1
|
Hsu HT, Lin YM, Hsing MT, Yeh KT, Lu JW, Yang SF. Correlation Between Low Cytoplasmic Expression of XBP1 and the Likelihood of Surviving Hepatocellular Carcinoma. In Vivo 2024; 38:1316-1324. [PMID: 38688649 PMCID: PMC11059868 DOI: 10.21873/invivo.13571] [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: 12/18/2023] [Revised: 01/23/2024] [Accepted: 01/31/2024] [Indexed: 05/02/2024]
Abstract
BACKGROUND/AIM Our objectives in this study were to (i) evaluate the clinical significance of X-box-binding protein 1 (XBP1) expression in cases of hepatocellular carcinoma (HCC) and (ii) assess the potential of XBP1 to be used as a prognostic biomarker. PATIENTS AND METHODS The expression of XBP1 protein in 267 HCC tissue specimens was measured using immunohistochemistry in order to characterize the associations among XBP1 expression, clinicopathological factors and survival outcomes. Survival analysis using follow-up data was used to assess the prognostic value of XBP1 in cases of HCC. Immunohistochemistry revealed a significant decrease in cytoplasmic XBP1 protein expression in HCC tumor tissue. RESULTS Immunoreactivity results showed that low cytoplasmic XBP1 expression was significantly associated with vascular invasion, as well as poor 5-year overall survival and long-term disease-specific (DSS) and disease-free (DFS) survival rates. Kaplan-Meier survival curves further confirmed a significant association between low cytoplasmic XBP1 protein expression and poor DSS and DFS. Univariate and multivariate analyses revealed that XBP1 expression, tumor differentiation, vascular invasion, tumor stage, and the rate of recurrence were linked to DSS, while low cytoplasmic XBP1 expression remained an independent predictor of poor DSS. Our analysis also revealed that XBP1 expression, tumor differentiation, vascular invasion, and T classification were linked to DFS, while low cytoplasmic XBP1 expression remained an independent predictor of poor DFS. CONCLUSION Low cytoplasmic XBP1 protein expression may play an important role in the pathogenesis of HCC, which suggests that XBP1 could potentially be targeted to benefit therapeutic strategies for HCC.
Collapse
Affiliation(s)
- Hui-Ting Hsu
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan, R.O.C
- School of Medicine, Chung Shan Medical University Hospital, Taichung, Taiwan, R.O.C
- Department of Pathology, China Medical University Hospital, Taichung, Taiwan, R.O.C
| | - Yueh-Min Lin
- Department of Pathology, Changhua Christian Hospital, Changhua, Taiwan, R.O.C
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan, R.O.C
| | - Ming-Tai Hsing
- Department of Neurosurgery, Show Chwan Memorial Hospital, Changhua, Taiwan, R.O.C
| | - Kun-Tu Yeh
- Department of Pathology, Changhua Christian Hospital, Changhua, Taiwan, R.O.C
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan, R.O.C
| | - Jeng-Wei Lu
- Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen, Denmark;
- The Finsen Laboratory, Rigshospitalet/National University Hospital, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Shun-Fa Yang
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan, R.O.C.;
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan, R.O.C
| |
Collapse
|
2
|
Gouttefangeas C, Klein R, Maia A. The good and the bad of T cell cross-reactivity: challenges and opportunities for novel therapeutics in autoimmunity and cancer. Front Immunol 2023; 14:1212546. [PMID: 37409132 PMCID: PMC10319254 DOI: 10.3389/fimmu.2023.1212546] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 05/24/2023] [Indexed: 07/07/2023] Open
Abstract
T cells are main actors of the immune system with an essential role in protection against pathogens and cancer. The molecular key event involved in this absolutely central task is the interaction of membrane-bound specific T cell receptors with peptide-MHC complexes which initiates T cell priming, activation and recall, and thus controls a range of downstream functions. While textbooks teach us that the repertoire of mature T cells is highly diverse, it is clear that this diversity cannot possibly cover all potential foreign peptides that might be encountered during life. TCR cross-reactivity, i.e. the ability of a single TCR to recognise different peptides, offers the best solution to this biological challenge. Reports have shown that indeed, TCR cross-reactivity is surprisingly high. Hence, the T cell dilemma is the following: be as specific as possible to target foreign danger and spare self, while being able to react to a large spectrum of body-threatening situations. This has major consequences for both autoimmune diseases and cancer, and significant implications for the development of T cell-based therapies. In this review, we will present essential experimental evidence of T cell cross-reactivity, implications for two opposite immune conditions, i.e. autoimmunity vs cancer, and how this can be differently exploited for immunotherapy approaches. Finally, we will discuss the tools available for predicting cross-reactivity and how improvements in this field might boost translational approaches.
Collapse
Affiliation(s)
- Cécile Gouttefangeas
- Nencki Institute of Experimental Biology of the Polish Academy of Sciences, Warsaw, Poland
- Cluster of Excellence iFIT (EXC2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tübingen, Tübingen, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ) partner site Tübingen, Tübingen, Germany
| | - Reinhild Klein
- Department of Hematology, Oncology, Clinical Immunology and Rheumatology, University Hospital Tübingen, Tübingen, Germany
| | - Ana Maia
- Nencki Institute of Experimental Biology of the Polish Academy of Sciences, Warsaw, Poland
- Cluster of Excellence iFIT (EXC2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tübingen, Tübingen, Germany
| |
Collapse
|
3
|
Dima D, Jiang D, Singh DJ, Hasipek M, Shah HS, Ullah F, Khouri J, Maciejewski JP, Jha BK. Multiple Myeloma Therapy: Emerging Trends and Challenges. Cancers (Basel) 2022; 14:cancers14174082. [PMID: 36077618 PMCID: PMC9454959 DOI: 10.3390/cancers14174082] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 08/16/2022] [Accepted: 08/18/2022] [Indexed: 11/16/2022] Open
Abstract
Multiple myeloma (MM) is a complex hematologic malignancy characterized by the uncontrolled proliferation of clonal plasma cells in the bone marrow that secrete large amounts of immunoglobulins and other non-functional proteins. Despite decades of progress and several landmark therapeutic advancements, MM remains incurable in most cases. Standard of care frontline therapies have limited durable efficacy, with the majority of patients eventually relapsing, either early or later. Induced drug resistance via up-modulations of signaling cascades that circumvent the effect of drugs and the emergence of genetically heterogeneous sub-clones are the major causes of the relapsed-refractory state of MM. Cytopenias from cumulative treatment toxicity and disease refractoriness limit therapeutic options, hence creating an urgent need for innovative approaches effective against highly heterogeneous myeloma cell populations. Here, we present a comprehensive overview of the current and future treatment paradigm of MM, and highlight the gaps in therapeutic translations of recent advances in targeted therapy and immunotherapy. We also discuss the therapeutic potential of emerging preclinical research in multiple myeloma.
Collapse
Affiliation(s)
- Danai Dima
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Center for Immunotherapy and Precision Immuno-Oncology, Lerner Research Institute, Cleveland, OH 44195, USA
- Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Dongxu Jiang
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Center for Immunotherapy and Precision Immuno-Oncology, Lerner Research Institute, Cleveland, OH 44195, USA
| | - Divya Jyoti Singh
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Center for Immunotherapy and Precision Immuno-Oncology, Lerner Research Institute, Cleveland, OH 44195, USA
| | - Metis Hasipek
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Haikoo S. Shah
- Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Fauzia Ullah
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Jack Khouri
- Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA
- Cleveland Clinic Lerner College of Medicine, Cleveland, OH 44195, USA
| | - Jaroslaw P. Maciejewski
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA
- Cleveland Clinic Lerner College of Medicine, Cleveland, OH 44195, USA
| | - Babal K. Jha
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Center for Immunotherapy and Precision Immuno-Oncology, Lerner Research Institute, Cleveland, OH 44195, USA
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA
- Cleveland Clinic Lerner College of Medicine, Cleveland, OH 44195, USA
- Correspondence:
| |
Collapse
|
4
|
Borrello MT, Martin MB, Pin CL. The unfolded protein response: An emerging therapeutic target for pancreatitis and pancreatic ductal adenocarcinoma. Pancreatology 2022; 22:148-159. [PMID: 34774415 DOI: 10.1016/j.pan.2021.10.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 10/23/2021] [Accepted: 10/25/2021] [Indexed: 12/11/2022]
Abstract
Pancreatitis is a debilitating disease involving inflammation and fibrosis of the exocrine pancreas. Recurrent or chronic forms of pancreatitis are a significant risk factor for pancreatic ductal adenocarcinoma. While genetic factors have been identified for both pathologies, environmental stresses play a large role in their etiology. All cells have adapted mechanisms to handle acute environmental stress that alters energy demands. A common pathway involved in the stress response involves endoplasmic reticulum stress and the unfolded protein response (UPR). While rapidly activated by many external stressors, in the pancreas the UPR plays a fundamental biological role, likely due to the high protein demands in acinar cells. Despite this, increased UPR activity is observed in response to acute injury or following exposure to risk factors associated with pancreatitis and pancreatic cancer. Studies in animal and cell cultures models show the importance of affecting the UPR in the context of both diseases, and inhibitors have been developed for several specific mediators of the UPR. Given the importance of the UPR to normal acinar cell function, efforts to affect the UPR in the context of disease must be able to specifically target pathology vs. physiology. In this review, we highlight the importance of the UPR to normal and pathological conditions of the exocrine pancreas. We discuss recent studies suggesting the UPR may be involved in the initiation and progression of pancreatitis and PDAC, as well as contributing to chemoresistance that occurs in pancreatic cancer. Finally, we discuss the potential of targeting the UPR for treatment.
Collapse
Affiliation(s)
- M Teresa Borrello
- Newcastle Fibrosis Research Group, Biosciences Institute, Newcastle University, Newcastle Upon Tyne, UK
| | - Mickenzie B Martin
- Depts. of Physiology and Pharmacology, Paediatrics, and Oncology, Schulich School of Medicine and Dentistry, The University of Western Ontario, Canada; Children's Health Research Institute, Lawson Health Research Institute, London, Ontario, Canada
| | - Christopher L Pin
- Depts. of Physiology and Pharmacology, Paediatrics, and Oncology, Schulich School of Medicine and Dentistry, The University of Western Ontario, Canada; Children's Health Research Institute, Lawson Health Research Institute, London, Ontario, Canada.
| |
Collapse
|
5
|
Tagliamonte M, Mauriello A, Cavalluzzo B, Ragone C, Manolio C, Luciano A, Barbieri A, Palma G, Scognamiglio G, Di Mauro A, Di Bonito M, Tornesello ML, Buonaguro FM, Vitagliano L, Caporale A, Ruvo M, Buonaguro L. MHC-Optimized Peptide Scaffold for Improved Antigen Presentation and Anti-Tumor Response. Front Immunol 2021; 12:769799. [PMID: 34745146 PMCID: PMC8564487 DOI: 10.3389/fimmu.2021.769799] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 10/04/2021] [Indexed: 11/17/2022] Open
Abstract
Tumor Associated Antigens (TAAs) may suffer from an immunological tolerance due to expression on normal cells. In order to potentiate their immunogenicity, heteroclitic peptides (htcPep) were designed according to prediction algorithms. In particular, specific modifications were introduced in peptide residues facing to TCR. Moreover, a MHC-optimized scaffold was designed for improved antigen presentation to TCR by H-2Db allele. The efficacy of such htcPep was assessed in C57BL/6 mice injected with syngeneic melanoma B16F10 or lung TC1 tumor cell lines, in combination with metronomic chemotherapy and immune checkpoint inhibitors. The immunogenicity of htcPep was significantly stronger than the corresponding wt peptide and the modification involving both MHC and TCR binding residues scored the strongest. In particular, the H-2Db-specific scaffold significantly potentiated the peptides' immunogenicity and control of tumor growth was comparable to wt peptide in a therapeutic setting. Overall, we demonstrated that modified TAAs show higher immunogenicity compared to wt peptide. In particular, the MHC-optimized scaffold can present different antigen sequences to TCR, retaining the conformational characteristics of the corresponding wt. Cross-reacting CD8+ T cells are elicited and efficiently kill tumor cells presenting the wild-type antigen. This novel approach can be of high clinical relevance in cancer vaccine development.
Collapse
MESH Headings
- Animals
- Antigen Presentation/drug effects
- Antigen Presentation/immunology
- Antigens, Neoplasm/immunology
- Antigens, Neoplasm/metabolism
- Antineoplastic Combined Chemotherapy Protocols/administration & dosage
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- Cancer Vaccines/administration & dosage
- Cancer Vaccines/immunology
- Cell Line, Tumor
- Combined Modality Therapy
- Female
- Histocompatibility Antigens/immunology
- Humans
- Mice, Inbred C57BL
- Neoplasms, Experimental/immunology
- Neoplasms, Experimental/metabolism
- Neoplasms, Experimental/prevention & control
- Peptides/immunology
- Peptides/metabolism
- Protein Binding
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/metabolism
- Treatment Outcome
- Tumor Burden/drug effects
- Tumor Burden/immunology
- Vaccines, Subunit/administration & dosage
- Vaccines, Subunit/immunology
- Mice
Collapse
Affiliation(s)
- Maria Tagliamonte
- Innovative Immunological Models Lab, Istituto Nazionale Tumori - Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) - “Fond G. Pascale”, Naples, Italy
| | - Angela Mauriello
- Innovative Immunological Models Lab, Istituto Nazionale Tumori - Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) - “Fond G. Pascale”, Naples, Italy
| | - Beatrice Cavalluzzo
- Innovative Immunological Models Lab, Istituto Nazionale Tumori - Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) - “Fond G. Pascale”, Naples, Italy
| | - Concetta Ragone
- Innovative Immunological Models Lab, Istituto Nazionale Tumori - Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) - “Fond G. Pascale”, Naples, Italy
| | - Carmen Manolio
- Innovative Immunological Models Lab, Istituto Nazionale Tumori - Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) - “Fond G. Pascale”, Naples, Italy
| | - Antonio Luciano
- Animal Facility, Istituto Nazionale Tumori - Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) - “Fond G. Pascale”, Naples, Italy
| | - Antonio Barbieri
- Animal Facility, Istituto Nazionale Tumori - Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) - “Fond G. Pascale”, Naples, Italy
| | - Giuseppe Palma
- Animal Facility, Istituto Nazionale Tumori - Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) - “Fond G. Pascale”, Naples, Italy
| | - Giosuè Scognamiglio
- Pathology Unit, Istituto Nazionale Tumori - Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) - “Fond G. Pascale”, Naples, Italy
| | - Annabella Di Mauro
- Pathology Unit, Istituto Nazionale Tumori - Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) - “Fond G. Pascale”, Naples, Italy
| | - Maurizio Di Bonito
- Pathology Unit, Istituto Nazionale Tumori - Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) - “Fond G. Pascale”, Naples, Italy
| | - Maria Lina Tornesello
- Molecular Biology and Viral Oncogenesis, Istituto Nazionale Tumori - Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) - “Fond G. Pascale”, Naples, Italy
| | - Franco M. Buonaguro
- Molecular Biology and Viral Oncogenesis, Istituto Nazionale Tumori - Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) - “Fond G. Pascale”, Naples, Italy
| | - Luigi Vitagliano
- Institute of Biostructures and Bioimaging, Consiglio Nazionale delle Ricerche (CNR), Napoli, Italy
| | - Andrea Caporale
- Institute of Biostructures and Bioimaging, Consiglio Nazionale delle Ricerche (CNR), Napoli, Italy
| | - Menotti Ruvo
- Institute of Biostructures and Bioimaging, Consiglio Nazionale delle Ricerche (CNR), Napoli, Italy
| | - Luigi Buonaguro
- Innovative Immunological Models Lab, Istituto Nazionale Tumori - Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) - “Fond G. Pascale”, Naples, Italy
| |
Collapse
|
6
|
Ragone C, Manolio C, Cavalluzzo B, Mauriello A, Tornesello ML, Buonaguro FM, Castiglione F, Vitagliano L, Iaccarino E, Ruvo M, Tagliamonte M, Buonaguro L. Identification and validation of viral antigens sharing sequence and structural homology with tumor-associated antigens (TAAs). J Immunother Cancer 2021; 9:jitc-2021-002694. [PMID: 34049932 PMCID: PMC8166618 DOI: 10.1136/jitc-2021-002694] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/08/2021] [Indexed: 11/11/2022] Open
Abstract
Background The host’s immune system develops in equilibrium with both cellular self-antigens and non-self-antigens derived from microorganisms which enter the body during lifetime. In addition, during the years, a tumor may arise presenting to the immune system an additional pool of non-self-antigens, namely tumor antigens (tumor-associated antigens, TAAs; tumor-specific antigens, TSAs). Methods In the present study, we looked for homology between published TAAs and non-self-viral-derived epitopes. Bioinformatics analyses and ex vivo immunological validations have been performed. Results Surprisingly, several of such homologies have been found. Moreover, structural similarities between paired TAAs and viral peptides as well as comparable patterns of contact with HLA and T cell receptor (TCR) α and β chains have been observed. Therefore, the two classes of non-self-antigens (viral antigens and tumor antigens) may converge, eliciting cross-reacting CD8+ T cell responses which possibly drive the fate of cancer development and progression. Conclusions An established antiviral T cell memory may turn out to be an anticancer T cell memory, able to control the growth of a cancer developed during the lifetime if the expressed TAA is similar to the viral epitope. This may ultimately represent a relevant selective advantage for patients with cancer and may lead to a novel preventive anticancer vaccine strategy.
Collapse
Affiliation(s)
- Concetta Ragone
- Experimental Oncology - Innovative Immunological Models, Istituto Nazionale per lo Studio e la Cura dei Tumori, "Fondazione Pascale"- IRCCS, Naples, Italy
| | - Carmen Manolio
- Experimental Oncology - Innovative Immunological Models, Istituto Nazionale per lo Studio e la Cura dei Tumori, "Fondazione Pascale"- IRCCS, Naples, Italy
| | - Beatrice Cavalluzzo
- Experimental Oncology - Innovative Immunological Models, Istituto Nazionale per lo Studio e la Cura dei Tumori, "Fondazione Pascale"- IRCCS, Naples, Italy
| | - Angela Mauriello
- Experimental Oncology - Innovative Immunological Models, Istituto Nazionale per lo Studio e la Cura dei Tumori, "Fondazione Pascale"- IRCCS, Naples, Italy
| | - Maria Lina Tornesello
- Esperimental Oncology - Molecular Biology and Viral Oncogenesis, Istituto Nazionale per lo Studio e la Cura dei Tumori, "Fondazione Pascale"- IRCCS, Naples, Italy
| | - Franco M Buonaguro
- Esperimental Oncology - Molecular Biology and Viral Oncogenesis, Istituto Nazionale per lo Studio e la Cura dei Tumori, "Fondazione Pascale"- IRCCS, Naples, Italy
| | | | | | | | - Menotti Ruvo
- Institute for Biostructures and Bioimages, CNR, Roma, Italy
| | - Maria Tagliamonte
- Experimental Oncology - Innovative Immunological Models, Istituto Nazionale per lo Studio e la Cura dei Tumori, "Fondazione Pascale"- IRCCS, Naples, Italy
| | - Luigi Buonaguro
- Experimental Oncology - Innovative Immunological Models, Istituto Nazionale per lo Studio e la Cura dei Tumori, "Fondazione Pascale"- IRCCS, Naples, Italy
| |
Collapse
|
7
|
Huang Y, Jiang L, Wei G. Circ_0006168 Promotes the Migration, Invasion and Proliferation of Esophageal Squamous Cell Carcinoma Cells via miR-516b-5p-Dependent Regulation of XBP1. Onco Targets Ther 2021; 14:2475-2488. [PMID: 33883902 PMCID: PMC8053607 DOI: 10.2147/ott.s293180] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 03/22/2021] [Indexed: 12/18/2022] Open
Abstract
Background Circular RNAs (circRNAs) exert important roles in carcinogenesis. Here, we aimed to uncover the working mechanism of circ_0006168 in esophageal squamous cell carcinoma (ESCC) development. Methods Western blot assay and real-time quantitative polymerase chain reaction (RT-qPCR) were used to determine protein and RNA expression, respectively. Wound healing assay and transwell migration assay were performed to assess cell migration ability, whereas cell invasion ability was evaluated by transwell invasion assay. 3-(4,5-dimethyl-thiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay and colony formation assay were utilized to analyze cell proliferation ability. Xenograft tumor model was utilized to assess the role of X-box binding protein 1 (XBP1) in xenograft tumor growth in vivo. Dual-luciferase reporter assay, RNA immunoprecipitation (RIP) assay and RNA pull down assay were used to verify intermolecular interactions. Results XBP1 silencing suppressed the migration, invasion and proliferation of ESCC cells in vitro and restrained the xenograft tumor growth in vivo. MicroRNA-516b-5p (miR-516b-5p) interacted with the 3ʹ untranslated region (3ʹUTR) of XBP1 in ESCC cells. MiR-516b-5p overexpression inhibited the proliferation and motility of ESCC cells. MiR-516b-5p was a molecular target of circ_0006168 in ESCC cells. The interference of circ_0006168 restrained the motility and proliferation of ESCC cells. Circ_0006168 acted as miR-516b-5p sponge to up-regulate XBP1 expression in ESCC cells. MiR-516b-5p silencing or the accumulation of XBP1 largely rescued the proliferation ability and motility in circ_0006168-silenced ESCC cells. Conclusion In conclusion, circ_0006168 contributed to ESCC development through promoting the proliferation and motility of ESCC cells via mediating miR-516b-5p/XBP1 axis.
Collapse
Affiliation(s)
- Yunhe Huang
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, People's Republic of China
| | - Lei Jiang
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, People's Republic of China
| | - Guangxia Wei
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, People's Republic of China
| |
Collapse
|
8
|
Triazoloacridone C-1305 impairs XBP1 splicing by acting as a potential IRE1α endoribonuclease inhibitor. Cell Mol Biol Lett 2021; 26:11. [PMID: 33730996 PMCID: PMC7968329 DOI: 10.1186/s11658-021-00255-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 03/09/2021] [Indexed: 02/06/2023] Open
Abstract
Inositol requiring enzyme 1 alpha (IRE1α) is one of three signaling sensors in the unfolding protein response (UPR) that alleviates endoplasmic reticulum (ER) stress in cells and functions to promote cell survival. During conditions of irrevocable stress, proapoptotic gene expression is induced to promote cell death. One of the three signaling stressors, IRE1α is an serine/threonine-protein kinase/endoribonuclease (RNase) that promotes nonconventional splicing of XBP1 mRNA that is translated to spliced XBP1 (XBP1s), an active prosurvival transcription factor. Interestingly, elevated IRE1α and XBP1s are both associated with poor cancer survival and drug resistance. In this study, we used next-generation sequencing analyses to demonstrate that triazoloacridone C-1305, a microtubule stabilizing agent that also has topoisomerase II inhibitory activity, dramatically decreases XBP1s mRNA levels and protein production during ER stress conditions, suggesting that C-1305 does this by decreasing IRE1α’s endonuclease activity.
Collapse
|
9
|
Cavalluzzo B, Ragone C, Mauriello A, Petrizzo A, Manolio C, Caporale A, Vitagliano L, Ruvo M, Buonaguro L, Tagliamonte M. Identification and characterization of heteroclitic peptides in TCR-binding positions with improved HLA-binding efficacy. J Transl Med 2021; 19:89. [PMID: 33637105 PMCID: PMC7913412 DOI: 10.1186/s12967-021-02757-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 02/16/2021] [Indexed: 01/03/2023] Open
Abstract
The antigenicity as well as the immunogenicity of tumor associated antigens (TAAs) may need to be potentiated in order to break the immunological tolerance. To this aim, heteroclitic peptides were designed introducing specific substitutions in the residue at position 4 (p4) binding to TCR. The effect of such modifications also on the affinity to the major histocompatibility class I (MHC-I) molecule was assessed. The Trp2 antigen, specific for the mouse melanoma B16F10 cells, as well as the HPV-E7 antigen, specific for the TC1 tumor cell lines, were used as models. Affinity of such heteroclitic peptides to HLA was predicted by bioinformatics tools and the most promising ones were validated by structural conformational and HLA binding analyses. Overall, we demonstrated that TAAs modified at the TCR-binding p4 residue are predicted to have higher affinity to MHC-I molecules. Experimental evaluation confirms the stronger binding, suggesting that this strategy may be very effective for designing new vaccines with improved antigenic efficacy.
Collapse
Affiliation(s)
- Beatrice Cavalluzzo
- Innovative Immunological Models Lab, Istituto Nazionale Tumori "Fond. G. Pascale", Via Mariano Semmola, 1, 80131, Naples, Italy
| | - Concetta Ragone
- Innovative Immunological Models Lab, Istituto Nazionale Tumori "Fond. G. Pascale", Via Mariano Semmola, 1, 80131, Naples, Italy
| | - Angela Mauriello
- Innovative Immunological Models Lab, Istituto Nazionale Tumori "Fond. G. Pascale", Via Mariano Semmola, 1, 80131, Naples, Italy
| | - Annacarmen Petrizzo
- Innovative Immunological Models Lab, Istituto Nazionale Tumori "Fond. G. Pascale", Via Mariano Semmola, 1, 80131, Naples, Italy
| | - Carmen Manolio
- Innovative Immunological Models Lab, Istituto Nazionale Tumori "Fond. G. Pascale", Via Mariano Semmola, 1, 80131, Naples, Italy
| | - Andrea Caporale
- Institute of Biostructures and Bioimaging, CNR, Naples, Italy.,Istituto Di Cristallografia-CNR, c/o area Science Park S.S. 14 Km 163.5 Basovizza, 34149, Trieste, Italy
| | | | - Menotti Ruvo
- Institute of Biostructures and Bioimaging, CNR, Naples, Italy
| | - Luigi Buonaguro
- Innovative Immunological Models Lab, Istituto Nazionale Tumori "Fond. G. Pascale", Via Mariano Semmola, 1, 80131, Naples, Italy
| | - Maria Tagliamonte
- Innovative Immunological Models Lab, Istituto Nazionale Tumori "Fond. G. Pascale", Via Mariano Semmola, 1, 80131, Naples, Italy.
| |
Collapse
|
10
|
Cao X, Xue LD, Di Y, Li T, Tian YJ, Song Y. MSC-derived exosomal lncRNA SNHG7 suppresses endothelial-mesenchymal transition and tube formation in diabetic retinopathy via miR-34a-5p/XBP1 axis. Life Sci 2021; 272:119232. [PMID: 33600866 DOI: 10.1016/j.lfs.2021.119232] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 01/28/2021] [Accepted: 02/10/2021] [Indexed: 01/13/2023]
Abstract
AIMS Diabetic retinopathy (DR) is the most common complication of type 2 diabetes mellitus, which could result in visual impairment. Accumulating studies have shown the implication of long non-coding RNAs (lncRNAs) in the pathogenesis of DR. Our aims are to investigate whether lncRNA SNHG7 plays a role during DR pathogenesis. MAIN METHODS Human retinal microvascular endothelial cells (HRMECs) were treated with high glucose (HG) to build cell model. Relative expression of RNAs were examined using qPCR, and western blot or immunofluorescence analysis was adopted to detect the protein expression. Cell viability, migration and angiogenic capacity of HRMECs were estimated through CCK-8, transwell and tube formation experiments, respectively. Dual-luciferase reporter and RNA pull down assays were employed to verify the interplay between miR-34a-5p and SNHG7 or XBP1. Mesenchymal stem cells (MSCs) were identified by examining typical surface makers using flow cytometry and the differentiation abilities via Alizarin red, Oil red O and Alcian blue staining. MSC-derived exosomes were verified by transmission electron microscopy and western blot. KEY FINDINGS LncRNA SNHG7 sponged to and negatively regulated miR-34a-5p. SNHG7 overexpression repressed HG induced endothelial-mesenchymal transition (EndMT) and tube formation of HRMECs, while miR-34a-5p overexpression could reverse this effect. miR-34a-5p targeted and negative regulated XBP1. Knockdown of miR-34a-5p repressed HG induced EndMT and tube formation, which were partially blocked by XBP1 inhibition. MSC-derived exosomes could transfer SNHG7 to HRMECs and modulated EndMT and tube formation. SIGNIFICANCE The MSC-derived exosomal lncRNA SNHG7 suppresses EndMT and tube formation in HRMECs via miR-34a-5p/XBP1 axis.
Collapse
Affiliation(s)
- Xin Cao
- Department of Ophthalmology, The Second Affiliated Hospital of Nantong University, Nantong 226000, Jiangsu Province, PR China
| | - Li-Dan Xue
- Department of Ophthalmology, The Second Affiliated Hospital of Nantong University, Nantong 226000, Jiangsu Province, PR China
| | - Yue Di
- Department of Ophthalmology, The Second Affiliated Hospital of Nantong University, Nantong 226000, Jiangsu Province, PR China
| | - Tao Li
- Department of Ophthalmology, The Second Affiliated Hospital of Nantong University, Nantong 226000, Jiangsu Province, PR China
| | - Ya-Jing Tian
- Department of Ophthalmology, The Second Affiliated Hospital of Nantong University, Nantong 226000, Jiangsu Province, PR China
| | - Yu Song
- Department of Ophthalmology, The Second Affiliated Hospital of Nantong University, Nantong 226000, Jiangsu Province, PR China.
| |
Collapse
|
11
|
Buonaguro L, Tagliamonte M. Selecting Target Antigens for Cancer Vaccine Development. Vaccines (Basel) 2020; 8:vaccines8040615. [PMID: 33080888 PMCID: PMC7711972 DOI: 10.3390/vaccines8040615] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 10/12/2020] [Accepted: 10/13/2020] [Indexed: 12/11/2022] Open
Abstract
One of the principal goals of cancer immunotherapy is the development of efficient therapeutic cancer vaccines that are able to elicit an effector as well as memory T cell response specific to tumor antigens. In recent years, the attention has been focused on the personalization of cancer vaccines. However, the efficacy of therapeutic cancer vaccines is still disappointing despite the large number of vaccine strategies targeting different tumors that have been evaluated in recent years. While the preclinical data have frequently shown encouraging results, clinical trials have not provided satisfactory data to date. The main reason for such failures is the complexity of identifying specific target tumor antigens that should be unique or overexpressed only by the tumor cells compared to normal cells. Most of the tumor antigens included in cancer vaccines are non-mutated overexpressed self-antigens, eliciting mainly T cells with low-affinity T cell receptors (TCR) unable to mediate an effective anti-tumor response. In this review, the target tumor antigens employed in recent years in the development of therapeutic cancer vaccine strategies are described, along with potential new classes of tumor antigens such as the human endogenous retroviral elements (HERVs), unconventional antigens, and/or heteroclitic peptides.
Collapse
|
12
|
Chen S, Chen J, Hua X, Sun Y, Cui R, Sha J, Zhu X. The emerging role of XBP1 in cancer. Biomed Pharmacother 2020; 127:110069. [PMID: 32294597 DOI: 10.1016/j.biopha.2020.110069] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 02/21/2020] [Accepted: 03/03/2020] [Indexed: 12/20/2022] Open
Abstract
X-box binding protein 1 (XBP1) is a unique basic-region leucine zipper (bZIP) transcription factor whose dynamic form is controlled by an alternative splicing response upon disturbance of homeostasis in the endoplasmic reticulum (ER) and activation of the unfolded protein response (UPR). XBP1 was first distinguished as a key regulator of major histocompatibility complex (MHC) class II gene expression in B cells. XBP1 communicates with the foremost conserved signalling component of the UPR and is essential for cell fate determination in response to ER stress (ERS). Here, we review recent advances in our understanding of this multifaceted translation component in cancer. In this review, we briefly discuss the role of XBP1 mediators in the UPR and the transcriptional function of XBP1. In addition, we describe how XBP1 operates as a key factor in tumour progression and metastasis. We mainly review XBP1's expression, function and prognostic value in research on solid tumours. Finally, we discuss multiple approaches, especially those involving XBP1, that overcome the immunosuppressive effect of the UPR in cancer that could potentially be useful as antitumour therapies.
Collapse
Affiliation(s)
- Shanshan Chen
- School of Medicine, Southeast University, Nanjing, 210009, China.
| | - Jing Chen
- Department of Respiratory, Zhongda Hospital of Southeast University, Nanjing, 210009, China
| | - Xin Hua
- School of Medicine, Southeast University, Nanjing, 210009, China
| | - Yue Sun
- School of Medicine, Southeast University, Nanjing, 210009, China
| | - Rui Cui
- School of Medicine, Southeast University, Nanjing, 210009, China
| | - Jun Sha
- School of Medicine, Southeast University, Nanjing, 210009, China
| | - Xiaoli Zhu
- School of Medicine, Southeast University, Nanjing, 210009, China; Department of Respiratory, Zhongda Hospital of Southeast University, Nanjing, 210009, China.
| |
Collapse
|
13
|
Shi W, Chen Z, Li L, Liu H, Zhang R, Cheng Q, Xu D, Wu L. Unravel the molecular mechanism of XBP1 in regulating the biology of cancer cells. J Cancer 2019; 10:2035-2046. [PMID: 31205564 PMCID: PMC6548171 DOI: 10.7150/jca.29421] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Accepted: 04/02/2019] [Indexed: 02/07/2023] Open
Abstract
Cancer cells are usually exposed to stressful environments, such as hypoxia, nutrient deprivation, and other metabolic dysfunctional regulation, leading to continuous endoplasmic reticulum (ER) stress. As the most conserved branch among the three un-folded protein response (UPR) pathways, Inositol-requiring enzyme 1α (IRE1α)-X-box-binding protein 1 (XBP1) signaling has been implicated in cancer development and progression. Active XBP1 with transactivation domain functions as a transcription factor to regulate the expression of downstream target genes, including many oncogenic factors. The regulatory activity of XBP1 in cell proliferation, apoptosis, metastasis, and drug resistance promotes cell survival, leading to tumorigenesis and tumor progression. In addition, the XBP1 peptides-based vaccination and/or combination with immune-modulatory drug administration have been developed for effective management for several cancers. Potentially, XBP1 is the biomarker of cancer development and progression and the strategy for clinical cancer management.
Collapse
Affiliation(s)
- Weimei Shi
- College of Pharmacy, Gannan Medical University, Ganzhou China, 341000
| | - Zhixi Chen
- College of Pharmacy, Gannan Medical University, Ganzhou China, 341000
| | - Linfu Li
- College of Pharmacy, Gannan Medical University, Ganzhou China, 341000
| | - Hai Liu
- College of Pharmacy, Gannan Medical University, Ganzhou China, 341000
| | - Rui Zhang
- College of Pharmacy, Gannan Medical University, Ganzhou China, 341000
| | - Qilai Cheng
- College of Pharmacy, Gannan Medical University, Ganzhou China, 341000
| | - Daohua Xu
- Department of Pharmacology, Guangdong Medical University, Dongguan China, 523808
| | - Longhuo Wu
- College of Pharmacy, Gannan Medical University, Ganzhou China, 341000
| |
Collapse
|
14
|
Hang J, Huang J, Zhou S, Wu L, Zhu Y, Zhu L, Zhou H, Xu K, Jiang H, Yang X. The clinical implication of CD45RA + naïve T cells and CD45RO + memory T cells in advanced pancreatic cancer: a proxy for tumor biology and outcome prediction. Cancer Med 2019; 8:1326-1335. [PMID: 30767430 PMCID: PMC6434335 DOI: 10.1002/cam4.1988] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 12/20/2018] [Accepted: 12/23/2018] [Indexed: 01/18/2023] Open
Abstract
Naïve and memory T cells play a pivotal role in solid tumor pathogenesis but their role in pancreatic cancer progression remains elusive. Thus, we aimed to investigate their clinical potential in advanced pancreatic cancer (APC). Flow cytometry was performed to evaluate the level of baseline peripheral naïve and memory T cells from 137 APC patients before receiving first‐line chemotherapy. Interrelationships between naïve, memory T cells and clinicopathological variables were evaluated using Pearson’s correlation. The prognostic impact of naïve and memory T cells were assessed by Kaplan‐Meier analysis and Cox regression. The correlation between naïve/memory T cells and tumor progression was investigated by Student’s t test. CD4+ naïve/memory ratio showed close correlations with hemoglobin, red blood cell (RBC), absolute neutrophil count (ANC) and platelet while CD8+ naïve/memory ratio was correlated with hemoglobin, RBC and CEA. Higher baseline lever of CD4+CD45RO+/CD4+ was correlated with better overall survival (OS) (P = 0.036). Patients with CD4+ naïve/memory ratio ≥0.36 had a poorer OS than those with CD4+ naïve/memory ratio <0.36 (P = 0.021). In addition, CD4+ naïve/memory ratio showed independent prognostic impact (HR 1.427, 95% CI 1.033‐1.973, P = 0.031). Furthermore, poorer clinical response was correlated with higher level of CD8+ naïve/memory ratio after the third cycle of chemotherapy (P = 0.01). Besides, patients with a lower level of CD8+ naïve/memory ratio had longer progression‐free survival (PFS) (P = 0.028). We propose CD4+ naïve/memory ratio as a novel prognostic biomarker for APC. In addition, CD8+ naïve/memory ratio can be a candidate marker for predicting PFS and the change of its level may reflect the progression of APC.
Collapse
Affiliation(s)
- Junjie Hang
- Department of Oncology, Changzhou No.2 People's Hospital, Affiliated Hospital of Nanjing Medical University, Changzhou, Jiangsu, China
| | - Junjie Huang
- JC School of Public Health and Primary Care, Chinese University of Hong Kong, Hong Kong, China
| | - Siyuan Zhou
- Department of Oncology, Changzhou No.2 People's Hospital, Affiliated Hospital of Nanjing Medical University, Changzhou, Jiangsu, China
| | - Lixia Wu
- Department of Oncology, Shanghai JingAn District ZhaBei Central Hospital, Shanghai, China
| | - Yingwei Zhu
- Department of Oncology, Changzhou No.2 People's Hospital, Affiliated Hospital of Nanjing Medical University, Changzhou, Jiangsu, China
| | - Lina Zhu
- Department of Oncology, Changzhou No.2 People's Hospital, Affiliated Hospital of Nanjing Medical University, Changzhou, Jiangsu, China
| | - Hanyu Zhou
- Department of Oncology, Changzhou No.2 People's Hospital, Affiliated Hospital of Nanjing Medical University, Changzhou, Jiangsu, China
| | - Kequn Xu
- Department of Oncology, Changzhou No.2 People's Hospital, Affiliated Hospital of Nanjing Medical University, Changzhou, Jiangsu, China
| | - Hua Jiang
- Department of Oncology, Changzhou No.2 People's Hospital, Affiliated Hospital of Nanjing Medical University, Changzhou, Jiangsu, China
| | - Xuguang Yang
- State Key Laboratory for Oncogenes and Related Genes, Department of Oncology, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
15
|
Nooka AK, Wang ML, Yee AJ, Kaufman JL, Bae J, Peterkin D, Richardson PG, Raje NS. Assessment of Safety and Immunogenicity of PVX-410 Vaccine With or Without Lenalidomide in Patients With Smoldering Multiple Myeloma: A Nonrandomized Clinical Trial. JAMA Oncol 2018; 4:e183267. [PMID: 30128502 DOI: 10.1001/jamaoncol.2018.3267] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Importance Increasing evidence suggests the significance of the role of the immune system in the progression of smoldering multiple myeloma (SMM) to symptomatic multiple myeloma (MM). Boosting the immune system via vaccination in the earlier, asymptomatic SMM stage may provide a novel strategy to prevent or slow progression to active MM. Objective To determine the safety, tolerability, immunogenicity, and anti-MM activity of the PVX-410 multipeptide vaccine with or without lenalidomide. Design, Setting, and Participants This 3-cohort phase 1/2a multicenter dose-escalation study accrued 22 adults (≥18 years) with SMM with normal organ/marrow function who were human leukocyte antigen A2-positive and at moderate or high risk of progression to MM. Interventions Patients received 6 doses of PVX-410 emulsified in Montanide ISA 720 VG, 0.4 mg total (0.1 mg/peptide) (n = 3) or 0.8 mg total (0.2 mg/peptide) (n = 9), biweekly via subcutaneous injection. In the combination cohort (n = 10), patients also received three 21-day cycles of lenalidomide, 25 mg, orally daily every 28 days. All patients received 0.5 mL (1 mg) poly-ICLC (2 mg/mL) via intramuscular injection with each PVX-410 dose. Main Outcomes and Measures Adverse events (AEs) were evaluated using the Common Terminology Criteria for Adverse Events, version 4.03. PVX-410-specific T lymphocytes by flow cytometry to assess tetramer and interferon (IFN)-γ response. Disease response was assessed by investigators using the International Myeloma Working Group (IMWG) and modified European Group for Bone Marrow Transplantation (EBMT) criteria. Results Overall, 14 (64%) patients were men and the median age at enrollment was 56 years in the monotherapy and 57 years in the combination cohorts (overall range, 39-82 years). Six of 12 patients in the monotherapy and 9 of 10 in the combination cohorts were at moderate risk. The PVX-410 vaccine was well tolerated. The most common AEs were mild-to-moderate injection site reactions and constitutional symptoms. Of note, PVX-410 was immunogenic as monotherapy (10 of 11 patients) and in combination with lenalidomide (9 of 9 patients), as demonstrated by an increase in percentage of tetramer-positive cells and IFN-γ cells in the CD3+CD8+ cell population. The combination resulted in greater mean fold increases in proportions of CD3+CD8+ T cells that were tetramer-positive and IFN-γ-positive, statistically significant for IFN-γ-positive cells after 2 and 4 vaccinations. An increase and persistence of vaccine-specific effector memory cells was noted. In total, 7 of 12 patients in the PVX-410-alone cohort had stable disease with 2 of 3 (low-dose cohort) and 1 of 9 of the target-dose cohort progressing (median TTP, 36 weeks), whereas 5 of 12 patients in the combination cohort showed, clinical response, with 1 patient progressing (median TTP not reached). Conclusions and Relevance Overall, these results suggest that the vaccine is safe and immunogenic in this patient population and support continued study of PVX-410 in SMM. Trial Registration ClinicalTrials.gov identifier: NCT01718899.
Collapse
Affiliation(s)
- Ajay K Nooka
- Department of Hematology and Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia
| | | | - Andrew J Yee
- Center for Multiple Myeloma, Massachusetts General Hospital Cancer Center, Boston, Massachusetts
| | - Jonathan L Kaufman
- Department of Hematology and Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia
| | - Jooeun Bae
- Dana-Farber Cancer Institute, Boston, Massachusetts
| | | | | | - Noopur S Raje
- Center for Multiple Myeloma, Massachusetts General Hospital Cancer Center, Boston, Massachusetts
| |
Collapse
|
16
|
Bae J, Hideshima T, Tai YT, Song Y, Richardson P, Raje N, Munshi NC, Anderson KC. Histone deacetylase (HDAC) inhibitor ACY241 enhances anti-tumor activities of antigen-specific central memory cytotoxic T lymphocytes against multiple myeloma and solid tumors. Leukemia 2018; 32:1932-1947. [PMID: 29487385 PMCID: PMC6537609 DOI: 10.1038/s41375-018-0062-8] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Revised: 11/27/2017] [Accepted: 12/15/2017] [Indexed: 12/17/2022]
Abstract
Histone deacetylases (HDAC) are therapeutic targets in multiple cancers. ACY241, an HDAC6 selective inhibitor, has shown anti-multiple myeloma (MM) activity in combination with immunomodulatory drugs and proteasome inhibitors. Here we show ACY241 significantly reduces the frequency of CD138+ MM cells, CD4+CD25+FoxP3+ regulatory T cells, and HLA-DRLow/-CD11b+CD33+ myeloid-derived suppressor cells; and decreases expression of PD1/PD-L1 on CD8+ T cells and of immune checkpoints in bone marrow cells from myeloma patients. ACY241 increased B7 (CD80, CD86) and MHC (Class I, Class II) expression on tumor and dendritic cells. We further evaluated the effect of ACY241 on antigen-specific cytotoxic T lymphocytes (CTL) generated with heteroclitic XBP1unspliced184-192 (YISPWILAV) and XBP1spliced367-375 (YLFPQLISV) peptides. ACY241 induces co-stimulatory (CD28, 41BB, CD40L, OX40) and activation (CD38) molecule expression in a dose- and time-dependent manner, and anti-tumor activities, evidenced by increased perforin/CD107a expression, IFN-γ/IL-2/TNF-α production, and antigen-specific central memory CTL. These effects of ACY241 on antigen-specific memory T cells were associated with activation of downstream AKT/mTOR/p65 pathways and upregulation of transcription regulators including Bcl-6, Eomes, HIF-1 and T-bet. These studies therefore demonstrate mechanisms whereby ACY241 augments immune response, providing the rationale for its use, alone and in combination, to restore host anti-tumor immunity and improve patient outcome.
Collapse
MESH Headings
- Antigens, Neoplasm/genetics
- Antigens, Neoplasm/immunology
- Biomarkers
- Cell Line, Tumor
- Cytotoxicity, Immunologic/drug effects
- Epitopes, T-Lymphocyte/genetics
- Epitopes, T-Lymphocyte/immunology
- Gene Expression Regulation, Neoplastic/drug effects
- Histone Deacetylase Inhibitors/pharmacology
- Histone Deacetylases/metabolism
- Humans
- Immunologic Memory
- Lymphocyte Activation/drug effects
- Lymphocyte Activation/immunology
- Multiple Myeloma/drug therapy
- Multiple Myeloma/genetics
- Multiple Myeloma/immunology
- Multiple Myeloma/metabolism
- Neoplasms/drug therapy
- Neoplasms/genetics
- Neoplasms/immunology
- Neoplasms/metabolism
- Peptides/immunology
- Signal Transduction/drug effects
- T-Lymphocyte Subsets/drug effects
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/metabolism
- T-Lymphocytes, Cytotoxic/drug effects
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Cytotoxic/metabolism
- X-Box Binding Protein 1/chemistry
- X-Box Binding Protein 1/genetics
- X-Box Binding Protein 1/immunology
Collapse
Affiliation(s)
- Jooeun Bae
- Dana-Farber Cancer Institute, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
| | - Teru Hideshima
- Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Yu-Tzu Tai
- Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Yan Song
- Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Paul Richardson
- Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Noopur Raje
- Harvard Medical School, Boston, MA, USA
- Massachusetts General Hospital, Boston, MA, USA
| | - Nikhil C Munshi
- Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- VA Boston Healthcare System, Boston, MA, USA
| | - Kenneth C Anderson
- Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| |
Collapse
|
17
|
Garcia-Carbonero N, Li W, Cabeza-Morales M, Martinez-Useros J, Garcia-Foncillas J. New Hope for Pancreatic Ductal Adenocarcinoma Treatment Targeting Endoplasmic Reticulum Stress Response: A Systematic Review. Int J Mol Sci 2018; 19:E2468. [PMID: 30134550 PMCID: PMC6165247 DOI: 10.3390/ijms19092468] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 08/10/2018] [Accepted: 08/18/2018] [Indexed: 12/28/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal types of tumours, and its incidence is rising worldwide. Although survival can be improved by surgical resection when these tumours are detected at an early stage, this cancer is usually asymptomatic, and disease only becomes apparent after metastasis. Several risk factors are associated with this disease, the most relevant being chronic pancreatitis, diabetes, tobacco and alcohol intake, cadmium, arsenic and lead exposure, certain infectious diseases, and the mutational status of some genes associated to a familial component. PDAC incidence has increased in recent decades, and there are few alternatives for chemotherapeutic treatment. Endoplasmic reticulum (ER) stress factors such as GRP78/BiP (78 kDa glucose-regulated protein), ATF6α (activating transcription factor 6 isoform α), IRE1α (inositol-requiring enzyme 1 isoform α), and PERK (protein kinase RNA-like endoplasmic reticulum kinase) activate the transcription of several genes involved in both survival and apoptosis. Some of these factors aid in inducing a non-proliferative state in cancer called dormancy. Modulation of endoplasmic reticulum stress could induce dormancy of tumour cells, thus prolonging patient survival. In this systematic review, we have compiled relevant results concerning those endoplasmic reticulum stress factors involved in PDAC, and we have analysed the mechanism of dormancy associated to endoplasmic reticulum stress and its potential use as a chemotherapeutic target against PDAC.
Collapse
MESH Headings
- Activating Transcription Factor 6/genetics
- Activating Transcription Factor 6/metabolism
- Animals
- Antibodies/pharmacology
- Carcinoma, Pancreatic Ductal/etiology
- Carcinoma, Pancreatic Ductal/genetics
- Carcinoma, Pancreatic Ductal/metabolism
- Carcinoma, Pancreatic Ductal/therapy
- Communicable Diseases/complications
- Communicable Diseases/genetics
- Communicable Diseases/metabolism
- Communicable Diseases/pathology
- Deoxycytidine/analogs & derivatives
- Deoxycytidine/pharmacology
- Diabetes Complications/genetics
- Diabetes Complications/metabolism
- Diabetes Complications/pathology
- Disease Models, Animal
- Endoplasmic Reticulum Chaperone BiP
- Endoplasmic Reticulum Stress/drug effects
- Endoplasmic Reticulum Stress/genetics
- Endoribonucleases/genetics
- Endoribonucleases/metabolism
- Gene Expression Regulation
- Heat-Shock Proteins/antagonists & inhibitors
- Heat-Shock Proteins/genetics
- Heat-Shock Proteins/metabolism
- Humans
- Pancreatic Neoplasms/etiology
- Pancreatic Neoplasms/genetics
- Pancreatic Neoplasms/metabolism
- Pancreatic Neoplasms/therapy
- Pancreatitis, Chronic/complications
- Pancreatitis, Chronic/genetics
- Pancreatitis, Chronic/metabolism
- Pancreatitis, Chronic/pathology
- Protein Serine-Threonine Kinases/genetics
- Protein Serine-Threonine Kinases/metabolism
- RNA, Small Interfering/genetics
- RNA, Small Interfering/metabolism
- Risk Factors
- Sulfones/pharmacology
- eIF-2 Kinase/genetics
- eIF-2 Kinase/metabolism
- Gemcitabine
Collapse
Affiliation(s)
- Nuria Garcia-Carbonero
- Translational Oncology Division, OncoHealth Institute, Health Research Institute-University Hospital Fundación Jiménez Díaz-UAM, Avda Reyes Catolicos 2, 28040 Madrid, Spain.
| | - Weiyao Li
- Translational Oncology Division, OncoHealth Institute, Health Research Institute-University Hospital Fundación Jiménez Díaz-UAM, Avda Reyes Catolicos 2, 28040 Madrid, Spain.
| | - Marticela Cabeza-Morales
- Translational Oncology Division, OncoHealth Institute, Health Research Institute-University Hospital Fundación Jiménez Díaz-UAM, Avda Reyes Catolicos 2, 28040 Madrid, Spain.
| | - Javier Martinez-Useros
- Translational Oncology Division, OncoHealth Institute, Health Research Institute-University Hospital Fundación Jiménez Díaz-UAM, Avda Reyes Catolicos 2, 28040 Madrid, Spain.
| | - Jesus Garcia-Foncillas
- Translational Oncology Division, OncoHealth Institute, Health Research Institute-University Hospital Fundación Jiménez Díaz-UAM, Avda Reyes Catolicos 2, 28040 Madrid, Spain.
| |
Collapse
|
18
|
Bae J, Hideshima T, Zhang GL, Zhou J, Keskin DB, Munshi NC, Anderson KC. Identification and characterization of HLA-A24-specific XBP1, CD138 (Syndecan-1) and CS1 (SLAMF7) peptides inducing antigens-specific memory cytotoxic T lymphocytes targeting multiple myeloma. Leukemia 2018; 32:752-764. [PMID: 29089645 PMCID: PMC5953209 DOI: 10.1038/leu.2017.316] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Revised: 09/17/2017] [Accepted: 10/03/2017] [Indexed: 12/27/2022]
Abstract
X-box binding protein 1 (XBP1), CD138 (Syndecan-1) and CS1 (SLAMF7) are highly expressed antigens in cancers including multiple myeloma (MM). Here, we identify and characterize immunogenic HLA-A24 peptides derived from these antigens for potential vaccination therapy of HLA-A24+ patients with MM. The identified immunogenic HLA-A24-specific XBP1 unspliced (UN)185-193 (I S P W I L A V L), XBP1 spliced (SP)223-231 (V Y P E G P S S L), CD138265-273 (I F A V C L V G F) and CS1240-248 (L F V L G L F L W) peptides induced antigen-specific CTL with anti-MM activity in an HLA-A24 restricted manner. Furthermore, a cocktail containing the four HLA-A24 peptides evoked MM-specific CTL with distinct phenotypic profiles (CD28, CD40L, 41BB, CD38, CD69) and anti-tumor activities, evidenced by perforin upregulation, CD107a degranulation (cytotoxicity) and Th1-type cytokines (IFN-γ/IL-2/TNF-α) production in response to HLA-A24+ MM cells. The multipeptide-specific CTL included antigen-specific memory CD8+ T cells expressing both T-cell activation (CD38, CD69) and immune checkpoints antigens (CTLA, PD-1, LAG-3, TIM-3). These results provide the framework for a multipeptide vaccination therapy to induce tumor-specific CTL in HLA-A24-positive patients with myeloma and other cancers expressing these antigens.
Collapse
Affiliation(s)
- Jooeun Bae
- Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Teru Hideshima
- Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | | | - Jun Zhou
- Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Derin B. Keskin
- Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Nikhil C. Munshi
- Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
- VA Boston Healthcare System, Boston, Massachusetts, USA
| | - Kenneth C. Anderson
- Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
19
|
Aberrant Glycosylation of Anchor-Optimized MUC1 Peptides Can Enhance Antigen Binding Affinity and Reverse Tolerance to Cytotoxic T Lymphocytes. Biomolecules 2016; 6:biom6030031. [PMID: 27367740 PMCID: PMC5039417 DOI: 10.3390/biom6030031] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2016] [Revised: 06/07/2016] [Accepted: 06/07/2016] [Indexed: 12/22/2022] Open
Abstract
Cancer vaccines have often failed to live up to their promise, although recent results with checkpoint inhibitors are reviving hopes that they will soon fulfill their promise. Although mutation-specific vaccines are under development, there is still high interest in an off-the-shelf vaccine to a ubiquitous antigen, such as MUC1, which is aberrantly expressed on most solid and many hematological tumors, including more than 90% of breast carcinomas. Clinical trials for MUC1 have shown variable success, likely because of immunological tolerance to a self-antigen and to poor immunogenicity of tandem repeat peptides. We hypothesized that MUC1 peptides could be optimized, relying on heteroclitic optimizations of potential anchor amino acids with and without tumor-specific glycosylation of the peptides. We have identified novel MUC1 class I peptides that bind to HLA-A*0201 molecules with significantly higher affinity and function than the native MUC1 peptides. These peptides elicited CTLs from normal donors, as well as breast cancer patients, which were highly effective in killing MUC1-expressing MCF-7 breast cancer cells. Each peptide elicited lytic responses in greater than 6/8 of normal individuals and 3/3 breast cancer patients. The CTLs generated against the glycosylated-anchor modified peptides cross reacted with the native MUC1 peptide, STAPPVHNV, suggesting these analog peptides may offer substantial improvement in the design of epitope-based vaccines.
Collapse
|
20
|
Kroemer G, Galluzzi L, Zitvogel L, Fridman WH. Colorectal cancer: the first neoplasia found to be under immunosurveillance and the last one to respond to immunotherapy? Oncoimmunology 2015; 4:e1058597. [PMID: 26140250 DOI: 10.1080/2162402x.2015.1058597] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Accepted: 06/02/2015] [Indexed: 12/13/2022] Open
Abstract
The first study demonstrating that human colorectal carcinoma (CRC) is under robust immunosurveillance was published a decade ago. Today, it is clear that CRC patients with Stage III lesions abundantly infiltrated by effector memory T cells have a better prognosis than subjects with Stage I neoplasms exhibiting no or poor immune infiltration. Thus, immunological parameters have a superior prognostic value for CRC patients than TNM staging or the Dukes classification. In spite of the fact that CRC is the first neoplasia found to be under immunological control, most attempts made so far to cure this malignancy with immunotherapy have failed. With the exception of a minority of lesions characterized by microsatellite instability (MSI), CRC seems to be insensitive to the blockade of immunological checkpoints with monoclonal antibodies (mAbs) specific for cytotoxic T lymphocyte-associated protein 4 (CTLA4), programmed cell death 1 (PDCD1, best known as PD-1) and the PD-1 ligand CD274 (best known as PD-L1). Thus, CRC stands in contrast with an increasing number of malignancies that respond to checkpoint blockers. Efforts should therefore be dedicated to the development of strategies to (re)instate immunosurveillance in patients with MSI- CRC, perhaps based on the identification of novel, locally relevant immunological checkpoints.
Collapse
Affiliation(s)
- Guido Kroemer
- INSERM; U1138 ; Paris, France ; Université Paris Descartes/Paris V; Sorbonne Paris Cité ; Paris, France ; Université Pierre et Marie Curie/Paris VI ; Paris, France ; Equipe 11 labellisée Ligue contre le Cancer; Center de Recherche des Cordeliers ; Paris, France ; Metabolomics and Cell Biology Platforms; Gustave Roussy Comprehensive Cancer Institute ; Villejuif, France ; Pôle de Biologie, Hôpital Européen Georges Pompidou; AP-HP ; Paris, France
| | - Lorenzo Galluzzi
- INSERM; U1138 ; Paris, France ; Université Paris Descartes/Paris V; Sorbonne Paris Cité ; Paris, France ; Université Pierre et Marie Curie/Paris VI ; Paris, France ; Equipe 11 labellisée Ligue contre le Cancer; Center de Recherche des Cordeliers ; Paris, France ; Gustave Roussy Comprehensive Cancer Institute ; Villejuif, France
| | - Laurence Zitvogel
- Gustave Roussy Comprehensive Cancer Institute ; Villejuif, France ; INSERM; U1015; Equipe 11 labellisée Ligue contre le Cancer ; Villejuif, France ; Center of Clinical Investigations in Biotherapies of Cancer 507 (CICBT507) ; Villejuif, France ; Université Paris Sud/Paris XI; Le Kremlin-Bicêtre , France
| | - Wolf Hervé Fridman
- INSERM; U1138 ; Paris, France ; Université Paris Descartes/Paris V; Sorbonne Paris Cité ; Paris, France ; Université Pierre et Marie Curie/Paris VI ; Paris, France ; Equipe 11 labellisée Ligue contre le Cancer; Center de Recherche des Cordeliers ; Paris, France
| |
Collapse
|
21
|
Bae J, Keskin DB, Cowens K, Lee AH, Dranoff G, Munshi NC, Anderson KC. Lenalidomide Polarizes Th1-specific Anti-tumor Immune Response and Expands XBP1 Antigen-Specific Central Memory CD3 +CD8 + T cells against Various Solid Tumors. ACTA ACUST UNITED AC 2015; 3. [PMID: 27668268 PMCID: PMC5032910 DOI: 10.4172/2329-6917.1000178] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Introduction Effective combination immunotherapeutic strategies may be required to enhance effector cells’ anti-tumor activities and improve clinical outcomes. Methods XBP1 antigen-specific cytotoxic T lymphocytes (XBP1-CTL) generated using immunogenic heteroclitic XBP1 US184-192 (YISPWILAV) and XBP1 SP367-375 (YLFPQLISV) peptides or various solid tumor cells over-expressing XBP1 target antigen were evaluated, either alone or in combination with lenalidomide, for phenotype and immune functional activity. Results Lenalidomide treatment of XBP1-CTL increased the proportion of CD45RO+ memory CD3+CD8+ T cells, but not the total CD3+CD8+ T cells. Lenalidomide upregulated critical T cell activation markers and costimulatory molecules (CD28, CD38, CD40L, CD69, ICOS), especially within the central memory CTL subset of XBP1-CTL, while decreasing TCRαβ and T cell checkpoint blockade (CTLA-4, PD-1). Lenalidomide increased the anti-tumor activities of XBP1-CTL memory subsets, which were associated with expression of Th1 transcriptional regulators (T-bet, Eomes) and Akt activation, thereby resulting in enhanced IFN-γ production, granzyme B upregulation and specific CD28/CD38-positive and CTLA-4/PD-1-negative cell proliferation. Conclusions These studies suggest the potential benefit of lenalidomide treatment to boost anti-tumor activities of XBP1-specific CTL against a variety of solid tumors and enhance response to an XBP1-directing cancer vaccine regime.
Collapse
Affiliation(s)
- Jooeun Bae
- Dana-Farber Cancer Institute, Boston, Massachusetts, USA; Harvard Medical School, Boston, Massachusetts, USA
| | - Derin B Keskin
- Dana-Farber Cancer Institute, Boston, Massachusetts, USA; Harvard Medical School, Boston, Massachusetts, USA
| | - Kristen Cowens
- Dana-Farber Cancer Institute, Boston, Massachusetts, USA; Harvard Medical School, Boston, Massachusetts, USA
| | - Ann-Hwee Lee
- Weill Cornell Medical College, New York, NY, USA
| | - Glen Dranoff
- Dana-Farber Cancer Institute, Boston, Massachusetts, USA; Harvard Medical School, Boston, Massachusetts, USA
| | - Nikhil C Munshi
- Dana-Farber Cancer Institute, Boston, Massachusetts, USA; Harvard Medical School, Boston, Massachusetts, USA; VA Boston Healthcare System, Boston, Massachusetts, USA
| | - Kenneth C Anderson
- Dana-Farber Cancer Institute, Boston, Massachusetts, USA; Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|