1
|
Bhutada AS, Adhikari S, Cuoco JA, In A, Rogers CM, Jane JA, Marvin EA. Prognostic Factors and Nomogram for Choroid Plexus Tumors: A Population-Based Retrospective Surveillance, Epidemiology, and End Results Database Analysis. Cancers (Basel) 2024; 16:610. [PMID: 38339361 PMCID: PMC10854689 DOI: 10.3390/cancers16030610] [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: 12/15/2023] [Revised: 01/23/2024] [Accepted: 01/29/2024] [Indexed: 02/12/2024] Open
Abstract
Background: Choroid plexus tumors (CPTs) are rare neoplasms found in the central nervous system, comprising 1% of all brain tumors. These tumors include choroid plexus papilloma (CPP), atypical choroid plexus papilloma (aCPP), and choroid plexus carcinoma (CPC). Although gross total resection for choroid plexus papillomas (CPPs) is associated with long-term survival, there is a scarcity of prospective data concerning the role and sequence of neoadjuvant therapy in treating aCPP and CPC. Methods: From the years 2000 to 2019, 679 patients with CPT were identified from the Surveillance, Epidemiology, and End Result (SEER) database. Among these patients, 456 patients had CPP, 75 patients had aCPP, and 142 patients had CPC. Univariate and multivariable Cox proportional hazard models were run to identify variables that had a significant impact on the primary endpoint of overall survival (OS). A predictive nomogram was built for patients with CPC to predict 5-year and 10-year survival probability. Results: Histology was a significant predictor of OS, with 5-year OS rates of 90, 79, and 61% for CPP, aCPP, and CPC, respectively. Older age and African American race were prognostic for worse OS for patients with CPP. Older age was also associated with reduced OS for patients with aCPP. American Indian/Alaskan Native race was linked to poorer OS for patients with CPC. Overall, treatment with gross total resection or subtotal resection had no difference in OS in patients with CPP or aCPP. Meanwhile, in patients with CPC, gross total resection (GTR) was associated with significantly better OS than subtotal resection (STR) only. However, there is no difference in OS between patients that receive GTR and patients that receive STR with adjuvant therapy. The nomogram for CPC considers types of treatments received. It demonstrates acceptable accuracy in estimating survival probability at 5-year and 10-year intervals, with a C-index of 0.608 (95% CI of 0.446 to 0.77). Conclusions: This is the largest study on CPT to date and highlights the optimal treatment strategies for these rare tumors. Overall, there is no difference in OS with GTR vs. STR in CPP or aCPP. Furthermore, OS is equivalent for CPC with GTR and STR plus adjuvant therapy.
Collapse
Affiliation(s)
- Abhishek S. Bhutada
- Virginia Tech Carilion School of Medicine, 2 Riverside Circle, Roanoke, VA 24016, USA; (S.A.); (J.A.C.); (A.I.); (C.M.R.); (J.A.J.J.); (E.A.M.)
| | - Srijan Adhikari
- Virginia Tech Carilion School of Medicine, 2 Riverside Circle, Roanoke, VA 24016, USA; (S.A.); (J.A.C.); (A.I.); (C.M.R.); (J.A.J.J.); (E.A.M.)
- Department of Neurosurgery, Carilion Clinic, 1906 Belleview Avenue, Roanoke, VA 24014, USA
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
| | - Joshua A. Cuoco
- Virginia Tech Carilion School of Medicine, 2 Riverside Circle, Roanoke, VA 24016, USA; (S.A.); (J.A.C.); (A.I.); (C.M.R.); (J.A.J.J.); (E.A.M.)
- Department of Neurosurgery, Carilion Clinic, 1906 Belleview Avenue, Roanoke, VA 24014, USA
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
| | - Alexander In
- Virginia Tech Carilion School of Medicine, 2 Riverside Circle, Roanoke, VA 24016, USA; (S.A.); (J.A.C.); (A.I.); (C.M.R.); (J.A.J.J.); (E.A.M.)
| | - Cara M. Rogers
- Virginia Tech Carilion School of Medicine, 2 Riverside Circle, Roanoke, VA 24016, USA; (S.A.); (J.A.C.); (A.I.); (C.M.R.); (J.A.J.J.); (E.A.M.)
- Department of Neurosurgery, Carilion Clinic, 1906 Belleview Avenue, Roanoke, VA 24014, USA
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
| | - John A. Jane
- Virginia Tech Carilion School of Medicine, 2 Riverside Circle, Roanoke, VA 24016, USA; (S.A.); (J.A.C.); (A.I.); (C.M.R.); (J.A.J.J.); (E.A.M.)
- Department of Neurosurgery, Carilion Clinic, 1906 Belleview Avenue, Roanoke, VA 24014, USA
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
| | - Eric A. Marvin
- Virginia Tech Carilion School of Medicine, 2 Riverside Circle, Roanoke, VA 24016, USA; (S.A.); (J.A.C.); (A.I.); (C.M.R.); (J.A.J.J.); (E.A.M.)
- Department of Neurosurgery, Carilion Clinic, 1906 Belleview Avenue, Roanoke, VA 24014, USA
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
| |
Collapse
|
2
|
Estevez-Ordonez D, Gary SE, Atchley TJ, Maleknia PD, George JA, Laskay NMB, Gross EG, Devulapalli RK, Johnston JM. Immunotherapy for Pediatric Brain and Spine Tumors: Current State and Future Directions. Pediatr Neurosurg 2022; 58:313-336. [PMID: 36549282 PMCID: PMC10233708 DOI: 10.1159/000528792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 12/15/2022] [Indexed: 12/24/2022]
Abstract
BACKGROUND Brain tumors are the most common solid tumors and the leading cause of cancer-related deaths in children. Incidence in the USA has been on the rise for the last 2 decades. While therapeutic advances in diagnosis and treatment have improved survival and quality of life in many children, prognosis remains poor and current treatments have significant long-term sequelae. SUMMARY There is a substantial need for the development of new therapeutic approaches, and since the introduction of immunotherapy by immune checkpoint inhibitors, there has been an exponential increase in clinical trials to adopt these and other immunotherapy approaches in children with brain tumors. In this review, we summarize the current immunotherapy landscape for various pediatric brain tumor types including choroid plexus tumors, embryonal tumors (medulloblastoma, AT/RT, PNETs), ependymoma, germ cell tumors, gliomas, glioneuronal and neuronal tumors, and mesenchymal tumors. We discuss the latest clinical trials and noteworthy preclinical studies to treat these pediatric brain tumors using checkpoint inhibitors, cellular therapies (CAR-T, NK, T cell), oncolytic virotherapy, radioimmunotherapy, tumor vaccines, immunomodulators, and other targeted therapies. KEY MESSAGES The current landscape for immunotherapy in pediatric brain tumors is still emerging, but results in certain tumors have been promising. In the age of targeted therapy, genetic tumor profiling, and many ongoing clinical trials, immunotherapy will likely become an increasingly effective tool in the neuro-oncologist armamentarium.
Collapse
Affiliation(s)
- Dagoberto Estevez-Ordonez
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, Alabama, USA,
- Division of Pediatric Neurosurgery, Children's of Alabama, Birmingham, Alabama, USA,
| | - Sam E Gary
- Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Travis J Atchley
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, Alabama, USA
- Division of Pediatric Neurosurgery, Children's of Alabama, Birmingham, Alabama, USA
| | - Pedram D Maleknia
- Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Jordan A George
- Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Nicholas M B Laskay
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, Alabama, USA
- Division of Pediatric Neurosurgery, Children's of Alabama, Birmingham, Alabama, USA
| | - Evan G Gross
- Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Rishi K Devulapalli
- Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - James M Johnston
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, Alabama, USA
- Division of Pediatric Neurosurgery, Children's of Alabama, Birmingham, Alabama, USA
| |
Collapse
|
3
|
Memarnejadian A, Meilleur CE, Shaler CR, Khazaie K, Bennink JR, Schell TD, Haeryfar SMM. PD-1 Blockade Promotes Epitope Spreading in Anticancer CD8 + T Cell Responses by Preventing Fratricidal Death of Subdominant Clones To Relieve Immunodomination. THE JOURNAL OF IMMUNOLOGY 2017; 199:3348-3359. [PMID: 28939757 DOI: 10.4049/jimmunol.1700643] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Accepted: 08/26/2017] [Indexed: 12/15/2022]
Abstract
The interactions between programmed death-1 (PD-1) and its ligands hamper tumor-specific CD8+ T cell (TCD8) responses, and PD-1-based "checkpoint inhibitors" have shown promise in certain cancers, thus revitalizing interest in immunotherapy. PD-1-targeted therapies reverse TCD8 exhaustion/anergy. However, whether they alter the epitope breadth of TCD8 responses remains unclear. This is an important question because subdominant TCD8 are more likely than immunodominant clones to escape tolerance mechanisms and may contribute to protective anticancer immunity. We have addressed this question in an in vivo model of TCD8 responses to well-defined epitopes of a clinically relevant oncoprotein, large T Ag. We found that unlike other coinhibitory molecules (CTLA-4, LAG-3, TIM-3), PD-1 was highly expressed by subdominant TCD8, which correlated with their propensity to favorably respond to PD-1/PD-1 ligand-1 (PD-L1)-blocking Abs. PD-1 blockade increased the size of subdominant TCD8 clones at the peak of their primary response, and it also sustained their presence, thus giving rise to an enlarged memory pool. The expanded population was fully functional as judged by IFN-γ production and MHC class I-restricted cytotoxicity. The selective increase in subdominant TCD8 clonal size was due to their enhanced survival, not proliferation. Further mechanistic studies utilizing peptide-pulsed dendritic cells, recombinant vaccinia viruses encoding full-length T Ag or epitope mingenes, and tumor cells expressing T Ag variants revealed that anti-PD-1 invigorates subdominant TCD8 responses by relieving their lysis-dependent suppression by immunodominant TCD8 To our knowledge, our work constitutes the first report that interfering with PD-1 signaling potentiates epitope spreading in tumor-specific responses, a finding with clear implications for cancer immunotherapy and vaccination.
Collapse
Affiliation(s)
- Arash Memarnejadian
- Department of Microbiology and Immunology, Western University, London, Ontario N6A 5C1, Canada
| | - Courtney E Meilleur
- Department of Microbiology and Immunology, Western University, London, Ontario N6A 5C1, Canada
| | - Christopher R Shaler
- Department of Microbiology and Immunology, Western University, London, Ontario N6A 5C1, Canada
| | | | - Jack R Bennink
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Todd D Schell
- Department of Microbiology and Immunology, Pennsylvania State University, Hershey, PA 17033
| | - S M Mansour Haeryfar
- Department of Microbiology and Immunology, Western University, London, Ontario N6A 5C1, Canada; .,Division of Clinical Immunology and Allergy, Department of Medicine, Western University, London, Ontario N6G 5W9, Canada.,Centre for Human Immunology, Western University, London, Ontario N6A 5C1, Canada; and.,Lawson Health Research Institute, London, Ontario N6C 2R5, Canada
| |
Collapse
|
4
|
Cozza EM, Cooper TK, Budgeon LR, Christensen ND, Schell TD. Protection from tumor recurrence following adoptive immunotherapy varies with host conditioning regimen despite initial regression of autochthonous murine brain tumors. Cancer Immunol Immunother 2014; 64:325-36. [PMID: 25408469 DOI: 10.1007/s00262-014-1635-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Accepted: 11/04/2014] [Indexed: 12/18/2022]
Abstract
Adoptive T cell transfer (ACT) has achieved clinical success in treating established cancer, particularly in combination with lymphodepleting regimens. Our group previously demonstrated that ACT following whole-body irradiation (WBI) promotes high-level T cell accumulation, regression of established brain tumors, and long-term protection from tumor recurrence in a mouse model of SV40 T antigen-induced choroid plexus tumors. Here we asked whether an approach that can promote strong donor T-cell responses in the absence of WBI might also produce this dramatic and durable tumor elimination following ACT. Agonist anti-CD40 antibody can enhance antigen-specific CD8(+) T-cell responses and has shown clinical efficacy as a monotherapy in the setting of cancer. We show that anti-CD40 conditioning promotes rapid accumulation of tumor-specific donor CD8(+) T cells in the brain and regression of autochthonous T antigen-induced choroid plexus tumors, similar to WBI. Despite a significant increase in the lifespan, tumors eventually recurred in anti-CD40-conditioned mice coincident with loss of T-cell persistence from both the brain and lymphoid organs. Depletion of CD8(+) T cells from the peripheral lymphoid organs of WBI-conditioned recipients failed to promote tumor recurrence, but donor cells persisted in the brains long-term in CD8-depleted mice. These results demonstrate that anti-CD40 conditioning effectively enhances ACT-mediated acute elimination of autochthonous tumors, but suggest that mechanisms associated with WBI conditioning, such as the induction of long-lived T cells, may be critical for protection from tumor recurrence.
Collapse
Affiliation(s)
- Eugene M Cozza
- Department of Microbiology and Immunology, Penn State Hershey College of Medicine, 500 University Drive, H107, Hershey, PA, 17033, USA
| | | | | | | | | |
Collapse
|
5
|
Rytelewski M, Meilleur CE, Atef Yekta M, Szabo PA, Garg N, Schell TD, Jevnikar AM, Sharif S, Singh B, Haeryfar SMM. Suppression of immunodominant antitumor and antiviral CD8+ T cell responses by indoleamine 2,3-dioxygenase. PLoS One 2014; 9:e90439. [PMID: 24587363 PMCID: PMC3938761 DOI: 10.1371/journal.pone.0090439] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2013] [Accepted: 01/30/2014] [Indexed: 11/19/2022] Open
Abstract
Indoleamine 2,3-dioxygenase (IDO) is a tryptophan-degrading enzyme known to suppress antitumor CD8+ T cells (TCD8). The role of IDO in regulation of antiviral TCD8 responses is far less clear. In addition, whether IDO controls both immunodominant and subdominant TCD8 is not fully understood. This is an important question because the dominance status of tumor- and virus-specific TCD8 may determine their significance in protective immunity and in vaccine design. We evaluated the magnitude and breadth of cross-primed TCD8 responses to simian virus 40 (SV40) large T antigen as well as primary and recall TCD8 responses to influenza A virus (IAV) in the absence or presence of IDO. IDO−/− mice and wild-type mice treated with 1-methyl-D-tryptophan, a pharmacological inhibitor of IDO, exhibited augmented responses to immunodominant epitopes encoded by T antigen and IAV. IDO-mediated suppression of these responses was independent of CD4+CD25+FoxP3+ regulatory T cells, which remained numerically and functionally intact in IDO−/− mice. Treatment with L-kynurenine failed to inhibit TCD8 responses, indicating that tryptophan metabolites are not responsible for the suppressive effect of IDO in our models. Immunodominant T antigen-specific TCD8 from IDO−/− mice showed increased Ki-67 expression, suggesting that they may have acquired a more vigorous proliferative capacity in vivo. In conclusion, IDO suppresses immunodominant TCD8 responses to tumor and viral antigens. Our work also demonstrates that systemic primary and recall TCD8 responses to IAV are controlled by IDO. Inhibition of IDO thus represents an attractive adjuvant strategy in boosting anticancer and antiviral TCD8 targeting highly immunogenic antigens.
Collapse
MESH Headings
- Animals
- Antigens, Polyomavirus Transforming/immunology
- Antigens, Viral/immunology
- CD4 Antigens/genetics
- CD4 Antigens/immunology
- CD8-Positive T-Lymphocytes/cytology
- CD8-Positive T-Lymphocytes/drug effects
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- Forkhead Transcription Factors/genetics
- Forkhead Transcription Factors/immunology
- Gene Expression
- Immune Tolerance/genetics
- Immunity, Innate
- Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors
- Indoleamine-Pyrrole 2,3,-Dioxygenase/deficiency
- Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics
- Indoleamine-Pyrrole 2,3,-Dioxygenase/immunology
- Influenza A virus/immunology
- Interleukin-2 Receptor alpha Subunit/genetics
- Interleukin-2 Receptor alpha Subunit/immunology
- Kynurenine/pharmacology
- Lymphocyte Activation
- Mice
- Mice, Knockout
- T-Lymphocytes, Regulatory/cytology
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- Tryptophan/analogs & derivatives
- Tryptophan/pharmacology
Collapse
Affiliation(s)
- Mateusz Rytelewski
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada
| | - Courtney E. Meilleur
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada
| | - Maryam Atef Yekta
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada
| | - Peter A. Szabo
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada
| | - Nitan Garg
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada
| | - Todd D. Schell
- Department of Microbiology and Immunology, The Pennsylvania State University, Hershey, Pennsylvania, United States of America
| | - Anthony M. Jevnikar
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada
- Department of Medicine, Western University, London, Ontario, Canada
- Department of Pathology, Western University, London, Ontario, Canada
- Centre for Human Immunology, Western University, London, Ontario, Canada
| | - Shayan Sharif
- Department of Pathobiology, University of Guelph, Guelph, Ontario, Canada
| | - Bhagirath Singh
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada
- Centre for Human Immunology, Western University, London, Ontario, Canada
| | - S. M. Mansour Haeryfar
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada
- Centre for Human Immunology, Western University, London, Ontario, Canada
- * E-mail:
| |
Collapse
|
6
|
Yorty JL. Short-term corticosterone treatment decreases the early CD8+ T cell response to simian virus 40 tumor antigen but has no impact on the late CD8+ T cell response. Brain Behav Immun 2013; 28:139-48. [PMID: 23164951 DOI: 10.1016/j.bbi.2012.11.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2012] [Revised: 11/09/2012] [Accepted: 11/09/2012] [Indexed: 10/27/2022] Open
Abstract
CD8+ T cells (T(CD8)) help control tumor growth in vivo through recognition of distinct tumor antigens and cytolysis of tumor cells. The T(CD8) immune response in C57BL/6 mice to the Simian Virus 40 oncoprotein, large tumor antigen (Tag), targets multiple epitopes and is well-characterized. Epitope IV, an H-2K(b)-restricted epitope, is immunodominant while epitope I, an H-2D(b)-restricted epitope is subdominant. GCs alter many aspects of T cell function. Indeed, the current studies demonstrate that exposure of mice to the immunosuppressive GC, corticosterone (CORT), over the entire course of the primary immune response limits activation of endogenous Tag-specific T(CD8). Even short-term CORT treatment from day -1 to day +2 post-immunization significantly reduced splenic size and the absolute number of Tag-specific T(CD8) on day 6 post-immunization. In vivo killing activity was also reduced. However, by day 10 post-immunization, the peak of the immune response, the absolute number of Tag-specific T(CD8) and their in vivo killing of epitope I or epitope IV-expressing target cells had recovered in CORT treated mice. Adoptive transfer of transgenic T cells post-CORT removal demonstrated that CORT decreased the ability of the endogenous antigen-presenting cells to induce proliferation of the exogenous transgenic T cells. Combined, these studies have implications about the timing of clinical steroid treatment relative to immunization or adoptive transfer for cancer immunotherapy.
Collapse
Affiliation(s)
- Jodi L Yorty
- Department of Biology, Elizabethtown College, One Alpha Drive, Elizabethtown, PA 17022, United States.
| |
Collapse
|
7
|
Watson AM, Mylin LM, Thompson MM, Schell TD. Modification of a tumor antigen determinant to improve peptide/MHC stability is associated with increased immunogenicity and cross-priming a larger fraction of CD8+ T cells. THE JOURNAL OF IMMUNOLOGY 2012; 189:5549-60. [PMID: 23175697 DOI: 10.4049/jimmunol.1102221] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Altered peptide ligands (APLs) with enhanced binding to MHC class I can increase the CD8(+) T cell response to native Ags, including tumor Ags. In this study, we investigate the influence of peptide-MHC (pMHC) stability on recruitment of tumor Ag-specific CD8(+) T cells through cross-priming. Among the four known H-2(b)-restricted CD8(+) T cell determinants within SV40 large tumor Ag (TAg), the site V determinant ((489)QGINNLDNL(497)) forms relatively low-stability pMHC and is characteristically immunorecessive. Absence of detectable site V-specific CD8(+) T cells following immunization with wild-type TAg is due in part to inefficient cross-priming. We mutated nonanchor residues within the TAg site V determinant that increased pMHC stability but preserved recognition by both TCR-transgenic and polyclonal endogenous T cells. Using a novel approach to quantify the fraction of naive T cells triggered through cross-priming in vivo, we show that immunization with TAg variants expressing higher-stability determinants increased the fraction of site V-specific T cells cross-primed and effectively overcame the immunorecessive phenotype. In addition, using MHC class I tetramer-based enrichment, we demonstrate for the first time, to our knowledge, that endogenous site V-specific T cells are primed following wild-type TAg immunization despite their low initial frequency, but that the magnitude of T cell accumulation is enhanced following immunization with a site V variant TAg. Our results demonstrate that site V APLs cross-prime a higher fraction of available T cells, providing a potential mechanism for high-stability APLs to enhance immunogenicity and accumulation of T cells specific for the native determinant.
Collapse
Affiliation(s)
- Alan M Watson
- Department of Microbiology and Immunology, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | | | | | | |
Collapse
|
8
|
Tatum AM, Watson AM, Schell TD. Direct presentation regulates the magnitude of the CD8+ T cell response to cell-associated antigen through prolonged T cell proliferation. THE JOURNAL OF IMMUNOLOGY 2010; 185:2763-72. [PMID: 20660711 DOI: 10.4049/jimmunol.0903920] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The magnitude and complexity of Ag-specific CD8(+) T cell responses is determined by intrinsic properties of the immune system and extrinsic factors, such as vaccination. We evaluated mechanisms that regulate the CD8(+) T cell response to two distinct determinants derived from the same protein Ag, SV40 T Ag (T Ag), following immunization of C57BL/6 mice with T Ag-transformed cells. The results show that direct presentation of T cell determinants by T Ag-transformed cells regulates the magnitude of the CD8(+) T cell response in vivo but not the immunodominance hierarchy. The immunodominance hierarchy was reversed in a dose-dependent manner by addition of excess naive T cells targeting the subdominant determinant. However, T cell competition played only a minor role in limiting T cell accumulation under physiological conditions. We found that the magnitude of the T cell response was regulated by the ability of T Ag-transformed cells to directly present the T Ag determinants. The hierarchy of the CD8(+) T cell response was maintained when Ag presentation in vivo was restricted to cross-presentation, but the presence of T Ag-transformed cells capable of direct presentation dramatically enhanced T cell accumulation at the peak of the response. This enhancement was due to a prolonged period of T cell proliferation, resulting in a delay in T cell contraction. Our findings reveal that direct presentation by nonprofessional APCs can dramatically enhance accumulation of CD8(+) T cells during the primary response, revealing a potential strategy to enhance vaccination approaches.
Collapse
Affiliation(s)
- Angela M Tatum
- Department of Microbiology and Immunology, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | | | | |
Collapse
|
9
|
Hamzah J, Altin JG, Herringson T, Parish CR, Hämmerling GJ, O'Donoghue H, Ganss R. Targeted liposomal delivery of TLR9 ligands activates spontaneous antitumor immunity in an autochthonous cancer model. THE JOURNAL OF IMMUNOLOGY 2009; 183:1091-8. [PMID: 19561111 DOI: 10.4049/jimmunol.0900736] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Accessibility of tumors for highly effective local treatment represents a major challenge for anticancer therapy. Immunostimulatory oligodeoxynucleotides (ODN) with CpG motifs are ligands of TLR9, which prime spontaneous antitumor immunity, but are less effective when applied systemically. We therefore developed a liposome-based agent for selective delivery of CpG-ODN into the tumor environment. A peptide that specifically targets angiogenic endothelial cells in a transgenic tumor model for islet cell carcinogenesis was engrafted into CpG-ODN containing liposomes. Intravenous injection of these liposomes resulted in specific accumulation around tumor vessels, increased uptake by tumor-resident macrophages, and retention over time. In contrast, nontargeted liposomes did not localize to the tumor vasculature. Consequently, only vascular targeting of CpG-ODN liposomes provoked a marked inflammatory response at vessel walls with enhanced CD8(+) and CD4(+) T cell infiltration and, importantly, activation of spontaneous, tumor-specific cytotoxicity. In a therapeutic setting, 40% of tumor-bearing, transgenic mice survived beyond week 45 after systemic administration of vascular-directed CpG-ODN liposomes. In contrast, control mice survived up to 30 wk. Therapeutic efficacy was further improved by increasing the frequency of tumor-specific effector cells through adoptive transfers. NK cells and CD8(+) T cells were major effectors which induced tumor cell death and acted in conjunction with antivascular effects. Thus, tumor homing with CpG-ODN-loaded liposomes is as potent as direct injection of free CpG-ODN and has the potential to overcome some major limitations of conventional CpG-ODN monotherapy.
Collapse
Affiliation(s)
- Juliana Hamzah
- Western Australian Institute for Medical Research, University of Western Australia Centre for Medical Research, Perth, Australia
| | | | | | | | | | | | | |
Collapse
|
10
|
Tatum AM, Mylin LM, Bender SJ, Fischer MA, Vigliotti BA, Tevethia MJ, Tevethia SS, Schell TD. CD8+ T cells targeting a single immunodominant epitope are sufficient for elimination of established SV40 T antigen-induced brain tumors. THE JOURNAL OF IMMUNOLOGY 2008; 181:4406-17. [PMID: 18768900 DOI: 10.4049/jimmunol.181.6.4406] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Immunotherapy of established solid tumors is rarely achieved, and the mechanisms leading to success remain to be elucidated. We previously showed that extended control of advanced-stage autochthonous brain tumors is achieved following adoptive transfer of naive C57BL/6 splenocytes into sublethally irradiated line SV11 mice expressing the SV40 T Ag (T Ag) oncoprotein, and was associated with in vivo priming of CD8(+) T cells (T(CD8)) specific for the dominant epitope IV (T Ag residues 404-411). Using donor lymphocytes derived from mice that are tolerant to epitope IV or a newly characterized transgenic mouse line expressing an epitope IV-specific TCR, we show that epitope IV-specific T(CD8) are a necessary component of the donor pool and that purified naive epitope IV-specific T(CD8) are sufficient to promote complete and rapid regression of established tumors. While transfer of naive TCR-IV cells alone induced some initial tumor regression, increased survival of tumor-bearing mice required prior conditioning of the host with a sublethal dose of gamma irradiation and was associated with complete tumor eradication. Regression of established tumors was associated with rapid accumulation of TCR-IV T cells within the brain following initial priming against the endogenous T Ag in the peripheral lymphoid organs. Additionally, persistence of functional TCR-IV cells in both the brain and peripheral lymphoid organs was associated with long-term tumor-free survival. Finally, we show that production of IFN-gamma, but not perforin or TNF-alpha, by the donor lymphocytes is critical for control of autochthonous brain tumors.
Collapse
Affiliation(s)
- Angela M Tatum
- Department of Microbiology and Immunology, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | | | | | | | | | | | | | | |
Collapse
|
11
|
Couty JP, Crain AM, Gerbaud S, Labasque M, Marchiol C, Fradelizi D, Boudaly S, Guettier C, Vignuzzi M, van der Werf S, Escriou N, Viguier M. Delivery of mengovirus-derived RNA replicons into tumoural liver enhances the anti-tumour efficacy of a peripheral peptide-based vaccine. Cancer Immunol Immunother 2008; 57:1161-71. [PMID: 18256833 PMCID: PMC11030607 DOI: 10.1007/s00262-007-0448-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2007] [Accepted: 12/27/2007] [Indexed: 01/22/2023]
Abstract
Hepatocellular carcinoma is a deadly cancer with growing incidence for which immunotherapy is one of the most promising therapeutic approach. Peptide-based vaccines designed to induce strong, sustained CD8+ T cell responses are effective in animal models and cancer patients. We demonstrated the efficacy of curative peptide-based immunisation against a unique epitope of SV40 tumour antigen, through the induction of a strong CD8+ T cell-specific response, in our liver tumour model. However, as in human clinical trials, most tumour antigen epitopes did not induce a therapeutic effect, despite inducing strong CD8+ T cell responses. We therefore modified the tumour environment to enhance peptide-based vaccine efficacy by delivering mengovirus (MV)-derived RNA autoreplicating sequences (MV-RNA replicons) into the liver. The injection of replication-competent RNA replicons into the liver converted partial tumour regression into tumour eradication, whereas non-replicating RNA had no such effect. Replicating RNA replicon injection induced local recruitment of innate immunity effectors (NK and NKT) to the tumour and did not affect specific CD8+ T cell populations or other myelolymphoid subsets. The local delivery of such RNA replicons into tumour stroma is therefore a promising strategy complementary to the use of peripheral peptide-based vaccines for treating liver tumours.
Collapse
Affiliation(s)
- Jean-Pierre Couty
- Institut Cochin, Département Endocrinologie-Métabolisme et Cancer (EMC), Institut National de la Santé et de la Recherche Médicale INSERM U567, 24, rue du Faubourg Saint-Jacques, 75014 Paris, France
- Centre National de la Recherche Scientifique, CNRS UMR 8104, 24, rue du Faubourg Saint-Jacques, 75014 Paris, France
- Université Paris-Descartes, Bâtiment Faculté, 24, rue du Faubourg Saint-Jacques, 75014 Paris, France
- Université Paris Diderot, 2, place Jussieu, 75251 Paris Cedex 05, France
| | - Anne-Marie Crain
- Institut Cochin, Département Endocrinologie-Métabolisme et Cancer (EMC), Institut National de la Santé et de la Recherche Médicale INSERM U567, 24, rue du Faubourg Saint-Jacques, 75014 Paris, France
- Centre National de la Recherche Scientifique, CNRS UMR 8104, 24, rue du Faubourg Saint-Jacques, 75014 Paris, France
- Université Paris-Descartes, Bâtiment Faculté, 24, rue du Faubourg Saint-Jacques, 75014 Paris, France
- Université Paris Diderot, 2, place Jussieu, 75251 Paris Cedex 05, France
| | - Sylvie Gerbaud
- Institut Pasteur, Unité de Génétique Moléculaire des Virus Respiratoires, Unité de Recherche Associée URA 1966, Centre National de la Recherche Scientifique, CNRS, 25, rue du Docteur Roux, 75724 Paris Cedex 15, France
- Université Paris Diderot, Laboratoire associé, 2 Place Jussieu, 75251 Paris Cedex 05, France
| | - Marilyne Labasque
- Institut Cochin, Département Endocrinologie-Métabolisme et Cancer (EMC), Institut National de la Santé et de la Recherche Médicale INSERM U567, 24, rue du Faubourg Saint-Jacques, 75014 Paris, France
- Centre National de la Recherche Scientifique, CNRS UMR 8104, 24, rue du Faubourg Saint-Jacques, 75014 Paris, France
- Université Paris-Descartes, Bâtiment Faculté, 24, rue du Faubourg Saint-Jacques, 75014 Paris, France
- Université Paris Diderot, 2, place Jussieu, 75251 Paris Cedex 05, France
| | - Carmen Marchiol
- Institut Cochin, Département Endocrinologie-Métabolisme et Cancer (EMC), Institut National de la Santé et de la Recherche Médicale INSERM U567, 24, rue du Faubourg Saint-Jacques, 75014 Paris, France
- Centre National de la Recherche Scientifique, CNRS UMR 8104, 24, rue du Faubourg Saint-Jacques, 75014 Paris, France
- Université Paris-Descartes, Bâtiment Faculté, 24, rue du Faubourg Saint-Jacques, 75014 Paris, France
| | - Didier Fradelizi
- Institut Cochin, Département Endocrinologie-Métabolisme et Cancer (EMC), Institut National de la Santé et de la Recherche Médicale INSERM U567, 24, rue du Faubourg Saint-Jacques, 75014 Paris, France
- Centre National de la Recherche Scientifique, CNRS UMR 8104, 24, rue du Faubourg Saint-Jacques, 75014 Paris, France
- Université Paris-Descartes, Bâtiment Faculté, 24, rue du Faubourg Saint-Jacques, 75014 Paris, France
| | - Sarah Boudaly
- Université Paris Diderot, 2, place Jussieu, 75251 Paris Cedex 05, France
- Laboratoire d’Immunologie Cellulaire et Immunopathologie de l’Ecole Pratique des Hautes Etudes, Institut Universitaire d’Hématologie, Unité Mixte de Recherche UMR 7151, Centre National de la Recherche Scientifique, CNRS, Hôpital Saint-Louis, 75010 Paris, France
| | - Catherine Guettier
- Centre Hépatobiliaire, laboratoire d’Anatomie Pathologique, Hôpital Paul Brousse, 14, avenue Paul Vaillant Couturier, 94804 Villejuif, France
| | - Marco Vignuzzi
- Institut Pasteur, Unité de Génétique Moléculaire des Virus Respiratoires, Unité de Recherche Associée URA 1966, Centre National de la Recherche Scientifique, CNRS, 25, rue du Docteur Roux, 75724 Paris Cedex 15, France
- Université Paris Diderot, Laboratoire associé, 2 Place Jussieu, 75251 Paris Cedex 05, France
| | - Sylvie van der Werf
- Institut Pasteur, Unité de Génétique Moléculaire des Virus Respiratoires, Unité de Recherche Associée URA 1966, Centre National de la Recherche Scientifique, CNRS, 25, rue du Docteur Roux, 75724 Paris Cedex 15, France
- Université Paris Diderot, Laboratoire associé, 2 Place Jussieu, 75251 Paris Cedex 05, France
| | - Nicolas Escriou
- Institut Pasteur, Unité de Génétique Moléculaire des Virus Respiratoires, Unité de Recherche Associée URA 1966, Centre National de la Recherche Scientifique, CNRS, 25, rue du Docteur Roux, 75724 Paris Cedex 15, France
- Université Paris Diderot, Laboratoire associé, 2 Place Jussieu, 75251 Paris Cedex 05, France
| | - Mireille Viguier
- Institut Cochin, Département Endocrinologie-Métabolisme et Cancer (EMC), Institut National de la Santé et de la Recherche Médicale INSERM U567, 24, rue du Faubourg Saint-Jacques, 75014 Paris, France
- Centre National de la Recherche Scientifique, CNRS UMR 8104, 24, rue du Faubourg Saint-Jacques, 75014 Paris, France
- Université Paris-Descartes, Bâtiment Faculté, 24, rue du Faubourg Saint-Jacques, 75014 Paris, France
- Université Paris Diderot, 2, place Jussieu, 75251 Paris Cedex 05, France
| |
Collapse
|
12
|
Otahal P, Knowles BB, Tevethia SS, Schell TD. Anti-CD40 conditioning enhances the T(CD8) response to a highly tolerogenic epitope and subsequent immunotherapy of simian virus 40 T antigen-induced pancreatic tumors. THE JOURNAL OF IMMUNOLOGY 2007; 179:6686-95. [PMID: 17982058 DOI: 10.4049/jimmunol.179.10.6686] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Rapid loss of adoptively transferred tumor-specific CD8(+) T cells (T(CD8)) following Ag recognition in the periphery and their limited accumulation within the tumor stroma reduces the effectiveness of T cell-based immunotherapy. To better understand the role of T(CD8) in the control of autochthonous tumors, we have used mice of the RIP1-Tag4 lineage that develop pancreatic beta cell tumors due to expression of the SV40 large T Ag from the rat insulin promoter. We previously showed that the kinetics of functional T(CD8) tolerance varies toward two distinct epitopes derived from T Ag. Epitope I ((206)SAINNYAQKL(215))-specific T(CD8) are rapidly deleted whereas T(CD8) targeting epitope IV ((404)VVYDFLKC(411)) persist over the lifetime of tumor-bearing animals. In this report, we show that the conditioning of tumor-bearing RIP1-Tag4 mice with agonistic anti-CD40 Ab induces extensive expansion of naive epitope I-specific TCR transgenic (TCR-I) T cells in this tolerogenic environment and delays their loss from the host. In addition, functional TCR-I T cells intensively infiltrate pancreatic tumors, resulting in increased survival of RIP1-Tag4 mice. These results suggest that a similar approach could effectively enhance T cell-based immunotherapies to cancer when targeting other highly tolerogenic epitopes.
Collapse
Affiliation(s)
- Pavel Otahal
- Department of Microbiology and Immunology, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | | | | | | |
Collapse
|
13
|
Yorty JL, Tevethia SS, Schell TD. Rapid accumulation of adoptively transferred CD8+ T cells at the tumor site is associated with long-term control of SV40 T antigen-induced tumors. Cancer Immunol Immunother 2007; 57:883-95. [PMID: 18004562 DOI: 10.1007/s00262-007-0424-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2007] [Accepted: 10/29/2007] [Indexed: 01/09/2023]
Abstract
We previously established a model to study CD8(+) T cell (T(CD8))-based adoptive immunotherapy of cancer using line SV11 mice that develop choroid plexus tumors in the brain due to transgenic expression of Simian Virus 40 large T antigen (Tag). These mice are tolerant to the three dominant T(CD8)-recognized Tag epitopes I, II/III and IV. However, adoptive transfer of spleen cells from naïve C57BL/6 (B6) mice prolongs SV11 survival following T(CD8) priming against the endogenous Tag epitope IV. In addition, survival of SV11 mice is dramatically increased following transfer of lymphocytes from Tag-immune B6 mice. In the current study, we compared the kinetics and magnitude of Tag-specific T(CD8) accumulation at the tumor site following adoptive transfer with a high dose of either Tag-immune or naïve donor cells or decreasing doses of Tag-immune lymphocytes. Following adoptive transfer of Tag-immune cells, epitope I- and IV-specific T(CD8) accumulated to high levels in the brain of SV11 mice, peaking at 5-7 days, while epitope IV-specific T(CD8 )derived from naïve donors required three weeks to achieve peak levels. A similar delay in the peak of epitope IV-specific T(CD8) accumulation was observed when tenfold fewer Tag-immune donor cells were administered, reducing control of tumor progression. These results suggest that efficient and prolonged control of established autochthonous tumors is associated with high-level early accumulation of adoptively transferred T cells. We also provide evidence that although multiple specificities are represented in the Tag immune donor lymphocytes, epitope IV-specific donor T(CD8) play a predominant role in control of tumor growth.
Collapse
Affiliation(s)
- Jodi L Yorty
- Department of Microbiology and Immunology, H107, The Pennsylvania State University College of Medicine, 500 University Drive, Hershey, PA 17033, USA
| | | | | |
Collapse
|
14
|
Abstract
With respect to CD8 effector T cells, interleukin-12 (IL-12) and transforming growth factor beta (TGFbeta) are 2 cytokines that exert opposing effects. IL-12 promotes antitumor immune responses by augmenting activated CD8 T-cell proliferation and interferon-gamma secretion. Conversely, TGFbeta generates a permissive environment for cancer growth, in part by antagonizing the effects of immunomodulatory cytokines, including IL-12. We demonstrate that TGFbeta-resistant T cells are capable of sustaining IL-12-induced mitogenesis and interferon-gamma secretion in a TGFbeta-rich milieu. Furthermore, in 2 murine tumor models associated with high TGFbeta1 levels in the local microenvironment, treatment with IL-12 and adoptively transferred TGFbeta-resistant T cells provided improved survival times. These results suggest that combining IL-12 with TGFbeta neutralization strategies may be effective in enhancing antitumor immune responses.
Collapse
MESH Headings
- Animals
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/pathology
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/pathology
- Cell Line
- Cell Proliferation
- Female
- Immunotherapy, Adoptive
- Interferon-gamma/biosynthesis
- Interleukin-12/immunology
- Interleukin-12/pharmacology
- Male
- Melanoma, Experimental/mortality
- Melanoma, Experimental/pathology
- Melanoma, Experimental/therapy
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Neoplasms, Experimental/mortality
- Neoplasms, Experimental/pathology
- Neoplasms, Experimental/therapy
- Papilloma, Choroid Plexus/mortality
- Papilloma, Choroid Plexus/pathology
- Papilloma, Choroid Plexus/therapy
- Receptors, Transforming Growth Factor beta/genetics
- T-Lymphocytes/immunology
- T-Lymphocytes/transplantation
- Transforming Growth Factor beta1/immunology
Collapse
Affiliation(s)
- Timothy M Fan
- Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, 1008 West Hazelwood Drive, Urbana, IL 61801, USA.
| | | | | |
Collapse
|
15
|
Mylin LM, Schell TD, Epler M, Kusuma C, Assis D, Matsko C, Smith A, Allebach A, Tevethia SS. Diversity of escape variant mutations in Simian virus 40 large tumor antigen (SV40 Tag) epitopes selected by cytotoxic T lymphocyte (CTL) clones. Virology 2007; 364:155-68. [PMID: 17368499 PMCID: PMC3866617 DOI: 10.1016/j.virol.2007.02.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2006] [Revised: 01/23/2007] [Accepted: 02/11/2007] [Indexed: 01/22/2023]
Abstract
To better understand the relationship between epitope variation and tumor escape from immune surveillance, SV40 T antigen-transformed B6/K-0 cells were subjected to selection with individual CTL clones specific for the SV40 T antigen H-2D(b)-restricted epitopes I or V. CTL-resistant populations were isolated from a majority of the selection cultures and substituted epitope sequences were identified within most of the resistant populations. Tag sequences deleted of all or portions of the selection-targeted epitope were identified, but in lower numbers compared to epitope sequences bearing single residue substitutions. Relatively few flanking residue substitutions were identified, and only in epitope I-targeted selections. The diversity (numbers and epitope residue locations) of substituted epitope residue positions varied between selections. These findings suggest that the scope of spontaneously occurring mutations that could allow for escape from individual CD8+ T cell clones is large.
Collapse
Affiliation(s)
- Lawrence M. Mylin
- Department of Microbiology and Immunology H107, The Pennsylvania State University College of Medicine, Hershey, PA 17033
- Department of Biological Sciences, Box 3030, Messiah College, Grantham, PA 17027
| | - Todd D. Schell
- Department of Microbiology and Immunology H107, The Pennsylvania State University College of Medicine, Hershey, PA 17033
| | - Melanie Epler
- Department of Microbiology and Immunology H107, The Pennsylvania State University College of Medicine, Hershey, PA 17033
| | - Carolyn Kusuma
- Department of Biological Sciences, Box 3030, Messiah College, Grantham, PA 17027
| | - David Assis
- Department of Biological Sciences, Box 3030, Messiah College, Grantham, PA 17027
| | - Chelsea Matsko
- Department of Biological Sciences, Box 3030, Messiah College, Grantham, PA 17027
| | - Alexandra Smith
- Department of Biological Sciences, Box 3030, Messiah College, Grantham, PA 17027
| | - April Allebach
- Department of Biological Sciences, Box 3030, Messiah College, Grantham, PA 17027
| | - Satvir S. Tevethia
- Department of Microbiology and Immunology H107, The Pennsylvania State University College of Medicine, Hershey, PA 17033
- Corresponding author. Tele: (717) 531-8872; Fax: (717) 531-6522; E-mail:
| |
Collapse
|
16
|
Nguyen van Binh P, Duc HT. Analyses and perspectives in cancer immunotherapy. Biomed Pharmacother 2006; 60:621-8. [PMID: 16978826 DOI: 10.1016/j.biopha.2006.07.092] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2006] [Accepted: 07/28/2006] [Indexed: 01/15/2023] Open
Abstract
Since the last two decades, rapid progress has been made in the field of cancer immunotherapy relevant to manipulation of adaptative cytotoxic T lymphocytes (CTLs) and innate immunity natural killer (NK) cells as well as antibodies. Many possibilities are now offered for therapeutic purposes contributing to better approaches in treatment of cancer.
Collapse
Affiliation(s)
- P Nguyen van Binh
- Inserm, U602, Micro-environnement et Physiopathologie de la Différenciation, Hôpital Paul-Brousse, 16, avenue Paul-Vaillant-Couturier, 94807 Villejuif cedex, France
| | | |
Collapse
|
17
|
Otahal P, Schell TD, Hutchinson SC, Knowles BB, Tevethia SS. Early Immunization Induces Persistent Tumor-Infiltrating CD8+T Cells against an Immunodominant Epitope and Promotes Lifelong Control of Pancreatic Tumor Progression in SV40 Tumor Antigen Transgenic Mice. THE JOURNAL OF IMMUNOLOGY 2006; 177:3089-99. [PMID: 16920946 DOI: 10.4049/jimmunol.177.5.3089] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The ability to recruit the host's CD8+ T lymphocytes (T(CD8)) against cancer is often limited by the development of peripheral tolerance toward the dominant tumor-associated Ags. Because multiple epitopes derived from a given tumor Ag (T Ag) can be targeted by T(CD8), vaccine approaches should be directed toward those T(CD8) that are more likely to survive under conditions of persistent Ag expression. In this study, we investigated the effect of peripheral tolerance on the endogenous T(CD8) response toward two epitopes, designated epitopes I and IV, from the SV40 large T Ag. Using rat insulin promoter (RIP) 1-Tag4 transgenic mice that express T Ag from the RIP and develop pancreatic insulinomas, we demonstrate that epitope IV- but not epitope I-specific T(CD8) are maintained long term in tumor-bearing RIP1-Tag4 mice. Even large numbers of TCR-transgenic T cells specific for epitope I were rapidly eliminated from RIP1-Tag4 mice after adoptive transfer and recognition of the endogenous T Ag. Importantly, immunization of RIP1-Tag4 mice at 5 wk of age against epitope IV resulted in complete protection from tumor progression over a 2-year period despite continued expression of T Ag in the pancreas. This extensive control of tumor progression was associated with the persistence of functional epitope IV-specific T(CD8) within the pancreas for the lifetime of the mice without the development of diabetes. This study indicates that an equilibrium is reached in which immune surveillance for spontaneous cancer can be achieved for the lifespan of the host while maintaining normal organ function.
Collapse
Affiliation(s)
- Pavel Otahal
- Department of Microbiology and Immunology, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | | | | | | | | |
Collapse
|
18
|
Ryan CM, Schell TD. Accumulation of CD8+T Cells in Advanced-Stage Tumors and Delay of Disease Progression following Secondary Immunization against an Immunorecessive Epitope. THE JOURNAL OF IMMUNOLOGY 2006; 177:255-67. [PMID: 16785521 DOI: 10.4049/jimmunol.177.1.255] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Self-reactive T cells that survive the process of positive and negative selection during thymocyte development represent potential effector cells against tumors that express these same self-Ags. We have previously shown that CD8+ T lymphocytes (T(CD8)) specific for an immunorecessive epitope, designated epitope V, from the SV40 large T Ag (Tag) escape thymic deletion in line SV11 Tag-transgenic mice. In contrast, these mice are tolerant to the three most dominant Tag epitopes. The majority of the residual epitope V-specific T(CD8) have a low avidity for the target epitope, but a prime/boost regimen can expand higher avidity clones in vivo. Whether higher avidity T(CD8) targeting this epitope are affected by Tag-expressing tumors in the periphery or can be recruited for control of tumor progression remains unknown. In the current study, we determined the fate of naive TCR-transgenic T(CD8) specific for Tag epitope V (TCR-V cells) following transfer into SV11 mice bearing advanced-stage choroid plexus tumors. The results indicate that TCR-V cells are rapidly triggered by the endogenous Tag and acquire effector function, but fail to accumulate within the tumors. Primary immunization enhanced TCR-V cell frequency in the periphery and promoted entry into the brain, but a subsequent booster immunization caused a dramatic accumulation of TCR-V T cells within the tumors and inhibited tumor progression. These results indicate that epitope V provides a target for CD8+ T cells against spontaneous tumors in vivo, and suggests that epitopes with similar properties can be harnessed for tumor immunotherapy.
Collapse
MESH Headings
- Adoptive Transfer
- Animals
- Antigens, Polyomavirus Transforming/administration & dosage
- Antigens, Polyomavirus Transforming/biosynthesis
- Antigens, Polyomavirus Transforming/genetics
- Antigens, Polyomavirus Transforming/immunology
- Brain Neoplasms/immunology
- Brain Neoplasms/mortality
- Brain Neoplasms/pathology
- Brain Neoplasms/prevention & control
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/pathology
- CD8-Positive T-Lymphocytes/transplantation
- Cancer Vaccines/administration & dosage
- Cancer Vaccines/genetics
- Cancer Vaccines/immunology
- Cell Line, Tumor
- Cell Movement/genetics
- Cell Movement/immunology
- Choroid Plexus/immunology
- Choroid Plexus/pathology
- Cytotoxicity Tests, Immunologic
- Disease Progression
- Epitopes, T-Lymphocyte/administration & dosage
- Epitopes, T-Lymphocyte/biosynthesis
- Epitopes, T-Lymphocyte/genetics
- Epitopes, T-Lymphocyte/immunology
- Female
- Genes, Recessive/immunology
- Immunization, Secondary/methods
- Lymphocyte Activation/genetics
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Neoplasm Staging
- Polyomavirus Infections/immunology
- Polyomavirus Infections/mortality
- Polyomavirus Infections/pathology
- Polyomavirus Infections/prevention & control
- Protein Structure, Tertiary/genetics
- Receptors, Antigen, T-Cell/genetics
- Survival Analysis
- Tumor Virus Infections/immunology
- Tumor Virus Infections/mortality
- Tumor Virus Infections/pathology
- Tumor Virus Infections/prevention & control
Collapse
Affiliation(s)
- Christina M Ryan
- Department of Microbiology and Immunology, Pennsylvania State University College of Medicine, 500 University Drive, Hershey, PA 17033, USA
| | | |
Collapse
|
19
|
Garbi N, Arnold B, Gordon S, Hämmerling GJ, Ganss R. CpG motifs as proinflammatory factors render autochthonous tumors permissive for infiltration and destruction. THE JOURNAL OF IMMUNOLOGY 2004; 172:5861-9. [PMID: 15128765 DOI: 10.4049/jimmunol.172.10.5861] [Citation(s) in RCA: 111] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
In a transgenic mouse model expressing SV40 T Ag (Tag) as a de novo tumor Ag, immune surveillance fails and islet cell carcinomas grow progressively. To develop an anticancer strategy that would be effective in eradicating solid, autochthonously growing tumors, we evaluated the effectiveness of immunostimulatory oligodeoxynucleotides (ODN) with cytosine-guanine-rich (CpG) motifs (CpG-ODN). In a classical vaccination protocol, Tag was administered with CpG-ODN as adjuvant. The antitumor vaccination, however, was only effective in a prophylactic setting, despite the successful activation of a Tag-specific CTL response in vivo. Histological examination demonstrated that even primed immune cells failed to infiltrate tumors once a malignant environment was established. To ensure that effector cells were not limiting, highly activated tumor Ag-specific T cells were transferred into tumor-bearing mice. However, this treatment also failed to result in tumor infiltration and rejection. Therefore, we further tested the efficacy of CpG-ODN as a proinflammatory agent in combination with the transfer of preactivated Tag-specific CD4(+) and CD8(+) T cells. Indeed, this combination therapy proved to be highly effective, because CpG-ODN rendered insulinomas permissive for massive infiltration and destruction. The opening of tumor tissue correlated with uptake of CpG-ODN by tissue-resident macrophages and a strong up-regulation of adhesion molecules such as ICAM and VCAM on blood vessel endothelia. These data demonstrate that systemic application of proinflammatory reagents drastically enhances extravasation of effector cells into tumor tissue, an observation that is of general importance for immunotherapy of solid tumors in a clinical setting.
Collapse
MESH Headings
- Adjuvants, Immunologic/administration & dosage
- Animals
- Antigens, Polyomavirus Transforming/administration & dosage
- Antigens, Polyomavirus Transforming/immunology
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Cell Movement/genetics
- Cell Movement/immunology
- CpG Islands/immunology
- Cytotoxicity, Immunologic/genetics
- Epitopes, T-Lymphocyte/immunology
- Immunity, Innate/genetics
- Immunotherapy, Adoptive
- Inflammation Mediators/administration & dosage
- Inflammation Mediators/immunology
- Islets of Langerhans/immunology
- Islets of Langerhans/metabolism
- Islets of Langerhans/pathology
- Lymphocyte Activation/genetics
- Mice
- Mice, Inbred C3H
- Mice, Inbred C57BL
- Mice, Transgenic
- Oligodeoxyribonucleotides/administration & dosage
- Oligodeoxyribonucleotides/immunology
- Pancreatic Neoplasms/genetics
- Pancreatic Neoplasms/immunology
- Pancreatic Neoplasms/pathology
- Pancreatic Neoplasms/therapy
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Cytotoxic/transplantation
Collapse
Affiliation(s)
- Natalio Garbi
- Department of Molecular Immunology, German Cancer Research Center, Heidelberg, Germany
| | | | | | | | | |
Collapse
|
20
|
Gawlick U, Kranz DM, Schepkin VD, Roy EJ. A Conjugate of a Tumor-Targeting Ligand and a T Cell Costimulatory Antibody To Treat Brain Tumors. Bioconjug Chem 2004; 15:1137-45. [PMID: 15366970 DOI: 10.1021/bc049911e] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
T cell immunotherapy is a potential strategy for the treatment of brain tumors because it offers a high degree of specificity, the ability to extravasate into solid tumors, and the potential for eliciting a long-term protective immune response. Various approaches have been developed to overcome T cell immune tolerance to cancer, including the use of cytokines and bispecific antibodies. T cell stimulation with the proinflammatory cytokine IL-12 can elicit antitumor immunity. T cell activation can be increased using bispecific antibodies against activating molecules on the surface of T cells and a tumor antigen. We studied the effects of systemic IL-12 administration in combination with a conjugate of an anti-CD28 antibody and a ligand for the folate receptor. The high affinity folate receptor is expressed on endogenously arising choroid plexus tumors of SV11 mice, which are transgenic for large T antigen under the control of the SV40 promoter. SV11 mice are immunocompetent, yet immunologically tolerant to large T antigen expressed by choroid plexus tumors. MRI analysis showed that the administration of IL-12 and anti-CD28 Fab/folate significantly slowed tumor growth. Proliferating CD8(+) T cells were found in choroid plexus tumors of treated animals. Treatment of animals with IL-12 + anti-CD28 Fab/folate prolonged survival compared to IL-12 alone. Cytokine treatment combined with tumor-targeted costimulation may be a useful adjunct treatment.
Collapse
Affiliation(s)
- Ute Gawlick
- Neuroscience Program and Department of Biochemistry, University of Illinois, Urbana, Illinois 61801, USA
| | | | | | | |
Collapse
|
21
|
Schell TD. In vivo expansion of the residual tumor antigen-specific CD8+ T lymphocytes that survive negative selection in simian virus 40 T-antigen-transgenic mice. J Virol 2004; 78:1751-62. [PMID: 14747540 PMCID: PMC369430 DOI: 10.1128/jvi.78.4.1751-1762.2004] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Mice that express the viral oncoprotein simian virus 40 (SV40) large T antigen (T-Ag) as a transgene provide useful models for the assessment of the state of the host immune response in the face of spontaneous tumor progression. Line SV11 (H2(b)) mice develop rapidly progressing choroid plexus tumors due to expression of full-length T-Ag from the SV40 promoter. In addition, T-Ag expression in the thymus of SV11 mice results in the deletion of CD8(+) T cells specific for the three H2(b)-restricted immunodominant epitopes of T-Ag. Whether CD8(+) T cells specific for the immunorecessive H2-D(b)-restricted epitope V of T-Ag survive negative selection in SV11 mice has not been determined. Immunization of SV11 mice with rVV-ES-V, a recombinant vaccinia virus expressing epitope V as a minigene, resulted in the induction of weak, but reproducible, epitope V-specific cytotoxic T-lymphocyte (CTL) responses. This weak lytic response corresponded with a decreased frequency of epitope V-specific CTL that could be recruited in SV11 mice. In addition, CTL lines derived from rVV-ES-V-immunized SV11 mice had reduced avidities compared to that seen with CTL derived from healthy mice. Despite this initial weak response, significant numbers of epitope V-specific CD8(+) T cells were detected in SV11 mice ex vivo following a priming-boosting approach and these cells demonstrated high avidity for epitope V. The results suggest that low numbers of tumor-reactive CD8(+) T cells with high avidity for epitope V survive negative selection in SV11 mice but can be expanded by specific boosting approaches in the tumor bearing host.
Collapse
Affiliation(s)
- Todd D Schell
- Department of Microbiology and Immunology, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033, USA.
| |
Collapse
|
22
|
Kjaergaard J, Peng L, Cohen PA, Shu S. Therapeutic efficacy of adoptive immunotherapy is predicated on in vivo antigen-specific proliferation of donor T cells. Clin Immunol 2003; 108:8-20. [PMID: 12865066 DOI: 10.1016/s1521-6616(03)00090-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Activated T cells with down-regulated L-selectin expression (L-sel(-)) from tumor-draining lymph nodes represent a potent source of specific immune effectors in adoptive immunotherapy. Using congenic pairs of mice and carboxyfluorescein diacetate succinimidyl ester-labeled L-sel(-) T cells, the current study analyzed in vivo proliferation of transferred cells. In the lung of MCA205 tumor-bearing mice, 6% or 0.3 x 10(6) of the 5 x 10(6) donor cells were identified 24 h after transfer. Vigorous proliferation of donor cells was evident on day 2, reaching a maximum on day 6. The proliferation was tumor-specific and CD4 T cells divided with greater magnitude than CD8 cells. Successful adoptive immunotherapy also required sublethal whole-body irradiation (WBI) of the recipient. WBI exerted its effects on facilitating specific T cell proliferation at the tumor site. Taken together, our results demonstrate that adoptively transferred T cells undergo extensive proliferation in response to the tumor and this response is associated with therapeutic efficacy.
Collapse
Affiliation(s)
- Jørgen Kjaergaard
- Center for Surgery Research, Cleveland Clinic Foundation, Cleveland, OH 44195, USA
| | | | | | | |
Collapse
|
23
|
Affiliation(s)
- Robert L Garcea
- Section of Pediatric Oncology, University of Colorado School of Medicine, Denver 80262, USA
| | | |
Collapse
|
24
|
Zheng X, Gao JX, Zhang H, Geiger TL, Liu Y, Zheng P. Clonal deletion of simian virus 40 large T antigen-specific T cells in the transgenic adenocarcinoma of mouse prostate mice: an important role for clonal deletion in shaping the repertoire of T cells specific for antigens overexpressed in solid tumors. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2002; 169:4761-9. [PMID: 12391185 DOI: 10.4049/jimmunol.169.9.4761] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
In addition to their overexpression in cancer cells, most of the tumor-associated Ags are expressed at low but detectable levels in normal tissues. It is not clear whether the repertoire of T cells specific for unmutated tumor Ags is shaped by negative selection during T cell development. The transgenic adenocarcinoma of mouse prostate (TRAMP) model is transgenic for the SV40 large T Ag (Tag) under the control of the rat probasin regulatory elements. Although it has been established that T lymphocytes from TRAMP mice are tolerant to SV40 Tag, the mechanism of the tolerance is largely unknown. To examine whether the T cell clonal deletion is responsible for the tolerance, we crossed the TRAMP mice with mice transgenic for a rearranged TCR specific for SV40 Tag presented by the H-2K(k). Double transgenic TRAMP/TCR mice showed profound thymic deletion of SV40 Tag-reactive T cells, including a 6- to 10-fold reduction in the total thymocyte numbers and a >50-fold reduction in phenotypically mature T cells. Consistent with this finding, we observed that the SV40 Tag and endogenous mouse probasin genes are expressed at low levels in the thymus. These results demonstrate that clonal deletion is a major mechanism for tolerance to Ags previously regarded as prostate-specific, and provide direct evidence that the T cell repertoire specific for an unmutated tumor Ag can be shaped by clonal deletion in the thymus.
Collapse
MESH Headings
- Adenocarcinoma/genetics
- Adenocarcinoma/immunology
- Amino Acid Sequence
- Androgen-Binding Protein/biosynthesis
- Animals
- Antigens, Neoplasm/biosynthesis
- Antigens, Neoplasm/genetics
- Antigens, Polyomavirus Transforming/biosynthesis
- Antigens, Polyomavirus Transforming/genetics
- Antigens, Polyomavirus Transforming/immunology
- Cell Line
- Clonal Deletion/genetics
- Epitopes, T-Lymphocyte/biosynthesis
- Epitopes, T-Lymphocyte/genetics
- Epitopes, T-Lymphocyte/immunology
- Female
- Immunodominant Epitopes/immunology
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic/genetics
- Mice, Transgenic/immunology
- Molecular Sequence Data
- Prostatic Neoplasms/genetics
- Prostatic Neoplasms/immunology
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/metabolism
- T-Lymphocyte Subsets/virology
- Thymus Gland/cytology
- Thymus Gland/immunology
- Thymus Gland/metabolism
- Thymus Gland/virology
Collapse
Affiliation(s)
- Xincheng Zheng
- Department of Pathology and Comprehensive Cancer Center, Ohio State University, Columbus 43210, USA
| | | | | | | | | | | |
Collapse
|
25
|
Velupillai P, Carroll JP, Benjamin TL. Susceptibility to polyomavirus-induced tumors in inbred mice: role of innate immune responses. J Virol 2002; 76:9657-63. [PMID: 12208944 PMCID: PMC136524 DOI: 10.1128/jvi.76.19.9657-9663.2002] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Mice of the PERA/Ei strain (PE mice) are highly susceptible to tumor induction by polyomavirus and transmit their susceptibility in a dominant manner in crosses with resistant C57BR/cdJ mice (BR mice). BR mice respond to polyomavirus infection with a type 1 cytokine response and develop effective cell-mediated immunity to the virus-induced tumors. By enumerating virus-specific CD8(+) T cells and measuring cytokine responses, we show that the susceptibility of PE mice is due to the absence of a type 1 cytokine response and a concomitant failure to sustain virus-specific cytotoxic T lymphocytes. (PE x BR)F(1) mice showed an initial type 1 response that became skewed toward type 2. Culture supernatants of splenocytes from infected PE mice stimulated in vitro contained high levels of interleukin-10 and no detectable gamma interferon, while those from BR mice showed the opposite pattern. Differences in the innate immune response to polyomavirus by antigen-presenting cells in PE mice and BR mice led to polarization of T-cell cytokine responses. Adherent cells from spleens of infected BR mice produced high levels of interleukin-12, while those from infected PE and F(1) mice produced predominantly interleukin-10. PE and F(1) mice infected by polyomavirus responded with increases in antigen-presenting cells expressing B7.2 costimulatory molecules, whereas BR mice responded with increased expression of B7.1. Administration of recombinant interleukin-12 along with virus resulted in partial protection of PE mice and provided complete protection against tumor development in F(1) animals.
Collapse
Affiliation(s)
- Palanivel Velupillai
- Department of Pathology, Harvard Medical School, Boston, Massachusetts 02115, USA
| | | | | |
Collapse
|
26
|
Mashishi T, Gray CM. The ELISPOT assay: an easily transferable method for measuring cellular responses and identifying T cell epitopes. Clin Chem Lab Med 2002; 40:903-10. [PMID: 12435107 DOI: 10.1515/cclm.2002.159] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Characterization of human leukocyte antigen (HLA) class I restricted epitopes derived from viral pathogens is imperative for formulating therapeutic interventions, as well as for vaccine design and monitoring. Sensitive, easy and cost-effective assays that measure the frequency of antigen-specific T lymphocytes are crucial for evaluating and improving vaccines and therapies. This paper reviews the ELISPOT technique that allows for quantifying HIV-specific T lymphocytes at the single cell level from peripheral blood by detection of antigen-induced cytokine secretion. The assay can be used successfully to quantify T cell immune responses in humans infected with different pathogens and to assess T cell immunogenicity of vaccines in phase I/II and III clinical trials. This review focuses on the ELISPOT methodology and discusses how it can be standardized and potentially used by multiple international laboratories attached to clinical trial sites.
Collapse
Affiliation(s)
- Tumelo Mashishi
- AIDS Research Unit, National Institute for Communicable Diseases, Johannesburg, South Africa
| | | |
Collapse
|