Kandarpa
- Department of Internal Medicine/Division of Hematology/Oncology, University of Michigan Rogel Cancer Center, Ann Arbor, MI
| | | | - Ashwini Bhide
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI
| | - Moshe Talpaz
- Department of Internal Medicine/Division of Hematology/Oncology, University of Michigan Rogel Cancer Center, Ann Arbor, MI
| | - Irina D Pogozheva
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, MI
| | - Malini Raghavan
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI
| |
Collapse
|
34
|
Handlos Grauslund J, Holmström MO, Jørgensen NG, Klausen U, Weis-Banke SE, El Fassi D, Schöllkopf C, Clausen MB, Gjerdrum LMR, Breinholt MF, Kjeldsen JW, Hansen M, Koschmieder S, Chatain N, Novotny GW, Petersen J, Kjær L, Skov V, Met Ö, Svane IM, Hasselbalch HC, Andersen MH. Therapeutic Cancer Vaccination With a Peptide Derived From the Calreticulin Exon 9 Mutations Induces Strong Cellular Immune Responses in Patients With CALR-Mutant Chronic Myeloproliferative Neoplasms. Front Oncol 2021; 11:637420. [PMID: 33718228 PMCID: PMC7952976 DOI: 10.3389/fonc.2021.637420] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 01/18/2021] [Indexed: 12/12/2022] Open
Abstract
Background The calreticulin (CALR) exon 9 mutations that are identified in 20% of patients with Philadelphia chromosome negative chronic myeloproliferative neoplasms (MPN) generate immunogenic antigens. Thus, therapeutic cancer vaccination against mutant CALR could be a new treatment modality in CALR-mutant MPN. Methods The safety and efficacy of vaccination with the peptide CALRLong36 derived from the CALR exon 9 mutations was tested in a phase I clinical vaccination trial with montanide as adjuvant. Ten patients with CALRmut MPN were included in the trial and received 15 vaccines over the course of one year. The primary end point was evaluation of safety and toxicity of the vaccine. Secondary endpoint was assessment of the immune response to the vaccination epitope (www.clinicaltrials.gov identifier NCT03566446). Results Patients had a median age of 59.5 years and a median disease duration of 6.5 years. All patients received the intended 15 vaccines, and the vaccines were deemed safe and tolerable as only two grade three AE were detected, and none of these were considered to be related to the vaccine. A decline in platelet counts relative to the platelets counts at baseline was detected during the first 100 days, however this did not translate into neither a clinical nor a molecular response in any of the patients. Immunomonitoring revealed that four of 10 patients had an in vitro interferon (IFN)-γ ELISPOT response to the CALRLong36 peptide at baseline, and four additional patients displayed a response in ELISPOT upon receiving three or more vaccines. The amplitude of the immune response increased during the entire vaccination schedule for patients with essential thrombocythemia. In contrast, the immune response in patients with primary myelofibrosis did not increase after three vaccines. Conclusion Therapeutic cancer vaccination with peptide vaccines derived from mutant CALR with montanide as an adjuvant, is safe and tolerable. The vaccines did not induce any clinical responses. However, the majority of patients displayed a marked T-cell response to the vaccine upon completion of the trial. This suggests that vaccines directed against mutant CALR may be used with other cancer therapeutic modalities to enhance the anti-tumor immune response.
Collapse
Affiliation(s)
- Jacob Handlos Grauslund
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
| | - Morten Orebo Holmström
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
| | - Nicolai Grønne Jørgensen
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
| | - Uffe Klausen
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
| | - Stine Emilie Weis-Banke
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
| | - Daniel El Fassi
- Department of Hematology, Copenhagen University Hospital, Herlev, Denmark.,Department of Medicine, Copenhagen University, Copenhagen, Denmark
| | - Claudia Schöllkopf
- Department of Hematology, Copenhagen University Hospital, Herlev, Denmark
| | - Mette Borg Clausen
- Department of Hematology, Copenhagen University Hospital, Herlev, Denmark
| | | | | | - Julie Westerlin Kjeldsen
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
| | - Morten Hansen
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
| | - Steffen Koschmieder
- Department of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation, Faculty of Medicine, RWTH Aachen University, Aachen, Germany
| | - Nicolas Chatain
- Department of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation, Faculty of Medicine, RWTH Aachen University, Aachen, Germany
| | - Guy Wayne Novotny
- Department of Hematology, Copenhagen University Hospital, Herlev, Denmark
| | - Jesper Petersen
- Department of Hematology, Copenhagen University Hospital, Herlev, Denmark
| | - Lasse Kjær
- Department of Hematology, Zealand University Hospital, Roskilde, Denmark
| | - Vibe Skov
- Department of Hematology, Zealand University Hospital, Roskilde, Denmark
| | - Özcan Met
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, Herlev, Denmark.,Institute for Immunology and Microbiology, Copenhagen University, Copenhagen, Denmark
| | - Inge Marie Svane
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
| | | | - Mads Hald Andersen
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, Herlev, Denmark.,Institute for Immunology and Microbiology, Copenhagen University, Copenhagen, Denmark
| |
Collapse
|
35
|
Guijarro-Hernández A, Vizmanos JL. A Broad Overview of Signaling in Ph-Negative Classic Myeloproliferative Neoplasms. Cancers (Basel) 2021; 13:cancers13050984. [PMID: 33652860 PMCID: PMC7956519 DOI: 10.3390/cancers13050984] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 02/19/2021] [Accepted: 02/22/2021] [Indexed: 12/11/2022] Open
Abstract
Simple Summary There is growing evidence that Ph-negative myeloproliferative neoplasms are disorders in which multiple signaling pathways are significantly disturbed. The heterogeneous phenotypes observed among patients have highlighted the importance of having a comprehensive knowledge of the molecular mechanisms behind these diseases. This review aims to show a broad overview of the signaling involved in myeloproliferative neoplasms (MPNs) and other processes that can modify them, which could be helpful to better understand these diseases and develop more effective targeted treatments. Abstract Ph-negative myeloproliferative neoplasms (polycythemia vera (PV), essential thrombocythemia (ET) and primary myelofibrosis (PMF)) are infrequent blood cancers characterized by signaling aberrations. Shortly after the discovery of the somatic mutations in JAK2, MPL, and CALR that cause these diseases, researchers extensively studied the aberrant functions of their mutant products. In all three cases, the main pathogenic mechanism appears to be the constitutive activation of JAK2/STAT signaling and JAK2-related pathways (MAPK/ERK, PI3K/AKT). However, some other non-canonical aberrant mechanisms derived from mutant JAK2 and CALR have also been described. Moreover, additional somatic mutations have been identified in other genes that affect epigenetic regulation, tumor suppression, transcription regulation, splicing and other signaling pathways, leading to the modification of some disease features and adding a layer of complexity to their molecular pathogenesis. All of these factors have highlighted the wide variety of cellular processes and pathways involved in the pathogenesis of MPNs. This review presents an overview of the complex signaling behind these diseases which could explain, at least in part, their phenotypic heterogeneity.
Collapse
Affiliation(s)
- Ana Guijarro-Hernández
- Department of Biochemistry and Genetics, School of Sciences, University of Navarra, 31008 Pamplona, Spain;
| | - José Luis Vizmanos
- Department of Biochemistry and Genetics, School of Sciences, University of Navarra, 31008 Pamplona, Spain;
- Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain
- Correspondence:
| |
Collapse
|
36
|
Mpakali A, Stratikos E. The Role of Antigen Processing and Presentation in Cancer and the Efficacy of Immune Checkpoint Inhibitor Immunotherapy. Cancers (Basel) 2021; 13:E134. [PMID: 33406696 PMCID: PMC7796214 DOI: 10.3390/cancers13010134] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Revised: 12/28/2020] [Accepted: 12/30/2020] [Indexed: 02/07/2023] Open
Abstract
Recent clinical successes of cancer immunotherapy using immune checkpoint inhibitors (ICIs) are rapidly changing the landscape of cancer treatment. Regardless of initial impressive clinical results though, the therapeutic benefit of ICIs appears to be limited to a subset of patients and tumor types. Recent analyses have revealed that the potency of ICI therapies depends on the efficient presentation of tumor-specific antigens by cancer cells and professional antigen presenting cells. Here, we review current knowledge on the role of antigen presentation in cancer. We focus on intracellular antigen processing and presentation by Major Histocompatibility class I (MHCI) molecules and how it can affect cancer immune evasion. Finally, we discuss the pharmacological tractability of manipulating intracellular antigen processing as a complementary approach to enhance tumor immunogenicity and the effectiveness of ICI immunotherapy.
Collapse
Affiliation(s)
- Anastasia Mpakali
- National Centre for Scientific Research Demokritos, Agia Paraskevi, 15341 Athens, Greece
| | - Efstratios Stratikos
- National Centre for Scientific Research Demokritos, Agia Paraskevi, 15341 Athens, Greece
- Laboratory of Biochemistry, Department of Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis Zographou, 15784 Athens, Greece
| |
Collapse
|
37
|
Roles of Calreticulin in Protein Folding, Immunity, Calcium Signaling and Cell Transformation. PROGRESS IN MOLECULAR AND SUBCELLULAR BIOLOGY 2021; 59:145-162. [PMID: 34050865 DOI: 10.1007/978-3-030-67696-4_7] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The endoplasmic reticulum (ER) is an organelle that mediates the proper folding and assembly of proteins destined for the cell surface, the extracellular space and subcellular compartments such as the lysosomes. The ER contains a wide range of molecular chaperones to handle the folding requirements of a diverse set of proteins that traffic through this compartment. The lectin-like chaperones calreticulin and calnexin are an important class of structurally-related chaperones relevant for the folding and assembly of many N-linked glycoproteins. Despite the conserved mechanism of action of these two chaperones in nascent protein recognition and folding, calreticulin has unique functions in cellular calcium signaling and in the immune response. The ER-related functions of calreticulin in the assembly of major histocompatibility complex (MHC) class I molecules are well-studied and provide many insights into the modes of substrate and co-chaperone recognition by calreticulin. Calreticulin is also detectable on the cell surface under some conditions, where it induces the phagocytosis of apoptotic cells. Furthermore, mutations of calreticulin induce cell transformation in myeloproliferative neoplasms (MPN). Studies of the functions of the mutant calreticulin in cell transformation and immunity have provided many insights into the normal biology of calreticulin, which are discussed.
Collapse
|
38
|
Benlabiod C, Cacemiro MDC, Nédélec A, Edmond V, Muller D, Rameau P, Touchard L, Gonin P, Constantinescu SN, Raslova H, Villeval JL, Vainchenker W, Plo I, Marty C. Calreticulin del52 and ins5 knock-in mice recapitulate different myeloproliferative phenotypes observed in patients with MPN. Nat Commun 2020; 11:4886. [PMID: 32985500 PMCID: PMC7522233 DOI: 10.1038/s41467-020-18691-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 09/04/2020] [Indexed: 12/20/2022] Open
Abstract
Somatic mutations in the calreticulin (CALR) gene are associated with approximately 30% of essential thrombocythemia (ET) and primary myelofibrosis (PMF). CALR mutations, including the two most frequent 52 bp deletion (del52) and 5 bp insertion (ins5), induce a frameshift to the same alternative reading frame generating new C-terminal tails. In patients, del52 and ins5 induce two phenotypically distinct myeloproliferative neoplasms (MPNs). They are equally found in ET, but del52 is more frequent in PMF. We generated heterozygous and homozygous conditional inducible knock-in (KI) mice expressing a chimeric murine CALR del52 or ins5 with the human mutated C-terminal tail to investigate their pathogenic effects on hematopoiesis. Del52 induces greater phenotypic changes than ins5 including thrombocytosis, leukocytosis, splenomegaly, bone marrow hypocellularity, megakaryocytic lineage amplification, expansion and competitive advantage of the hematopoietic stem cell compartment. Homozygosity amplifies these features, suggesting a distinct contribution of homozygous clones to human MPNs. Moreover, homozygous del52 KI mice display features of a penetrant myelofibrosis-like disorder with extramedullary hematopoiesis linked to splenomegaly, megakaryocyte hyperplasia and the presence of reticulin fibers. Overall, modeling del52 and ins5 mutations in mice successfully recapitulates the differences in phenotypes observed in patients. Calreticulin del52 and ins5 mutations induce two phenotypically distinct myeloproliferative neoplasms in patients. Here the authors show that modeling these mutations in knock-in mice recapitulate the two diseases and highlight how they impact the different hematopoietic compartments.
Collapse
Affiliation(s)
- Camélia Benlabiod
- INSERM, UMR 1287, Gustave Roussy, Villejuif, France.,Université Paris-Saclay, UMR 1287, Gustave Roussy, Villejuif, France.,Gustave Roussy, UMR 1287, Villejuif, France
| | - Maira da Costa Cacemiro
- INSERM, UMR 1287, Gustave Roussy, Villejuif, France.,Université Paris-Saclay, UMR 1287, Gustave Roussy, Villejuif, France.,Gustave Roussy, UMR 1287, Villejuif, France.,Department of Clinical Analysis, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo-USP, Ribeirão Preto, São Paulo, Brazil
| | - Audrey Nédélec
- Ludwig Institute for Cancer Research, Brussels, Belgium.,de Duve Institute, Walloon Excellence in Life Sciences and Biotechnology (WELBIO), Université catholique de Louvain, Brussels, Belgium
| | - Valérie Edmond
- INSERM, UMR 1287, Gustave Roussy, Villejuif, France.,Université Paris-Saclay, UMR 1287, Gustave Roussy, Villejuif, France.,Gustave Roussy, UMR 1287, Villejuif, France
| | - Delphine Muller
- INSERM, UMR 1287, Gustave Roussy, Villejuif, France.,Université Paris-Saclay, UMR 1287, Gustave Roussy, Villejuif, France.,Gustave Roussy, UMR 1287, Villejuif, France
| | - Philippe Rameau
- Integrated Biology Core Facility, Gustave Roussy, Villejuif, France
| | - Laure Touchard
- Preclinical Research Plateform, Unité Mixte de Service AMMICA 3655/US 23, Gustave Roussy, Villejuif, France
| | - Patrick Gonin
- Preclinical Research Plateform, Unité Mixte de Service AMMICA 3655/US 23, Gustave Roussy, Villejuif, France
| | - Stefan N Constantinescu
- Ludwig Institute for Cancer Research, Brussels, Belgium.,de Duve Institute, Walloon Excellence in Life Sciences and Biotechnology (WELBIO), Université catholique de Louvain, Brussels, Belgium
| | - Hana Raslova
- INSERM, UMR 1287, Gustave Roussy, Villejuif, France.,Université Paris-Saclay, UMR 1287, Gustave Roussy, Villejuif, France.,Gustave Roussy, UMR 1287, Villejuif, France
| | - Jean-Luc Villeval
- INSERM, UMR 1287, Gustave Roussy, Villejuif, France.,Université Paris-Saclay, UMR 1287, Gustave Roussy, Villejuif, France.,Gustave Roussy, UMR 1287, Villejuif, France
| | - William Vainchenker
- INSERM, UMR 1287, Gustave Roussy, Villejuif, France.,Université Paris-Saclay, UMR 1287, Gustave Roussy, Villejuif, France.,Gustave Roussy, UMR 1287, Villejuif, France
| | - Isabelle Plo
- INSERM, UMR 1287, Gustave Roussy, Villejuif, France.,Université Paris-Saclay, UMR 1287, Gustave Roussy, Villejuif, France.,Gustave Roussy, UMR 1287, Villejuif, France
| | - Caroline Marty
- INSERM, UMR 1287, Gustave Roussy, Villejuif, France. .,Université Paris-Saclay, UMR 1287, Gustave Roussy, Villejuif, France. .,Gustave Roussy, UMR 1287, Villejuif, France.
| |
Collapse
|
39
|
Zaitoua AJ, Kaur A, Raghavan M. Variations in MHC class I antigen presentation and immunopeptidome selection pathways. F1000Res 2020; 9. [PMID: 33014341 PMCID: PMC7525337 DOI: 10.12688/f1000research.26935.1] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/18/2020] [Indexed: 12/12/2022] Open
Abstract
Major histocompatibility class I (MHC-I) proteins mediate immunosurveillance against pathogens and cancers by presenting antigenic or mutated peptides to antigen receptors of CD8+ T cells and by engaging receptors of natural killer (NK) cells. In humans, MHC-I molecules are highly polymorphic. MHC-I variations permit the display of thousands of distinct peptides at the cell surface. Recent mass spectrometric studies have revealed unique and shared characteristics of the peptidomes of individual MHC-I variants. The cell surface expression of MHC-I–peptide complexes requires the functions of many intracellular assembly factors, including the transporter associated with antigen presentation (TAP), tapasin, calreticulin, ERp57, TAP-binding protein related (TAPBPR), endoplasmic reticulum aminopeptidases (ERAPs), and the proteasomes. Recent studies provide important insights into the structural features of these factors that govern MHC-I assembly as well as the mechanisms underlying peptide exchange. Conformational sensing of MHC-I molecules mediates the quality control of intracellular MHC-I assembly and contributes to immune recognition by CD8 at the cell surface. Recent studies also show that several MHC-I variants can follow unconventional assembly routes to the cell surface, conferring selective immune advantages that can be exploited for immunotherapy.
Collapse
Affiliation(s)
- Anita J Zaitoua
- Department of Microbiology and Immunology, Michigan Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Amanpreet Kaur
- Department of Microbiology and Immunology, Michigan Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Malini Raghavan
- Department of Microbiology and Immunology, Michigan Medicine, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
40
|
Abstract
Calreticulin (CALR) is an endoplasmic reticulum (ER)-resident protein involved in a spectrum of cellular processes. In healthy cells, CALR operates as a chaperone and Ca2+ buffer to assist correct protein folding within the ER. Besides favoring the maintenance of cellular proteostasis, these cell-intrinsic CALR functions support Ca2+-dependent processes, such as adhesion and integrin signaling, and ensure normal antigen presentation on MHC Class I molecules. Moreover, cancer cells succumbing to immunogenic cell death (ICD) expose CALR on their surface, which promotes the uptake of cell corpses by professional phagocytes and ultimately supports the initiation of anticancer immunity. Thus, loss-of-function CALR mutations promote oncogenesis not only as they impair cellular homeostasis in healthy cells, but also as they compromise natural and therapy-driven immunosurveillance. However, the prognostic impact of total or membrane-exposed CALR levels appears to vary considerably with cancer type. For instance, while genetic CALR defects promote pre-neoplastic myeloproliferation, patients with myeloproliferative neoplasms bearing CALR mutations often experience improved overall survival as compared to patients bearing wild-type CALR. Here, we discuss the context-dependent impact of CALR on malignant transformation, tumor progression and response to cancer therapy.
Collapse
|
41
|
Kepp O, Liu P, Zhao L, Plo I, Kroemer G. Surface-exposed and soluble calreticulin: conflicting biomarkers for cancer prognosis. Oncoimmunology 2020; 9:1792037. [PMID: 32923154 PMCID: PMC7458660 DOI: 10.1080/2162402x.2020.1792037] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Increased exposure of calreticulin (CALR) on malignant cells is associated with therapy-relevant adaptive immune responses and superior therapeutic outcome in solid tumors and haemato-oncological diseases, because surface-exposed CALR acts as an ‘eat-me’ signal facilitating the phagocytosis of stressed and dying cancer cells by immature dendritic cells, thus favoring antitumor immune responses. On the contrary, mutations of the CALR gene that cause the omission of the C-terminal KDEL endoplasmic reticulum retention motif from CALR protein, resulting in its secretion from cells, act as oncogenic drivers in myeloproliferative neoplasms via the autocrine activation of the thrombopoietin receptor. We recently showed that soluble CALR inhibited the phagocytosis of cancer cells by dendritic cells, thus dampening anticancer immune responses. Furthermore, systemic elevations of soluble CALR that is secreted from tumors or that is artificially supplied by injection of the recombinant protein decreased the efficacy of immunotherapy. Thus, depending on its location, CALR can have immunostimulatory or immunosuppressive functions.
Collapse
Affiliation(s)
- Oliver Kepp
- Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, INSERM UMR1138, Centre de Recherche des Cordeliers, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Villejuif, France
| | - Peng Liu
- Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, INSERM UMR1138, Centre de Recherche des Cordeliers, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Villejuif, France
| | - Liwei Zhao
- Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, INSERM UMR1138, Centre de Recherche des Cordeliers, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Villejuif, France
| | - Isabelle Plo
- INSERM UMR1287, Gustave Roussy Cancer Center, Villejuif, France
| | - Guido Kroemer
- Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, INSERM UMR1138, Centre de Recherche des Cordeliers, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Villejuif, France
- Suzhou Institute for Systems Medicine, Chinese Academy of Medical Sciences, Suzhou, China
- Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France
- Karolinska Institutet, Department of Women's and Children's Health, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
42
|
Holmström MO, Hasselbalch HC, Andersen MH. Cancer Immune Therapy for Philadelphia Chromosome-Negative Chronic Myeloproliferative Neoplasms. Cancers (Basel) 2020; 12:E1763. [PMID: 32630667 PMCID: PMC7407874 DOI: 10.3390/cancers12071763] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 06/19/2020] [Accepted: 06/29/2020] [Indexed: 02/07/2023] Open
Abstract
Philadelphia chromosome-negative chronic myeloproliferative neoplasms (MPN) are neoplastic diseases of the hematopoietic stem cells in the bone marrow. MPN are characterized by chronic inflammation and immune dysregulation. Of interest, the potent immunostimulatory cytokine interferon-α has been used to treat MPN for decades. A deeper understanding of the anti-cancer immune response and of the different immune regulatory mechanisms in patients with MPN has paved the way for an increased perception of the potential of cancer immunotherapy in MPN. Therapeutic vaccination targeting the driver mutations in MPN is one recently described potential new treatment modality. Furthermore, T cells can directly react against regulatory immune cells because they recognize proteins like arginase and programmed death ligand 1 (PD-L1). Therapeutic vaccination with arginase or PD-L1 therefore offers a novel way to directly affect immune inhibitory pathways, potentially altering tolerance to tumor antigens like mutant CALR and mutant JAK2. Other therapeutic options that could be used in concert with therapeutic cancer vaccines are immune checkpoint-blocking antibodies and interferon-α. For more advanced MPN, adoptive cellular therapy is a potential option that needs more preclinical investigation. In this review, we summarize current knowledge about the immune system in MPN and discuss the many opportunities for anti-cancer immunotherapy in patients with MPN.
Collapse
Affiliation(s)
- Morten Orebo Holmström
- National Center for Cancer Immune Therapy, Department of Oncology, Herlev University Hospital, DK-2730 Herlev, Denmark;
| | | | - Mads Hald Andersen
- National Center for Cancer Immune Therapy, Department of Oncology, Herlev University Hospital, DK-2730 Herlev, Denmark;
- Department of Immunology and Microbiology, University of Copenhagen, DK-2200 Copenhagen N, Denmark
| |
Collapse
|
43
|
Edahiro Y, Araki M, Komatsu N. Mechanism underlying the development of myeloproliferative neoplasms through mutant calreticulin. Cancer Sci 2020; 111:2682-2688. [PMID: 32462673 PMCID: PMC7419020 DOI: 10.1111/cas.14503] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 05/12/2020] [Accepted: 05/22/2020] [Indexed: 01/14/2023] Open
Abstract
Deregulation of cytokine signaling is frequently associated with various pathological conditions, including malignancies. In patients with myeloproliferative neoplasms (MPNs), recurrent somatic mutations in the calreticulin (CALR) gene, which encodes a molecular chaperone that resides in the endoplasmic reticulum, have been reported. Studies have defined mutant CALR as an oncogene promoting the development of MPN, and deciphered a novel molecular mechanism by which mutant CALR constitutively activates thrombopoietin receptor MPL and its downstream molecules to induce cellular transformation. The mechanism of interaction and activation of MPL by mutant CALR is unique, not only due to the latter forming a homomultimeric complex through a novel mutant‐specific sequence generated by frameshift mutation, but also for its ability to interact with immature asparagine‐linked glycan for eventual engagement with immature MPL in the endoplasmic reticulum. The complex formed between mutant CALR and MPL is then transported to the cell surface, where it induces constitutive activation of downstream kinase JAK2 bound to MPL. Refined structural and cell biological studies can provide an in‐depth understanding of this unusual mechanism of receptor activation by a mutant molecular chaperone. Mutant CALR is also involved in modulation of the immune response, transcription, and intracellular homeostasis, which could contribute to the development of MPN. In the present article, we comprehensively review the current understanding of the underlying molecular mechanisms for mutant molecular chaperone‐induced cellular transformation.
Collapse
Affiliation(s)
- Yoko Edahiro
- Department of Hematology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Marito Araki
- Department of Transfusion Medicine and Stem Cell Regulation, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Norio Komatsu
- Department of Hematology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| |
Collapse
|
44
|
Penter L, Wu CJ. Personal tumor antigens in blood malignancies: genomics-directed identification and targeting. J Clin Invest 2020; 130:1595-1607. [PMID: 31985488 PMCID: PMC7108890 DOI: 10.1172/jci129209] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Hematological malignancies have long been at the forefront of the development of novel immune-based treatment strategies. The earliest successful efforts originated from the extensive body of work in the field of allogeneic hematopoietic stem cell transplantation. These efforts laid the foundation for the recent exciting era of cancer immunotherapy, which includes immune checkpoint blockade, personal neoantigen vaccines, and adoptive T cell transfer. At the heart of the specificity of these novel strategies is the recognition of target antigens presented by malignant cells to T cells. Here, we review the advances in systematic identification of minor histocompatibility antigens and neoantigens arising from personal somatic alterations or recurrent driver mutations. These exciting efforts pave the path for the implementation of personalized combinatorial cancer therapy.
Collapse
Affiliation(s)
- Livius Penter
- Department of Hematology, Oncology, and Tumor Immunology, Charité – Universitätsmedizin Berlin (CVK), Berlin, Germany
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Catherine J. Wu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Broad Institute, Massachusetts Institute of Technology and Harvard University, Cambridge, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
- Department of Medicine, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| |
Collapse
|
45
|
Mutant calreticulin in myeloproliferative neoplasms. Blood 2020; 134:2242-2248. [PMID: 31562135 DOI: 10.1182/blood.2019000622] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Accepted: 09/19/2019] [Indexed: 01/03/2023] Open
Abstract
Recurrent mutations in calreticulin are present in ∼20% of patients with myeloproliferative neoplasms (MPNs). Since its discovery in 2013, we now have a more precise understanding of how mutant CALR, an endoplasmic reticulum chaperone protein, activates the JAK/STAT signaling pathway via a pathogenic binding interaction with the thrombopoietin receptor MPL to induce MPNs. In this Spotlight article, we review the current understanding of the biology underpinning mutant CALR-driven MPNs, discuss clinical implications, and highlight future therapeutic approaches.
Collapse
|
46
|
Kaur A, Raghavan M. A Calreticulin Tail: C-terminal Mutants of Calreticulin Allow Cancer Cells to Evade Phagocytosis. Mol Cell 2020; 77:683-685. [PMID: 32084350 PMCID: PMC7594649 DOI: 10.1016/j.molcel.2020.01.024] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
In the current issue of Molecular Cell, Liu et al. (2020) show that the secretion of cancer-linked forms of mutant calreticulin allow cancer cells to escape protective immune responses induced by chemotherapeutic and immunotherapeutic drugs, thereby promoting tumor growth.
Collapse
Affiliation(s)
- Amanpreet Kaur
- Department of Microbiology and Immunology, Michigan Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Malini Raghavan
- Department of Microbiology and Immunology, Michigan Medicine, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
47
|
Trowitzsch S, Tampé R. Multifunctional Chaperone and Quality Control Complexes in Adaptive Immunity. Annu Rev Biophys 2020; 49:135-161. [PMID: 32004089 DOI: 10.1146/annurev-biophys-121219-081643] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The fundamental process of adaptive immunity relies on the differentiation of self from nonself. Nucleated cells are continuously monitored by effector cells of the immune system, which police the peptide status presented via cell surface molecules. Recent integrative structural approaches have provided insights toward our understanding of how sophisticated cellular machineries shape such hierarchical immune surveillance. Biophysical and structural achievements were invaluable for defining the interconnection of many key factors during antigen processing and presentation, and helped to solve several conundrums that persisted for many years. In this review, we illuminate the numerous quality control machineries involved in different steps during the maturation of major histocompatibility complex class I (MHC I) proteins, from their synthesis in the endoplasmic reticulum to folding and trafficking via the secretory pathway, optimization of antigenic cargo, final release to the cell surface, and engagement with their cognate receptors on cytotoxic T lymphocytes.
Collapse
Affiliation(s)
- Simon Trowitzsch
- Institute of Biochemistry, Biocenter, Goethe University Frankfurt, 60438 Frankfurt am Main, Germany; ,
| | - Robert Tampé
- Institute of Biochemistry, Biocenter, Goethe University Frankfurt, 60438 Frankfurt am Main, Germany; ,
| |
Collapse
|
48
|
The role of calreticulin mutations in myeloproliferative neoplasms. Int J Hematol 2019; 111:200-205. [DOI: 10.1007/s12185-019-02800-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 12/09/2019] [Accepted: 12/10/2019] [Indexed: 12/11/2022]
|
49
|
Cruz P, Sosoniuk-Roche E, Maldonado I, Torres CG, Ferreira A. Trypanosoma cruzi calreticulin: In vitro modulation of key immunogenic markers of both canine tumors and relevant immune competent cells. Immunobiology 2019; 225:151892. [PMID: 31837774 DOI: 10.1016/j.imbio.2019.12.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 12/01/2019] [Accepted: 12/01/2019] [Indexed: 10/25/2022]
Abstract
Recombinant calreticulin from Trypanosoma cruzi (rTcCalr), the parasite responsible for Chagas' disease, binds to Canine Transmissible Venereal Tumor (CTVT) cells from primary cultures and to a canine mammary carcinoma cell line. A Complement-binding assay indicated that interaction of the first component C1q with these tumor cells operated independently of the rTcCalr-presence. This apparent independence could be explained by the important structural similarities that exist among rTcCarl, endogenous normal canine and/or mutated calreticulins present in several types of cancer. In phagocytosis assays, tumor cells treated with rTcCalr were readily engulfed by macrophages and, co-cultured with DCs, accelerated their maturation. In addition, DCs maturation, induced by tumor cells co-cultured with rTcCalr, activated T cells more efficiently than DCs, treated or not with LPS. In an apparent paradox, a decrease in MHC Class I expression was observed when these tumor cells were co-cultivated with rTcCalr. This decrease may be related to a down regulation signaling promoting the rescue of MHC I. Possibly, these in vitro assays may be valid correlates of in vivo sceneries. Based on these results, we propose that rTcCalr improves in vitro the immunogenicity of two widely different tumor cell lines, thus suggesting that the interesting properties of rTcCalr to boost immune responses warrant future studies.
Collapse
Affiliation(s)
- P Cruz
- Laboratory of Immunology of Microbial Aggression, Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, 8380453, Chile; Laboratory of Biomedicine and Regenerative Medicine, Department of Clinical Sciences, Faculty of Veterinary and Animal Sciences, University of Chile, Santiago, 8820808, Chile
| | - E Sosoniuk-Roche
- Laboratory of Immunology of Microbial Aggression, Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, 8380453, Chile
| | - I Maldonado
- Laboratory of Immunology of Microbial Aggression, Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, 8380453, Chile
| | - C G Torres
- Laboratory of Biomedicine and Regenerative Medicine, Department of Clinical Sciences, Faculty of Veterinary and Animal Sciences, University of Chile, Santiago, 8820808, Chile.
| | - A Ferreira
- Laboratory of Immunology of Microbial Aggression, Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, 8380453, Chile.
| |
Collapse
|
50
|
The Expression of Myeloproliferative Neoplasm-Associated Calreticulin Variants Depends on the Functionality of ER-Associated Degradation. Cancers (Basel) 2019; 11:cancers11121921. [PMID: 31810292 PMCID: PMC6966542 DOI: 10.3390/cancers11121921] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 11/26/2019] [Accepted: 11/28/2019] [Indexed: 12/15/2022] Open
Abstract
Background: Mutations in CALR observed in myeloproliferative neoplasms (MPN) were recently shown to be pathogenic via their interaction with MPL and the subsequent activation of the Janus Kinase – Signal Transducer and Activator of Transcription (JAK-STAT) pathway. However, little is known on the impact of those variant CALR proteins on endoplasmic reticulum (ER) homeostasis. Methods: The impact of the expression of Wild Type (WT) or mutant CALR on ER homeostasis was assessed by quantifying the expression level of Unfolded Protein Response (UPR) target genes, splicing of X-box Binding Protein 1 (XBP1), and the expression level of endogenous lectins. Pharmacological and molecular (siRNA) screens were used to identify mechanisms involved in CALR mutant proteins degradation. Coimmunoprecipitations were performed to define more precisely actors involved in CALR proteins disposal. Results: We showed that the expression of CALR mutants alters neither ER homeostasis nor the sensitivity of hematopoietic cells towards ER stress-induced apoptosis. In contrast, the expression of CALR variants is generally low because of a combination of secretion and protein degradation mechanisms mostly mediated through the ER-Associated Degradation (ERAD)-proteasome pathway. Moreover, we identified a specific ERAD network involved in the degradation of CALR variants. Conclusions: We propose that this ERAD network could be considered as a potential therapeutic target for selectively inhibiting CALR mutant-dependent proliferation associated with MPN, and therefore attenuate the associated pathogenic outcomes.
Collapse
|