1
|
Fines C, McCarthy H, Buckley N. The search for a TNBC vaccine: the guardian vaccine. Cancer Biol Ther 2025; 26:2472432. [PMID: 40089851 PMCID: PMC11913391 DOI: 10.1080/15384047.2025.2472432] [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: 11/22/2024] [Revised: 02/18/2025] [Accepted: 02/19/2025] [Indexed: 03/17/2025] Open
Abstract
Nearly 20 million people are diagnosed with cancer each year with breast cancer being the most common among women. Triple negative breast cancer (TNBC), defined by its no/low expression of ER and PR and lack of amplification of HER2, makes up 15-20% of all breast cancer cases. While patients overall have a higher response to chemotherapy, this subgroup is associated with the lowest survival rate indicating significant clinical and molecular heterogeneity demanding alternate treatment options. Therefore, new therapies have been explored, with a large focus on utilizing the immune system. A whole host of immunotherapies have been studied including immune checkpoint inhibitors, now standard of care for eligible patients, and possibly the most exciting and promising is that of a TNBC vaccine. While currently there are no approved TNBC vaccines, this review highlights many promising studies and points to an antigen, p53, which we believe is highly relevant for TNBC.
Collapse
Affiliation(s)
- Cory Fines
- School of Pharmacy, Queen's University Belfast, Belfast, UK
| | - Helen McCarthy
- School of Pharmacy, Queen's University Belfast, Belfast, UK
| | - Niamh Buckley
- School of Pharmacy, Queen's University Belfast, Belfast, UK
| |
Collapse
|
2
|
Song B, Yang P, Zhang S. Cell fate regulation governed by p53: Friends or reversible foes in cancer therapy. Cancer Commun (Lond) 2024; 44:297-360. [PMID: 38311377 PMCID: PMC10958678 DOI: 10.1002/cac2.12520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 01/03/2024] [Accepted: 01/11/2024] [Indexed: 02/10/2024] Open
Abstract
Cancer is a leading cause of death worldwide. Targeted therapies aimed at key oncogenic driver mutations in combination with chemotherapy and radiotherapy as well as immunotherapy have benefited cancer patients considerably. Tumor protein p53 (TP53), a crucial tumor suppressor gene encoding p53, regulates numerous downstream genes and cellular phenotypes in response to various stressors. The affected genes are involved in diverse processes, including cell cycle arrest, DNA repair, cellular senescence, metabolic homeostasis, apoptosis, and autophagy. However, accumulating recent studies have continued to reveal novel and unexpected functions of p53 in governing the fate of tumors, for example, functions in ferroptosis, immunity, the tumor microenvironment and microbiome metabolism. Among the possibilities, the evolutionary plasticity of p53 is the most controversial, partially due to the dizzying array of biological functions that have been attributed to different regulatory mechanisms of p53 signaling. Nearly 40 years after its discovery, this key tumor suppressor remains somewhat enigmatic. The intricate and diverse functions of p53 in regulating cell fate during cancer treatment are only the tip of the iceberg with respect to its equally complicated structural biology, which has been painstakingly revealed. Additionally, TP53 mutation is one of the most significant genetic alterations in cancer, contributing to rapid cancer cell growth and tumor progression. Here, we summarized recent advances that implicate altered p53 in modulating the response to various cancer therapies, including chemotherapy, radiotherapy, and immunotherapy. Furthermore, we also discussed potential strategies for targeting p53 as a therapeutic option for cancer.
Collapse
Affiliation(s)
- Bin Song
- Laboratory of Radiation MedicineWest China Second University HospitalSichuan UniversityChengduSichuanP. R. China
| | - Ping Yang
- Laboratory of Radiation MedicineWest China Second University HospitalSichuan UniversityChengduSichuanP. R. China
| | - Shuyu Zhang
- Laboratory of Radiation MedicineWest China Second University HospitalSichuan UniversityChengduSichuanP. R. China
- The Second Affiliated Hospital of Chengdu Medical CollegeChina National Nuclear Corporation 416 HospitalChengduSichuanP. R. China
- Laboratory of Radiation MedicineNHC Key Laboratory of Nuclear Technology Medical TransformationWest China School of Basic Medical Sciences & Forensic MedicineSichuan UniversityChengduSichuanP. R. China
| |
Collapse
|
3
|
Abstract
Mutations in the TP53 tumour suppressor gene are very frequent in cancer, and attempts to restore the functionality of p53 in tumours as a therapeutic strategy began decades ago. However, very few of these drug development programmes have reached late-stage clinical trials, and no p53-based therapeutics have been approved in the USA or Europe so far. This is probably because, as a nuclear transcription factor, p53 does not possess typical drug target features and has therefore long been considered undruggable. Nevertheless, several promising approaches towards p53-based therapy have emerged in recent years, including improved versions of earlier strategies and novel approaches to make undruggable targets druggable. Small molecules that can either protect p53 from its negative regulators or restore the functionality of mutant p53 proteins are gaining interest, and drugs tailored to specific types of p53 mutants are emerging. In parallel, there is renewed interest in gene therapy strategies and p53-based immunotherapy approaches. However, major concerns still remain to be addressed. This Review re-evaluates the efforts made towards targeting p53-dysfunctional cancers, and discusses the challenges encountered during clinical development.
Collapse
Affiliation(s)
- Ori Hassin
- grid.13992.300000 0004 0604 7563Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Moshe Oren
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
4
|
Abstract
The importance of cancer-cell-autonomous functions of the tumour suppressor p53 (encoded by TP53) has been established in many studies, but it is now clear that the p53 status of the cancer cell also has a profound impact on the immune response. Loss or mutation of p53 in cancers can affect the recruitment and activity of myeloid and T cells, allowing immune evasion and promoting cancer progression. p53 can also function in immune cells, resulting in various outcomes that can impede or support tumour development. Understanding the role of p53 in tumour and immune cells will help in the development of therapeutic approaches that can harness the differential p53 status of cancers compared with most normal tissue.
Collapse
Affiliation(s)
- Julianna Blagih
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Michael D Buck
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Karen H Vousden
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| |
Collapse
|
5
|
Targeting mutant p53-expressing tumours with a T cell receptor-like antibody specific for a wild-type antigen. Nat Commun 2019; 10:5382. [PMID: 31772160 PMCID: PMC6879612 DOI: 10.1038/s41467-019-13305-z] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Accepted: 11/02/2019] [Indexed: 01/09/2023] Open
Abstract
Accumulation of mutant p53 proteins is frequently found in a wide range of cancers. While conventional antibodies fail to target intracellular proteins, proteosomal degradation results in the presentation of p53-derived peptides on the tumour cell surface by class I molecules of the major histocompatibility complex (MHC). Elevated levels of such p53-derived peptide-MHCs on tumour cells potentially differentiate them from healthy tissues. Here, we report the engineering of an affinity-matured human antibody, P1C1TM, specific for the unmutated p53125-134 peptide in complex with the HLA-A24 class I MHC molecule. We show that P1C1TM distinguishes between mutant and wild-type p53 expressing HLA-A24+ cells, and mediates antibody dependent cellular cytotoxicity of mutant p53 expressing cells in vitro. Furthermore, we show that cytotoxic PNU-159682-P1C1TM drug conjugates specifically inhibit growth of mutant p53 expressing cells in vitro and in vivo. Hence, p53-associated peptide-MHCs are attractive targets for the immunotherapy against mutant p53 expressing tumours. Several cancers harbour mutant p53 and express higher levels of p53-derived peptide-MHCs. Here, the authors report affinity matured human antibody, P1C1TM, specific for the p53125-134 peptide in complex with the HLA-A24 class I MHC molecule and show its efficacy and specificity for mutant p53 expressing tumours.
Collapse
|
6
|
TP53 missense mutation is associated with increased tumor-infiltrating T cells in primary prostate cancer. Hum Pathol 2019; 87:95-102. [PMID: 30851334 DOI: 10.1016/j.humpath.2019.02.006] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 02/20/2019] [Accepted: 02/24/2019] [Indexed: 01/06/2023]
Abstract
The makeup of the tumor immune microenvironment may be associated with tumor somatic genomic alterations and plays a key role in tumor progression and response to immunotherapy. We examined the association of tumor-infiltrating T-cell density with TP53 status in surgically treated primary prostate cancer using 3 independent tissue microarray sets, including one set of tumors from grade-matched patients of European American or African American ancestry (n = 391), a retrospective case-cohort of intermediate- and high-risk patients enriched for adverse outcomes (n = 267), and a set of tumors with primary Gleason pattern 5 (n = 77). The presence of TP53 missense mutation, indicated by p53 nuclear accumulation using a genetically validated assay, was significantly associated with increased CD3+ T-cell density (median, 341 versus 231 CD3+ T cells/mm2; P = .004) in the matched European American and African American ancestry patient sets. The same association was present in patients of both ancestries when analyzed separately, despite the fact that p53 nuclear accumulation was less frequent among African American compared with European American tumors (7% versus 3%, P = .2). The validation cohorts of intermediate/high-risk and primary Gleason pattern 5 patients corroborated the association of increased CD3+ T-cell density with presence of p53 nuclear accumulation. In a pooled analysis of all sets, adjusting for clinicopathological variables, CD3+ and CD8+, but not FOXP3+, T-cell densities remained significantly higher in tumors with p53 nuclear accumulation compared with those without. TP53 mutation is associated with higher tumor-infiltrating T-cell density, which may be relevant in future clinical trials of immunotherapy in prostate cancer.
Collapse
|
7
|
Chiappori AA, Williams CC, Gray JE, Tanvetyanon T, Haura EB, Creelan BC, Thapa R, Chen DT, Simon GR, Bepler G, Gabrilovich DI, Antonia SJ. Randomized-controlled phase II trial of salvage chemotherapy after immunization with a TP53-transfected dendritic cell-based vaccine (Ad.p53-DC) in patients with recurrent small cell lung cancer. Cancer Immunol Immunother 2019; 68:517-527. [PMID: 30591959 PMCID: PMC6426813 DOI: 10.1007/s00262-018-2287-9] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 12/11/2018] [Indexed: 12/11/2022]
Abstract
Small cell lung cancer TP53 mutations lead to expression of tumor antigens that elicits specific cytotoxic T-cell immune responses. In this phase II study, dendritic cells transfected with wild-type TP53 (vaccine) were administered to patients with extensive-stage small cell lung cancer after chemotherapy. Patients were randomized 1:1:1 to arm A (observation), arm B (vaccine alone), or arm C (vaccine plus all-trans-retinoic acid). Vaccine was administered every 2 weeks (3 times), and all patients were to receive paclitaxel at progression. Our primary endpoint was overall response rate (ORR) to paclitaxel. The study was not designed to detect overall response rate differences between arms. Of 69 patients enrolled (performance status 0/1, median age 62 years), 55 were treated in stage 1 (18 in arm A, 20 in arm B, and 17 in arm C) and 14 in stage 2 (arm C only), per 2-stage Simon Minimax design. The vaccine was safe, with mostly grade 1/2 toxicities, although 1 arm-B patient experienced grade 3 fatigue and 8 arm-C patients experienced grade 3 toxicities. Positive immune responses were obtained in 20% of arm B (95% confidence interval [CI], 5.3-48.6) and 43.3% of arm C (95% CI 23.9-65.1). The ORRs to the second-line chemotherapy (including paclitaxel) were 15.4% (95% CI 2.7-46.3), 16.7% (95% CI 2.9-49.1), and 23.8% (95% CI 9.1-47.5) for arms A, B, and C, with no survival differences between arms. Although our vaccine failed to improve ORRs to the second-line chemotherapy, its safety profile and therapeutic immune potential remain. Combinations with the other immunotherapeutic agents are reasonable options.
Collapse
Affiliation(s)
- Alberto A Chiappori
- Department of Thoracic Oncology, H. Lee Moffitt Cancer Center and Research Institute, 12902, Magnolia Drive, FOB1, Tampa, FL, 33612, USA.
| | - Charles C Williams
- Department of Thoracic Oncology, H. Lee Moffitt Cancer Center and Research Institute, 12902, Magnolia Drive, FOB1, Tampa, FL, 33612, USA
| | - Jhanelle E Gray
- Department of Thoracic Oncology, H. Lee Moffitt Cancer Center and Research Institute, 12902, Magnolia Drive, FOB1, Tampa, FL, 33612, USA
| | - Tawee Tanvetyanon
- Department of Thoracic Oncology, H. Lee Moffitt Cancer Center and Research Institute, 12902, Magnolia Drive, FOB1, Tampa, FL, 33612, USA
| | - Eric B Haura
- Department of Thoracic Oncology, H. Lee Moffitt Cancer Center and Research Institute, 12902, Magnolia Drive, FOB1, Tampa, FL, 33612, USA
| | - Ben C Creelan
- Department of Thoracic Oncology, H. Lee Moffitt Cancer Center and Research Institute, 12902, Magnolia Drive, FOB1, Tampa, FL, 33612, USA
| | - Ram Thapa
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA
| | - Dung-Tsa Chen
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA
| | | | | | | | - Scott J Antonia
- Department of Thoracic Oncology, H. Lee Moffitt Cancer Center and Research Institute, 12902, Magnolia Drive, FOB1, Tampa, FL, 33612, USA
| |
Collapse
|
8
|
Calles A, Aguado G, Sandoval C, Álvarez R. The role of immunotherapy in small cell lung cancer. Clin Transl Oncol 2019; 21:961-976. [DOI: 10.1007/s12094-018-02011-9] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 12/07/2018] [Indexed: 12/31/2022]
|
9
|
A general strategy to optimize immunogenicity of HLA-B*0702 restricted cryptic peptides from tumor associated antigens: Design of universal neo-antigen like tumor vaccines for HLA-B*0702 positive patients. Oncotarget 2018; 7:59417-59428. [PMID: 27506946 PMCID: PMC5312321 DOI: 10.18632/oncotarget.11086] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Accepted: 07/13/2016] [Indexed: 11/27/2022] Open
Abstract
Tumor Associated Antigens (TAAs) are the privileged targets of almost all the cancer vaccines tested to date. Unfortunately all these vaccines failed to show a clinical efficacy. The main reason for this failure is the immune tolerance to TAAs that are self-proteins expressed by normal and cancer cells. Self-tolerance to TAAs is directed against their dominant rather than against their cryptic epitopes. The best way to overcome self-tolerance to TAAs would therefore be to target their cryptic epitopes. However, because of their low HLA-I affinity, cryptic peptides are non-immunogenic and cannot be used to stimulate an antitumor immune response unless their immunogenicity has been previously enhanced. In this paper we describe a general approach to enhance immunogenicity of almost all the HLA-B*0702 restricted cryptic peptides derived from TAAs. It consists in substituting residues at position 1 or 9 of low HLA-B*0702 affinity cryptic peptides by an Alanine or a Leucine respectively. These substitutions increase affinity of peptides for HLA-B*0702. These optimized cryptic peptides are strongly immunogenic and very importantly CTL they stimulate recognize their native counterparts. TAAs derived optimized cryptic peptides can be considered as universal antitumor vaccine since they escape self-tolerance, are immunogenic and are not patient specific.
Collapse
|
10
|
Li D, Bentley C, Anderson A, Wiblin S, Cleary KLS, Koustoulidou S, Hassanali T, Yates J, Greig J, Nordkamp MO, Trenevska I, Ternette N, Kessler BM, Cornelissen B, Cragg MS, Banham AH. Development of a T-cell Receptor Mimic Antibody against Wild-Type p53 for Cancer Immunotherapy. Cancer Res 2017; 77:2699-2711. [PMID: 28363997 DOI: 10.1158/0008-5472.can-16-3247] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 01/17/2017] [Accepted: 03/09/2017] [Indexed: 11/16/2022]
Abstract
The tumor suppressor p53 is widely dysregulated in cancer and represents an attractive target for immunotherapy. Because of its intracellular localization, p53 is inaccessible to classical therapeutic monoclonal antibodies, an increasingly successful class of anticancer drugs. However, peptides derived from intracellular antigens are presented on the cell surface in the context of MHC I and can be bound by T-cell receptors (TCR). Here, we report the development of a novel antibody, T1-116C, that acts as a TCR mimic to recognize an HLA-A*0201-presented wild-type p53 T-cell epitope, p5365-73(RMPEAAPPV). The antibody recognizes a wide range of cancers, does not bind normal peripheral blood mononuclear cells, and can activate immune effector functions to kill cancer cells in vitroIn vivo, the antibody targets p5365-73 peptide-expressing breast cancer xenografts, significantly inhibiting tumor growth. This represents a promising new agent for future cancer immunotherapy. Cancer Res; 77(10); 2699-711. ©2017 AACR.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/immunology
- Antibodies, Monoclonal/pharmacology
- Antibody-Dependent Cell Cytotoxicity/immunology
- Cell Line, Tumor
- Disease Models, Animal
- Epitopes, T-Lymphocyte/immunology
- Female
- HLA-A2 Antigen/chemistry
- HLA-A2 Antigen/immunology
- HLA-A2 Antigen/metabolism
- Humans
- Immunophenotyping
- Immunotherapy
- Mice
- Molecular Mimicry
- Neoplasms/drug therapy
- Neoplasms/genetics
- Neoplasms/immunology
- Neoplasms/metabolism
- Protein Binding
- Protein Multimerization
- Receptors, Antigen, T-Cell/antagonists & inhibitors
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/metabolism
- T-Lymphocytes, Cytotoxic/immunology
- Tumor Burden/drug effects
- Tumor Suppressor Protein p53/chemistry
- Tumor Suppressor Protein p53/genetics
- Tumor Suppressor Protein p53/metabolism
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Demin Li
- Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Headington, Oxford, United Kingdom.
| | - Carol Bentley
- Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Headington, Oxford, United Kingdom
| | - Amanda Anderson
- Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Headington, Oxford, United Kingdom
| | - Sarah Wiblin
- Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Headington, Oxford, United Kingdom
| | - Kirstie L S Cleary
- Antibody & Vaccine Group, Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, United Kingdom
| | - Sofia Koustoulidou
- CRUK/MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Tasneem Hassanali
- Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Headington, Oxford, United Kingdom
| | - Jenna Yates
- Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Headington, Oxford, United Kingdom
| | - Jenny Greig
- Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Headington, Oxford, United Kingdom
| | - Marloes Olde Nordkamp
- Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Headington, Oxford, United Kingdom
| | - Iva Trenevska
- Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Headington, Oxford, United Kingdom
| | - Nicola Ternette
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Benedikt M Kessler
- Target Discovery Institute, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Bart Cornelissen
- CRUK/MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Mark S Cragg
- Antibody & Vaccine Group, Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, United Kingdom
| | - Alison H Banham
- Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Headington, Oxford, United Kingdom.
| |
Collapse
|
11
|
Li D, Bentley C, Yates J, Salimi M, Greig J, Wiblin S, Hassanali T, Banham AH. Engineering chimeric human and mouse major histocompatibility complex (MHC) class I tetramers for the production of T-cell receptor (TCR) mimic antibodies. PLoS One 2017; 12:e0176642. [PMID: 28448627 PMCID: PMC5407768 DOI: 10.1371/journal.pone.0176642] [Citation(s) in RCA: 7] [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: 12/07/2016] [Accepted: 04/13/2017] [Indexed: 01/30/2023] Open
Abstract
Therapeutic monoclonal antibodies targeting cell surface or secreted antigens are among the most effective classes of novel immunotherapies. However, the majority of human proteins and established cancer biomarkers are intracellular. Peptides derived from these intracellular proteins are presented on the cell surface by major histocompatibility complex class I (MHC-I) and can be targeted by a novel class of T-cell receptor mimic (TCRm) antibodies that recognise similar epitopes to T-cell receptors. Humoural immune responses to MHC-I tetramers rarely generate TCRm antibodies and many antibodies recognise the α3 domain of MHC-I and β2 microglobulin (β2m) that are not directly involved in presenting the target peptide. Here we describe the production of functional chimeric human-murine HLA-A2-H2Dd tetramers and modifications that increase their bacterial expression and refolding efficiency. These chimeric tetramers were successfully used to generate TCRm antibodies against two epitopes derived from wild type tumour suppressor p53 (RMPEAAPPV and GLAPPQHLIRV) that have been used in vaccination studies. Immunisation with chimeric tetramers yielded no antibodies recognising the human α3 domain and β2m and generated TCRm antibodies capable of specifically recognising the target peptide/MHC-I complex in fully human tetramers and on the cell surface of peptide pulsed T2 cells. Chimeric tetramers represent novel immunogens for TCRm antibody production and may also improve the yield of tetramers for groups using these reagents to monitor CD8 T-cell immune responses in HLA-A2 transgenic mouse models of immunotherapy.
Collapse
Affiliation(s)
- Demin Li
- Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, Level 4, Academic Block, John Radcliffe Hospital, Headington, Oxford, United Kingdom
- * E-mail: (AHB); (DL)
| | - Carol Bentley
- Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, Level 4, Academic Block, John Radcliffe Hospital, Headington, Oxford, United Kingdom
| | - Jenna Yates
- Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, Level 4, Academic Block, John Radcliffe Hospital, Headington, Oxford, United Kingdom
| | - Maryam Salimi
- Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, Level 4, Academic Block, John Radcliffe Hospital, Headington, Oxford, United Kingdom
| | - Jenny Greig
- Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, Level 4, Academic Block, John Radcliffe Hospital, Headington, Oxford, United Kingdom
| | - Sarah Wiblin
- Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, Level 4, Academic Block, John Radcliffe Hospital, Headington, Oxford, United Kingdom
| | - Tasneem Hassanali
- Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, Level 4, Academic Block, John Radcliffe Hospital, Headington, Oxford, United Kingdom
| | - Alison H. Banham
- Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, Level 4, Academic Block, John Radcliffe Hospital, Headington, Oxford, United Kingdom
- * E-mail: (AHB); (DL)
| |
Collapse
|
12
|
Abstract
Oncolytic virus (OV) therapy utilizes replication-competent viruses to kill cancer cells, leaving non-malignant cells unharmed. With the first U.S. Food and Drug Administration-approved OV, dozens of clinical trials ongoing, and an abundance of translational research in the field, OV therapy is poised to be one of the leading treatments for cancer. A number of recombinant OVs expressing a transgene for p53 (TP53) or another p53 family member (TP63 or TP73) were engineered with the goal of generating more potent OVs that function synergistically with host immunity and/or other therapies to reduce or eliminate tumor burden. Such transgenes have proven effective at improving OV therapies, and basic research has shown mechanisms of p53-mediated enhancement of OV therapy, provided optimized p53 transgenes, explored drug-OV combinational treatments, and challenged canonical roles for p53 in virus-host interactions and tumor suppression. This review summarizes studies combining p53 gene therapy with replication-competent OV therapy, reviews preclinical and clinical studies with replication-deficient gene therapy vectors expressing p53 transgene, examines how wild-type p53 and p53 modifications affect OV replication and anti-tumor effects of OV therapy, and explores future directions for rational design of OV therapy combined with p53 gene therapy.
Collapse
|
13
|
Menez-Jamet J, Gallou C, Rougeot A, Kosmatopoulos K. Optimized tumor cryptic peptides: the basis for universal neo-antigen-like tumor vaccines. ANNALS OF TRANSLATIONAL MEDICINE 2016; 4:266. [PMID: 27563653 DOI: 10.21037/atm.2016.05.15] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
The very impressive clinical results recently obtained in cancer patients treated with immune response checkpoint inhibitors boosted the interest in immunotherapy as a therapeutic choice in cancer treatment. However, these inhibitors require a pre-existing tumor specific immune response and the presence of tumor infiltrating T cells to be efficient. This immune response can be triggered by cancer vaccines. One of the main issues in tumor vaccination is the choice of the right antigen to target. All vaccines tested to date targeted tumor associated antigens (TAA) that are self-antigens and failed to show a clinical efficacy because of the immune self-tolerance to TAA. A new class of tumor antigens has recently been described, the neo-antigens that are created by point mutations of tumor expressing proteins and are recognized by the immune system as non-self. Neo-antigens exhibit two main properties: they are not involved in the immune self-tolerance process and are immunogenic. However, the majority of the neo-antigens are patient specific and their use as cancer vaccines requires their previous identification in each patient individualy that can be done only in highly specialized research centers. It is therefore evident that neo-antigens cannot be used for patient vaccination worldwide. This raises the question of whether we can find neo-antigen like vaccines, which would not be patient specific. In this review we show that optimized cryptic peptides from TAA are neo-antigen like peptides. Optimized cryptic peptides are recognized by the immune system as non-self because they target self-cryptic peptides that escape self-tolerance; in addition they are strongly immunogenic because their sequence is modified in order to enhance their affinity for the HLA molecule. The first vaccine based on the optimized cryptic peptide approach, Vx-001, which targets the widely expressed tumor antigen telomerase reverse transcriptase (TERT), has completed a large phase I clinical study and is currently being tested in a randomized phase II trial in non-small cell lung cancer (NSCLC) patients.
Collapse
Affiliation(s)
| | | | - Aude Rougeot
- Vaxon Biotech, 3 rue de l'Arrivée 75015, Paris, France
| | | |
Collapse
|
14
|
Abstract
Overexpressed tumor-self antigens represent the largest group of candidate vaccine targets. Those exhibiting a role in oncogenesis may be some of the least studied but perhaps most promising. This review considers this subset of self antigens by highlighting vaccine efforts for some of the better known members and focusing on TPD52, a new promising vaccine target. We shed light on the importance of both preclinical and clinical vaccine studies demonstrating that tolerance and autoimmunity (presumed to preclude this class of antigens from vaccine development) can be overcome and do not present the obstacle that might have been expected. The potential of this class of antigens for broad application is considered, possibly in the context of low tumor burden or adjuvant therapy, as is the need to understand mechanisms of tolerance that are relatively understudied.
Collapse
Key Words
- ALK, Anaplastic lymphoma kinase
- AR, androgen receptor
- CTL, cytotoxic T lymphocyte
- CTLA-4, cytotoxic T lymphocyte-associated antigen 4
- HLA, human leukocyte antigen
- Her-2/neu, human epithelial growth factor receptor 2
- ODN, oligodeoxynucleotide
- Overexpressed tumor-self antigen
- TAA, tumor associated antigen
- TPD52
- TRAMP, Transgenic adenocarcinoma of the mouse prostate
- Treg, T regulatory cell
- VEGFR2, vascular endothelial growth factor receptor 2
- WT-1, Wilms tumor-1
- hD52
- hD52, human TPD52
- mD52
- mD52, murine TPD52
- oncogenic
- shared
- tumor protein D52
- universal
- vaccine
Collapse
Affiliation(s)
- Robert K Bright
- a Department of Immunology and Molecular Microbiology and the TTUHSC Cancer Center ; Texas Tech University Health Sciences Center ; Lubbock , TX USA
| | | | | |
Collapse
|
15
|
Targeting cryptic epitope with modified antigen coupled to the surface of liposomes induces strong antitumor CD8 T-cell immune responses in vivo. Oncol Rep 2015; 34:2827-36. [PMID: 26398429 PMCID: PMC4722887 DOI: 10.3892/or.2015.4299] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Accepted: 07/20/2015] [Indexed: 02/04/2023] Open
Abstract
Active cancer immunotherapy, such as cancer vaccine, is based on the fundamental knowledge that tumor-associated antigens (TAAs) are presented on MHC molecules for recognition by specific T cells. However, most TAAs are self-antigens and are also expressed on normal tissues, including the thymus. This fact raises the issue of the tolerance of the TAA-specific T-cell repertoire and consequently the inability to trigger a strong and efficient antitumor immune response. In the present study, we used antigens chemically coupled to the surface of liposomes to target telomerase reverse transcriptase (TERT), a widely expressed self/tumor antigen. Taking advantage of the high homology between mouse and human TERT, we investigated immunogenicity and antitumor efficiency of the liposomal TERT peptides in HLA-A*0201 transgenic HHD mice. Using the heteroclitical peptide-modifying approach with antigen-coupled liposomes, we identified a novel cryptic epitope with low affinity for HLA*0201 molecules derived from TERT. The heteroclitical variant derived from this novel low affinity peptide exhibited strong affinity for HLA*0201 molecules. However, it induced only weak CD8 T-cell immune responses in HHD mice when emulsified in IFA. By contrast, when coupled to the surface of the liposomes, it induced powerful CD8 T-cell immune responses which cross-reacted against the original cryptic epitope. The induced CD8 T cells also recognized endogenously TERT-expressing tumor cells and inhibited their growth in HHD mice. These data suggest that heteroclitical antigen derived from low affinity epitope of tumor antigens coupled to the surface of liposome may have a role as an effective cancer vaccine candidate.
Collapse
|
16
|
MU XIYAN, SANG YAXIONG, FANG CHUNJU, SHAO BIN, YANG LU, YAO KUI, ZHAO XITONG, GOU JINHAI, WEI YUQUAN, YI TAO, WU YANG, ZHAO XIA. Immunotherapy of tumors with human telomerase reverse transcriptase immortalized human umbilical vein endothelial cells. Int J Oncol 2015; 47:1901-11. [DOI: 10.3892/ijo.2015.3175] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Accepted: 08/19/2015] [Indexed: 11/06/2022] Open
|
17
|
Obenaus M, Leitão C, Leisegang M, Chen X, Gavvovidis I, van der Bruggen P, Uckert W, Schendel DJ, Blankenstein T. Identification of human T-cell receptors with optimal affinity to cancer antigens using antigen-negative humanized mice. Nat Biotechnol 2015; 33:402-7. [PMID: 25774714 DOI: 10.1038/nbt.3147] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Accepted: 01/12/2015] [Indexed: 12/21/2022]
Abstract
Identifying T-cell receptors (TCRs) that bind tumor-associated antigens (TAAs) with optimal affinity is a key bottleneck in the development of adoptive T-cell therapy of cancer. TAAs are unmutated self proteins, and T cells bearing high-affinity TCRs specific for such antigens are commonly deleted in the thymus. To identify optimal-affinity TCRs, we generated antigen-negative humanized mice with a diverse human TCR repertoire restricted to the human leukocyte antigen (HLA) A*02:01 (ref. 3). These mice were immunized with human TAAs, for which they are not tolerant, allowing induction of CD8⁺ T cells with optimal-affinity TCRs. We isolate TCRs specific for the cancer/testis (CT) antigen MAGE-A1 (ref. 4) and show that two of them have an anti-tumor effect in vivo. By comparison, human-derived TCRs have lower affinity and do not mediate substantial therapeutic effects. We also identify optimal-affinity TCRs specific for the CT antigen NY-ESO. Our humanized mouse model provides a useful tool for the generation of optimal-affinity TCRs for T-cell therapy.
Collapse
Affiliation(s)
| | | | | | - Xiaojing Chen
- Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
| | | | - Pierre van der Bruggen
- 1] Ludwig Institute for Cancer Research and WELBIO, Brussels, Belgium. [2] De Duve Institute, Université Catholique de Louvain, Brussels, Belgium
| | - Wolfgang Uckert
- 1] Max-Delbrück-Center for Molecular Medicine, Berlin, Germany. [2] Institute of Biology, Humboldt University, Berlin, Germany
| | | | - Thomas Blankenstein
- 1] Max-Delbrück-Center for Molecular Medicine, Berlin, Germany. [2] Institute of Immunology, Charité Campus Buch, Berlin, Germany
| |
Collapse
|
18
|
The first step of peptide selection in antigen presentation by MHC class I molecules. Proc Natl Acad Sci U S A 2015; 112:1505-10. [PMID: 25605945 DOI: 10.1073/pnas.1416543112] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
MHC class I molecules present a variable but limited repertoire of antigenic peptides for T-cell recognition. Understanding how peptide selection is achieved requires mechanistic insights into the interactions between the MHC I and candidate peptides. We find that, at first encounter, MHC I H-2K(b) considers a wide range of peptides, including those with expanded N termini and unfitting anchor residues. Discrimination occurs in the second step, when noncanonical peptides dissociate with faster exchange rates. This second step exhibits remarkable temperature sensitivity, as illustrated by numerous noncanonical peptides presented by H-2K(b) in cells cultured at 26 °C relative to 37 °C. Crystallographic analyses of H-2K(b)-peptide complexes suggest that a conformational adaptation of H-2K(b) drives the decisive step in peptide selection. We propose that MHC class I molecules consider initially a large peptide pool, subsequently refined by a temperature-sensitive induced-fit mechanism to retain the canonical peptide repertoire.
Collapse
|
19
|
Choi M, Thakur A. Identifying Appropriate Colorectal Cancer-Associated Antigens for the Clinical Trials. CURRENT COLORECTAL CANCER REPORTS 2014. [DOI: 10.1007/s11888-014-0256-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
20
|
Humar M, Azemar M, Maurer M, Groner B. Adaptive Resistance to Immunotherapy Directed Against p53 Can be Overcome by Global Expression of Tumor-Antigens in Dendritic Cells. Front Oncol 2014; 4:270. [PMID: 25340039 PMCID: PMC4186483 DOI: 10.3389/fonc.2014.00270] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Accepted: 09/17/2014] [Indexed: 12/13/2022] Open
Abstract
Immunotherapy of cancer utilizes dendritic cells (DCs) for antigen presentation and the induction of tumor-specific immune responses. However, the therapeutic induction of anti-tumor immunity is limited by tumor escape mechanisms. In this study, immortalized dendritic D2SC/1 cells were transduced with a mutated version of the p53 tumor suppressor gene, p53M234I, or p53C132F/E168G, which are overexpressed in MethA fibrosarcoma tumor cells. In addition, D2SC/1 cells were fused with MethA tumor cells to generate a vaccine that potentially expresses a large repertoire of tumor-antigens. Cellular vaccines were transplanted onto Balb/c mice and MethA tumor growth and anti-tumor immune responses were examined in vaccinated animals. D2SC/1–p53M234I and D2SC/1–p53C132F/E168G cells induced strong therapeutic and protective MethA tumor immunity upon transplantation in Balb/c mice. However, in a fraction of immunized mice MethA tumor growth resumed after an extended latency period. Analysis of these tumors indicated loss of p53 expression. Mice, pre-treated with fusion hybrids generated from D2SC/1 and MethA tumor cells, suppressed MethA tumor growth and averted adaptive immune escape. Polyclonal B-cell responses directed against various MethA tumor proteins could be detected in the sera of D2SC/1–MethA inoculated mice. Athymic nude mice and Balb/c mice depleted of CD4+ or CD8+ T-cells were not protected against MethA tumor cell growth after immunization with D2SC/1–MethA hybrids. Our results highlight a potential drawback of cancer immunotherapy by demonstrating that the induction of a specific anti-tumor response favors the acquisition of tumor phenotypes promoting immune evasion. In contrast, the application of DC/tumor cell fusion hybrids prevents adaptive immune escape by a T-cell dependent mechanism and provides a simple strategy for personalized anti-cancer treatment without the need of selectively priming the host immune system.
Collapse
Affiliation(s)
- Matjaz Humar
- Pharmaceutical Biology and Biotechnology, Albert-Ludwigs-University of Freiburg , Freiburg , Germany
| | - Marc Azemar
- Internistische Onkologie, Tumor Biology Center , Freiburg , Germany
| | - Martina Maurer
- Basilea Pharmaceutica International Ltd. , Basel , Switzerland
| | - Bernd Groner
- Institute for Biomedical Research, Georg Speyer Haus , Frankfurt am Main , Germany
| |
Collapse
|
21
|
Matsushita H, Hosoi A, Ueha S, Abe J, Fujieda N, Tomura M, Maekawa R, Matsushima K, Ohara O, Kakimi K. Cytotoxic T lymphocytes block tumor growth both by lytic activity and IFNγ-dependent cell-cycle arrest. Cancer Immunol Res 2014; 3:26-36. [PMID: 25127875 DOI: 10.1158/2326-6066.cir-14-0098] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
To understand global effector mechanisms of CTL therapy, we performed microarray gene expression analysis in a murine model using pmel-1 T-cell receptor (TCR) transgenic T cells as effectors and B16 melanoma cells as targets. In addition to upregulation of genes related to antigen presentation and the MHC class I pathway, and cytotoxic effector molecules, cell-cycle-promoting genes were downregulated in the tumor on days 3 and 5 after CTL transfer. To investigate the impact of CTL therapy on the cell cycle of tumor cells in situ, we generated B16 cells expressing a fluorescent ubiquitination-based cell-cycle indicator (B16-fucci) and performed CTL therapy in mice bearing B16-fucci tumors. Three days after CTL transfer, we observed diffuse infiltration of CTLs into the tumor with a large number of tumor cells arrested at the G1 phase of the cell cycle, and the presence of spotty apoptotic or necrotic areas. Thus, tumor growth suppression was largely dependent on G1 cell-cycle arrest rather than killing by CTLs. Neutralizing antibody to IFNγ prevented both tumor growth inhibition and G1 arrest. The mechanism of G1 arrest involved the downregulation of S-phase kinase-associated protein 2 (Skp2) and the accumulation of its target cyclin-dependent kinase inhibitor p27 in the B16-fucci tumor cells. Because tumor-infiltrating CTLs are far fewer in number than the tumor cells, we propose that CTLs predominantly regulate tumor growth via IFNγ-mediated profound cytostatic effects rather than via cytotoxicity. This dominance of G1 arrest over other mechanisms may be widespread but not universal because IFNγ sensitivity varied among tumors.
Collapse
Affiliation(s)
- Hirokazu Matsushita
- Department of Immunotherapeutics, The University of Tokyo Hospital, Tokyo, Japan
| | - Akihiro Hosoi
- Department of Immunotherapeutics, The University of Tokyo Hospital, Tokyo, Japan. Medinet Co Ltd., Yokohama, Japan
| | - Satoshi Ueha
- Department of Molecular Preventive Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Jun Abe
- Department of Molecular Preventive Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Nao Fujieda
- Department of Immunotherapeutics, The University of Tokyo Hospital, Tokyo, Japan. Medinet Co Ltd., Yokohama, Japan
| | - Michio Tomura
- Center for Innovation in Immunoregulative Technology and Therapeutics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | | | - Kouji Matsushima
- Department of Molecular Preventive Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Osamu Ohara
- Department of Human Genome Research, Kazusa DNA Research Institute, Chiba, Japan
| | - Kazuhiro Kakimi
- Department of Immunotherapeutics, The University of Tokyo Hospital, Tokyo, Japan.
| |
Collapse
|
22
|
Toomer KH, Chen Z. Autoimmunity as a double agent in tumor killing and cancer promotion. Front Immunol 2014; 5:116. [PMID: 24672527 PMCID: PMC3957029 DOI: 10.3389/fimmu.2014.00116] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2013] [Accepted: 03/05/2014] [Indexed: 12/19/2022] Open
Abstract
Cancer immunotherapy through manipulation of the immune system holds great potential for the treatment of human cancers. However, recent trials targeting the negative immune regulators cytotoxic T-lymphocyte antigen 4, programed death 1 (PD-1), and PD-1 receptor ligand (PD-L1) demonstrated that clinically significant antitumor responses were often associated with the induction of autoimmune toxicity. This finding suggests that the same immune mechanisms that elicit autoimmunity may also contribute to the destruction of tumors. Given the fact that the immunological identity of tumors might be largely an immunoprivileged self, autoimmunity may not represent a wholly undesirable outcome in the context of cancer immunotherapy. Rather, targeted killing of cancer cells and autoimmune damage to healthy tissues may be intricately linked through molecular mechanisms, in particular inflammatory cytokine signaling. On the other hand, since chronic inflammation is a well-recognized condition that promotes tumor development, it appears that autoimmunity can be a "double agent" in mediating either pro-tumor or antitumor effects. This review surveys the tumor-promoting and tumoricidal activities of several prominent cytokines: IFN-γ, TNF-α, TGF-β, IL-17, IL-23, IL-4, and IL-13, produced by three major subsets of T helper cells that interact with innate immune cells. Many of these cytokines exert divergent and seemingly contradictory effects on cancer development in different human and animal models, suggesting a high degree of context dependence in their functions. We hypothesize that these inflammatory cytokines could mediate a feedback loop of autoimmunity, antitumor immunity, and tumorigenesis. Understanding the diverse and paradoxical roles of cytokines from autoimmune responses in the setting of cancer will advance the long-term goal of improving cancer immunotherapy, while minimizing the hazards of immune-mediated tissue damage and the possibility of de novo tumorigenesis, through proper monitoring and preventive measures.
Collapse
Affiliation(s)
- Kevin H Toomer
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine , Miami, FL , USA
| | - Zhibin Chen
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine , Miami, FL , USA ; Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine , Miami, FL , USA
| |
Collapse
|
23
|
Miska J, Bas E, Devarajan P, Chen Z. Autoimmunity-mediated antitumor immunity: tumor as an immunoprivileged self. Eur J Immunol 2012; 42:2584-96. [PMID: 22777737 DOI: 10.1002/eji.201242590] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2012] [Revised: 05/22/2012] [Accepted: 06/20/2012] [Indexed: 11/09/2022]
Abstract
The association of autoimmunity with antitumor immunity challenges a paradigm of selective surveillance against tumors. Aided with well-characterized models of robust autoimmunity, we show that self-antigen-specific effector T (Teff) cell clones could eradicate tumor cells. However, a tumor microenvironment reinforced by Treg cells and myeloid-derived suppressor cells (MDSCs) presented a barrier to the autoimmune effectors, more so in tumors than in healthy tissues. This barrier required optimal CTLA4 expression in Teff cells. In a spontaneous model of breast cancer, subtle reductions in CTLA4 expression impeded tumor onset and progression, providing the first direct evidence that CTLA4 inhibits spontaneous tumor development. In an adoptive therapy model of lymphoma, self-antigen-specific Teff cells were potentiated by even a modest reduction of CTLA4. A subtle reduction of CTLA4 did not curtail Treg-cell suppression. Thus, Teff cells had an exquisite sensitivity to physiological levels of CTLA4 variations. However, both Treg and Teff cells were impacted by anti-CTLA4 antibody blockade. Therefore, whether CTLA4 impacts through Treg cells or Teff cells depends on its expression level. Overall, the results suggest that the tumor microenvironment represents an "immunoprivileged self" that could be overcome practically and at least partially by RNAi silencing of CTLA4 in Teff cells.
Collapse
Affiliation(s)
- Jason Miska
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | | | | | | |
Collapse
|
24
|
Vermeij R, Leffers N, Hoogeboom BN, Hamming ILE, Wolf R, Reyners AKL, Molmans BHW, Hollema H, Bart J, Drijfhout JW, Oostendorp J, van der Zee AGJ, Melief CJ, van der Burg SH, Daemen T, Nijman HW. Potentiation of a p53-SLP vaccine by cyclophosphamide in ovarian cancer: a single-arm phase II study. Int J Cancer 2012; 131:E670-80. [PMID: 22139992 DOI: 10.1002/ijc.27388] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2011] [Accepted: 10/21/2011] [Indexed: 01/21/2023]
Abstract
The purpose of the current phase II single-arm clinical trial was to evaluate whether pretreatment with low-dose cyclophosphamide improves immunogenicity of a p53-synthetic long peptide (SLP) vaccine in patients with recurrent ovarian cancer. Patients with ovarian cancer with elevated serum levels of CA-125 after primary treatment were immunized four times with the p53-SLP vaccine. Each immunization was preceded by administration of 300 mg/m2 intravenous cyclophosphamide as a means to affect regulatory T cells (Tregs). Vaccine-induced p53-specific interferon-gamma (IFN-γ)-producing T cells evaluated by IFN-γ ELISPOT were observed in 90% (9/10) and 87.5% (7/8) of evaluable patients after two and four immunizations, respectively. Proliferative p53-specific T cells, observed in 80.0% (8/10) and 62.5% (5/8) of patients, produced both T-helper 1 and T-helper-2 cytokines. Cyclophosphamide induced neither a quantitative reduction of Tregs determined by CD4+ FoxP3+ T cell levels nor a demonstrable qualitative difference in Treg function tested in vitro. Nonetheless, the number of vaccine-induced p53-specific IFN-γ-producing T cells was higher in our study compared to a study in which a similar patient group was treated with p53-SLP monotherapy (p≤0.012). Furthermore, the strong reduction in the number of circulating p53-specific T cells observed previously after four immunizations was currently absent. Stable disease was observed in 20.0% (2/10) of patients, and the remainder of patients (80.0%) showed clinical, biochemical and/or radiographic evidence of progressive disease. The outcome of this phase II trial warrants new studies on the use of low-dose cyclophosphamide to potentiate the immunogenicity of the p53-SLP vaccine or other antitumor vaccines.
Collapse
Affiliation(s)
- Renee Vermeij
- Department of Gynecologic Oncology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Quakkelaar ED, Melief CJM. Experience with synthetic vaccines for cancer and persistent virus infections in nonhuman primates and patients. Adv Immunol 2012; 114:77-106. [PMID: 22449779 DOI: 10.1016/b978-0-12-396548-6.00004-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Synthetic vaccines, in particular long synthetic peptides of approximately 25-50 amino acids in length, are attractive for HIV vaccine development and for induction of therapeutic immune responses in patients with (pre-)malignant disorders. In the case of preventive vaccine development against HIV, no major success has been achieved, but the possibilities are by no means exhausted. A long peptide vaccine consisting of 13 overlapping peptides, which together cover the entire length of the two oncogenic proteins E6 and E7 of high-risk human papilloma virus type 16 (HPV16), caused complete regression of all lesions and eradication of virus in 9 out of 20 women with high-grade vulvar intraepithelial neoplasia, a therapy-resistant preneoplastic disorder. The nature and strength of the vaccine-prompted T cell responses were significantly correlated with the clinical response. Synthetic peptide vaccines are attractive, because they allow rational improvement of vaccine design and detailed pharmacokinetic and pharmacodynamic studies not possible with conventional vaccines. Improvements are possible by addition or conjugation of adjuvants, notably TLR ligands, to the synthetic peptides.
Collapse
Affiliation(s)
- Esther D Quakkelaar
- Department of Immunohematology, Leiden University Medical Center, Leiden, The Netherlands
| | | |
Collapse
|
26
|
Abstract
T-cell receptor (TCR) gene therapy aims to induce immune reactivity against tumors by introducing genes encoding a tumor-reactive TCR into patient T cells. This approach has been extensively tested in preclinical mouse models, and initial clinical trials have demonstrated the feasibility and potential of TCR gene therapy as a cancer treatment. However, data obtained from preclinical and clinical studies suggest that both the therapeutic efficacy and the safety of TCR gene therapy can be and needs to be further enhanced. This review highlights those strategies that can be followed to develop TCR gene therapy into a clinically relevant treatment option for cancer patients.
Collapse
|
27
|
Gooden MJM, de Bock GH, Leffers N, Daemen T, Nijman HW. The prognostic influence of tumour-infiltrating lymphocytes in cancer: a systematic review with meta-analysis. Br J Cancer 2011; 105:93-103. [PMID: 21629244 PMCID: PMC3137407 DOI: 10.1038/bjc.2011.189] [Citation(s) in RCA: 980] [Impact Index Per Article: 70.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Background: Tumour-infiltrating lymphocytes (TILs) are often found in tumours, presumably reflecting an immune response against the tumour. We carried out a systematic review and meta-analysis, aiming to establish pooled estimates for survival outcomes based on the presence of TILs in cancer. Methods: A Pubmed and Embase literature search was designed. Studies were included, in which the prognostic significance of intratumoural CD3+, CD4+, CD8+, and FoxP3+ lymphocytes, as well as ratios between these subsets, were determined in solid tumours. Results: In pooled analysis, CD3+ TILs had a positive effect on survival with a hazard ratio (HR) of 0.58 (95% confidence interval (CI) 0.43–0.78) for death, as did CD8+ TILs with a HR of 0.71 (95% CI 0.62–0.82). FoxP3+ regulatory TILs were not linked to overall survival, with a HR of 1.19 (95% CI 0.84–1.67). The CD8/FoxP3 ratio produced a more impressive HR (risk of death: HR 0.48, 95% CI 0.34–0.68), but was used in relatively few studies. Sample size and follow-up time seemed to influence study outcomes. Conclusion: Any future studies should be carefully designed, to prevent overestimating the effect of TILs on prognosis. In this context, ratios between TIL subsets may be more informative.
Collapse
Affiliation(s)
- M J M Gooden
- Department of Obstetrics and Gynecology, University Medical Center Groningen, University of Groningen, PO Box 30.001, 9700 RB Groningen, The Netherlands
| | | | | | | | | |
Collapse
|
28
|
Immunological and clinical effects of vaccines targeting p53-overexpressing malignancies. J Biomed Biotechnol 2011; 2011:702146. [PMID: 21541192 PMCID: PMC3085500 DOI: 10.1155/2011/702146] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2010] [Revised: 12/13/2010] [Accepted: 01/18/2011] [Indexed: 12/20/2022] Open
Abstract
Approximately 50% of human malignancies carry p53 mutations, which makes it a potential antigenic target for cancer immunotherapy. Adoptive transfer with p53-specific cytotoxic T-lymphocytes (CTL) and CD4+ T-helper cells eradicates p53-overexpressing tumors in mice. Furthermore, p53 antibodies and p53-specific CTLs can be detected in cancer patients, indicating that p53 is immunogenic. Based on these results, clinical trials were initiated. In this paper, we review immunological and clinical responses observed in cancer patients vaccinated with p53 targeting vaccines. In most trials, p53-specific vaccine-induced immunological responses were observed. Unfortunately, no clinical responses with significant reduction of tumor-burden have occurred. We will elaborate on possible explanations for this lack of clinical effectiveness. In the second part of this paper, we summarize several immunopotentiating combination strategies suitable for clinical use. In our opinion, future p53-vaccine studies should focus on addition of these immunopotentiating regimens to achieve clinically effective therapeutic vaccination strategies for cancer patients.
Collapse
|
29
|
Jorritsma A, Schumacher TNM, Haanen JBAG. Immunotherapeutic strategies: the melanoma example. Immunotherapy 2011; 1:679-90. [PMID: 20635992 DOI: 10.2217/imt.09.20] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
T-cell-based immunotherapy can be induced by nonspecific activation, by antigen-specific immunization, or by adoptive immunotherapy. In this review, progress in these areas is discussed as based on data from clinical trials for the treatment of metastatic melanoma. Nonspecific immunotherapy has been shown to result in low, but in some cases significant, levels of objective tumor responses, and is often associated with autoimmune reactions. Antigen-specific targeting of tumors via vaccination has only resulted in low to very low levels of objective responses, and these strategies seem to have most value when the T-cell repertoire is not affected by tolerance. Finally, adoptive immunotherapy can be applied by in vitro expansion of autologous lymphocytes that have escaped tolerance or by genetic transfer of allogeneic T-cell receptors (TCRs). Autologous adoptive T-cell transfer has resulted in a very high frequency of clinical responses when combined with chemotherapy and IL-2 administration in single-center studies. Although TCR gene transfer has, until now, only resulted in a low frequency of clinical responses, it does have a broader application potential, and optimization of this strategy is likely to improve its efficacy.
Collapse
Affiliation(s)
- Annelies Jorritsma
- Division of Immunology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands.
| | | | | |
Collapse
|
30
|
Chiappori AA, Soliman H, Janssen WE, Antonia SJ, Gabrilovich DI. INGN-225: a dendritic cell-based p53 vaccine (Ad.p53-DC) in small cell lung cancer: observed association between immune response and enhanced chemotherapy effect. Expert Opin Biol Ther 2010; 10:983-91. [PMID: 20420527 DOI: 10.1517/14712598.2010.484801] [Citation(s) in RCA: 95] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
IMPORTANCE OF THE FIELD Novel approaches are needed for patients with small cell lung cancer (SCLC), as response after relapse is poor with standard therapies. p53 gene mutations often occur, resulting in tumoral protein overexpression and allowing for their recognition by p53-specific cytotoxic T cells. AREAS COVERED IN THIS REVIEW We describe the characteristics and manufacturing of INGN-225, a p53-modified adenovirus-tranduced dendritic cell vaccine, and review available data, to understand INGN-225's role in SCLC treatment. We discuss our pre-clinical, early Phase I/II, and ongoing randomized Phase II studies. WHAT THE READER WILL GAIN INGN-225 was well tolerated (all toxicities <or=grade 2) in the Phase I/II trial (54 patients receiving at least 1 dose). Specific anti-p53 immune response was positive in 18/43 (41.8%) patients, with overall post-INGN-225 response observed in 17/33 (51.5%) and immune response data available in 29 (14 positive, 15 negative). Post-INGN-225 response was observed in 11/14 (78.6%) and 5/15 (33%) patients with positive and negative immune responses, respectively. TAKE HOME MESSAGE INGN-225 is safe, induces a significant immune response, and appears to sensitize SCLC to subsequent chemotherapy. Improvements in immune response induction and understanding the chemotherapy-immunotherapy synergism will determine INGN-225's future role as an anticancer therapy.
Collapse
Affiliation(s)
- Alberto A Chiappori
- Department of Thoracic Oncology, H Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive, Tampa, FL 33612, USA.
| | | | | | | | | |
Collapse
|
31
|
Therapeutic cancer vaccines in combination with conventional therapy. J Biomed Biotechnol 2010; 2010:237623. [PMID: 20617155 PMCID: PMC2896846 DOI: 10.1155/2010/237623] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2010] [Revised: 05/05/2010] [Accepted: 05/17/2010] [Indexed: 12/16/2022] Open
Abstract
The clinical efficacy of most therapeutic vaccines against cancer has not yet met its promise. Data are emerging that strongly support the notion that combining immunotherapy with conventional therapies, for example, radiation and chemotherapy may improve efficacy. In particular combination with chemotherapy may lead to improved clinical efficacy by clearing suppressor cells, reboot of the immune system, by rendering tumor cells more susceptible to immune mediated killing, or by activation of cells of the immune system. In addition, a range of tumor antigens have been characterized to allow targeting of proteins coupled to intrinsic properties of cancer cells. For example, proteins associated with drug resistance can be targeted, and form ideal target structures for use in combination with chemotherapy for killing of surviving drug resistant cancer cells. Proteins associated with the malignant phenotype can be targeted to specifically target cancer cells, but proteins targeted by immunotherapy may also simultaneously target cancer cells as well as suppressive cells in the tumor stroma.
Collapse
|
32
|
Simmons O, Magee M, Nemunaitis J. Current vaccine updates for lung cancer. Expert Rev Vaccines 2010; 9:323-35. [PMID: 20218860 DOI: 10.1586/erv.10.12] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Current treatments for lung cancer are far from optimal. Several immunotherapeutic strategies involving vaccines incorporating different tumor-associated antigens to induce immune responses against tumors are being tested in clinical trials internationally. Although small, benefits have indeed been observed from the early studies of these vaccines, and the future is looking brighter for lung cancer patients as a handful of these immunotherapies reach Phase III trials. In addition, optimizing the induced immune response by these vaccines has become a priority, and a number of techniques are being considered, including addition of adjuvants and combining vaccines, which affect synergy based on their mechanism of action. This review is an update on the current vaccines in production, the benefits observed from their most recent studies, and the upcoming plans for improvements in these immunotherapies.
Collapse
|
33
|
Buschow C, Charo J, Anders K, Loddenkemper C, Jukica A, Alsamah W, Perez C, Willimsky G, Blankenstein T. In vivo imaging of an inducible oncogenic tumor antigen visualizes tumor progression and predicts CTL tolerance. THE JOURNAL OF IMMUNOLOGY 2010; 184:2930-8. [PMID: 20142365 DOI: 10.4049/jimmunol.0900893] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Visualizing oncogene/tumor Ag expression by noninvasive imaging is of great interest for understanding processes of tumor development and therapy. We established transgenic (Tg) mice conditionally expressing a fusion protein of the SV40 large T Ag and luciferase (TagLuc) that allows monitoring of oncogene/tumor Ag expression by bioluminescent imaging upon Cre recombinase-mediated activation. Independent of Cre-mediated recombination, the TagLuc gene was expressed at low levels in different tissues, probably due to the leakiness of the stop cassette. The level of spontaneous TagLuc expression, detected by bioluminescent imaging, varied between the different Tg lines, depended on the nature of the Tg expression cassette, and correlated with Tag-specific CTL tolerance. Following liver-specific Cre-loxP site-mediated excision of the stop cassette that separated the promoter from the TagLuc fusion gene, hepatocellular carcinoma development was visualized. The ubiquitous low level TagLuc expression caused the failure of transferred effector T cells to reject Tag-expressing tumors rather than causing graft-versus-host disease. This model may be useful to study different levels of tolerance, monitor tumor development at an early stage, and rapidly visualize the efficacy of therapeutic intervention versus potential side effects of low-level Ag expression in normal tissues.
Collapse
Affiliation(s)
- Christian Buschow
- Institute of Immunology, Charité Campus Benjamin Franklin, Berlin, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Vaccine Therapy for Lung Cancer. Lung Cancer 2010. [DOI: 10.1007/978-1-60761-524-8_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
35
|
Theoret MR, Cohen CJ, Nahvi AV, Ngo LT, Suri KB, Powell DJ, Dudley ME, Morgan RA, Rosenberg SA. Relationship of p53 overexpression on cancers and recognition by anti-p53 T cell receptor-transduced T cells. Hum Gene Ther 2009; 19:1219-32. [PMID: 19848582 DOI: 10.1089/hum.2008.083] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Tumor suppressor p53 is reported to be an attractive immunotherapy target because it is mutated in approximately half of human cancers, resulting in inactivation and often an accumulation of the protein in the tumor cells. Only low amounts of protein are detectable in normal tissues. The differential display of antigen in normal versus tumor tissues has been reported to create an opportunity to target p53 by immunotherapy. We sought to determine the relationship between p53 expression and its recognition by cognate T cells in human tumors including common epithelial malignancies. Inasmuch as nonsense or missense p53 mutations may disrupt processing and presentation, we studied tumors with either identified wild-type or mutated p53, based on our gene-sequencing studies or published data. T cells transduced with a high-affinity, p53(264-272)-reactive T cell receptor (TCR) derived from HLA-A2.1 transgenic mice recognized a wide panel of human tumor lines. There was no significant correlation between p53 expression in tumors and recognition by the anti-p53 TCR-transduced T cells. This conclusion was based on the study of 48 cell lines and is in contrast to several prior studies that used only a limited number of selected cell lines. A panel of normal cells was evaluated for recognition, and some of these populations were capable of stimulating anti-p53 T cells, albeit at low levels. These studies raise doubts concerning the suitability of targeting p53 in the immunotherapy of cancer patients.
Collapse
Affiliation(s)
- Marc R Theoret
- Surgery Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892-1201, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Solcia E, Klersy C, Mastracci L, Alberizzi P, Candusso ME, Diegoli M, Tava F, Riboni R, Manca R, Luinetti O. A combined histologic and molecular approach identifies three groups of gastric cancer with different prognosis. Virchows Arch 2009; 455:197-211. [PMID: 19672623 DOI: 10.1007/s00428-009-0813-z] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2009] [Revised: 07/06/2009] [Accepted: 07/15/2009] [Indexed: 12/22/2022]
Abstract
The limited prognostic value of currently used histologic classifications of gastric cancer and their failure to account for the complexity of the disease as revealed by more recent investigations prompted a combined reinvestigation of histologic, molecular, and clinicopathologic patterns in 294 extensively sampled, invasive gastric cancers representing all main histotypes and stages of the disease and followed for a median of 150 months. Among histologic parameters tested, only cellular atypia, angio-lympho- or neuroinvasion, Ki67 proliferation index, expansile/infiltrative type growth, and T8 cell-rich high lymphoid intra-/peritumor response (HLR) proved to be stage-independent predictors of patient survival. Among molecular tests, p53 gene exon 7 (loop 3) and 8 (loop-sheet-helix motif and S-10 band), but not p53 protein overexpression, TP53 LOH or 18qLOH, were found to worsen prognosis. Microsatellite DNA instability was a favorable prognostic factor when coupled with HLR. Patient survival analysis of the main histotypes and their subtypes confirmed the favorable prognosis of HLR, well-differentiated tubular, muconodular, and low grade diffuse desmoplastic cancers, and highlighted the worse prognosis of anaplastic and infiltrative-lymphoinvasive mucinous cancers compared to ordinary cohesive and diffuse cancers. Distinct roles of individual morphologic and molecular factors in tumor progression of the different histotypes have been recognized. The combination of survival-predictive histotypes and individual histologic or molecular parameters allowed us to develop a classification of all gastric cancers into three grades of increasing malignancy which proved to be of high prognostic value.
Collapse
Affiliation(s)
- Enrico Solcia
- Anatomic Pathology Service, IRCCS Policlinico S. Matteo Foundation, Pavia, Italy.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Passoni L, Gambacorti-Passerini C. ALK a Novel Lymphoma-associated Tumor Antigen for Vaccination Strategies. Leuk Lymphoma 2009; 44:1675-81. [PMID: 14692518 DOI: 10.1080/1042819031000099625] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
The discovery of Tumor Associated Antigens (TAAs) demonstrated that tumor cells can be specifically recognized by the immune system raising the hypothesis that tumors express antigens that Cytotoxic T Lymphocytes (CTLs) can potentially attack. The identification of immunogenic epitopes led to their use as targets to mediate the specific clearance of neoplastic cells by TAA targeting strategies such as vaccination strategies. One of the critical issues in the development of efficient vaccination protocols is the identification of the appropriate TAAs. The TAA should be effective as a "tumor rejection antigen" able to induce an immune response that will affect tumor growth. A distinct pathologic entity characterized by the expression of the Anaplastic Lymphoma Kinase (ALK) protein and named "ALKoma" has recently emerged within the heterogeneous group of CD30+ Anaplastic Large Cell Lymphoma (ALCL). ALK is a receptor tyrosine kinase whose expression is normally restricted to a few scattered cells in the nervous system. Its pathological expression in lymphoma cells is due to a chromosomal translocation that leads to the formation of an ALK-derived oncogenic fusion proteins. ALK fusion proteins ectopically over-expressed and constitutively activated in lymphoid cells play a key role in the neoplastic transformation by the aberrant phosphorylation of intracellular substrates that likely contributes to the molecular pathogenesis of ALCL. The high level of ALK expression in lymphoma cells and its direct role in lymphomagenesis, combined with the fact that normal ALK is expressed at low levels in the immune privileged nervous system, makes ALK an ideal lymphoma-specific target for immunotherapy of ALK+ALCL.
Collapse
Affiliation(s)
- Lorena Passoni
- Oncogenic Fusion Genes and Proteins Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori, Milan, Italy.
| | | |
Collapse
|
38
|
DeLeo AB, Whiteside TL. Development of multi-epitope vaccines targeting wild-type sequence p53 peptides. Expert Rev Vaccines 2008; 7:1031-40. [PMID: 18767952 DOI: 10.1586/14760584.7.7.1031] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Loss of p53 tumor-suppressor function is the most common abnormality in human cancer, which can result in enhanced presentation to immune cells of wild-type (wt)-sequence peptides from tumor p53 molecules, thus providing the rationale for wt p53 peptide-based cancer vaccines. We review evidence from preclinical murine tumor models and preclinical studies that led to the clinical introduction of wt p53 peptide-based vaccines for cancer immunotherapy. Overall, this review illustrates the complex process of wt p53 epitope selection and the issues and concerns involved in the application of p53-based vaccines for patients with cancer.
Collapse
Affiliation(s)
- Albert B DeLeo
- Department of Pathology, University of Pittsburgh School of Medicine and University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
| | | |
Collapse
|
39
|
Barve M, Bender J, Senzer N, Cunningham C, Greco FA, McCune D, Steis R, Khong H, Richards D, Stephenson J, Ganesa P, Nemunaitis J, Ishioka G, Pappen B, Nemunaitis M, Morse M, Mills B, Maples PB, Sherman J, Nemunaitis JJ. Induction of Immune Responses and Clinical Efficacy in a Phase II Trial of IDM-2101, a 10-Epitope Cytotoxic T-Lymphocyte Vaccine, in Metastatic Non–Small-Cell Lung Cancer. J Clin Oncol 2008; 26:4418-25. [DOI: 10.1200/jco.2008.16.6462] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Purpose Generation of broad cytotoxic T-lymphocyte responses against multiple epitopes and tumor-associated antigens (TAAs) may provide effective immunotherapy in patients with cancer. We evaluated a single-vial peptide vaccine consisting of nine HLA-A2 supertype-binding epitopes (two native and seven analog epitopes modified for optimal HLA binding or T-cell receptor stimulation) covering five TAAs and the universal helper pan-DR epitope, formulated as a stable emulsion with incomplete Freund's adjuvant (Montanide ISA 51; Seppic SA, Paris, France). The clinical efficacy, safety, and multiepitope immunogenicity of IDM-2101 was evaluated in patients with stage IIIB or IV non–small-cell lung cancer (NSCLC). Patients and Methods A total of 63 patients were enrolled who were positive for HLA-A2. End points included survival, safety, and immune response. IDM-2101 (previously EP-2101) was administered every 3 weeks for the first 15 weeks, then every 2 months through year 1, then quarterly through year 2, for a total of 13 doses. Epitope-specific cytotoxic and helper T-lymphocyte immunogenic responses were measured by the interferon gamma enzyme-linked immunosorbent spot assay. Results No significant adverse events were noted. Low-grade erythema and pain at the injection site were the most common adverse effects. One-year survival in the treated patients was 60%, and median survival was 17.3 months. One complete and one partial response were identified. Survival was longer in patients demonstrating an immune response to epitope peptides (P < .001). Conclusion IDM-2101 was well tolerated, and evidence of efficacy was suggested.
Collapse
Affiliation(s)
- Minal Barve
- From the Mary Crowley Cancer Research Centers; Baylor Sammons Cancer Center; Gradalis Inc; and Texas Oncology Physicians Association, Dallas; Tyler Cancer Center, Tyler, TX; IDM Pharma Inc, Irvine; Pharmexa-Epimmune, San Diego, CA; Sarah Cannon Cancer Center, Nashville, TN; Madigan Army Medical Center, Tacoma WA; Atlanta Cancer Care, Roswell, GA; University of South Alabama, Mitchell Cancer Institute, Mobile, AL; Cancer Center of the Carolinas, Greenville, SC; and Duke University Medical Center, Durham, NC
| | - James Bender
- From the Mary Crowley Cancer Research Centers; Baylor Sammons Cancer Center; Gradalis Inc; and Texas Oncology Physicians Association, Dallas; Tyler Cancer Center, Tyler, TX; IDM Pharma Inc, Irvine; Pharmexa-Epimmune, San Diego, CA; Sarah Cannon Cancer Center, Nashville, TN; Madigan Army Medical Center, Tacoma WA; Atlanta Cancer Care, Roswell, GA; University of South Alabama, Mitchell Cancer Institute, Mobile, AL; Cancer Center of the Carolinas, Greenville, SC; and Duke University Medical Center, Durham, NC
| | - Neil Senzer
- From the Mary Crowley Cancer Research Centers; Baylor Sammons Cancer Center; Gradalis Inc; and Texas Oncology Physicians Association, Dallas; Tyler Cancer Center, Tyler, TX; IDM Pharma Inc, Irvine; Pharmexa-Epimmune, San Diego, CA; Sarah Cannon Cancer Center, Nashville, TN; Madigan Army Medical Center, Tacoma WA; Atlanta Cancer Care, Roswell, GA; University of South Alabama, Mitchell Cancer Institute, Mobile, AL; Cancer Center of the Carolinas, Greenville, SC; and Duke University Medical Center, Durham, NC
| | - Casey Cunningham
- From the Mary Crowley Cancer Research Centers; Baylor Sammons Cancer Center; Gradalis Inc; and Texas Oncology Physicians Association, Dallas; Tyler Cancer Center, Tyler, TX; IDM Pharma Inc, Irvine; Pharmexa-Epimmune, San Diego, CA; Sarah Cannon Cancer Center, Nashville, TN; Madigan Army Medical Center, Tacoma WA; Atlanta Cancer Care, Roswell, GA; University of South Alabama, Mitchell Cancer Institute, Mobile, AL; Cancer Center of the Carolinas, Greenville, SC; and Duke University Medical Center, Durham, NC
| | - F. Anthony Greco
- From the Mary Crowley Cancer Research Centers; Baylor Sammons Cancer Center; Gradalis Inc; and Texas Oncology Physicians Association, Dallas; Tyler Cancer Center, Tyler, TX; IDM Pharma Inc, Irvine; Pharmexa-Epimmune, San Diego, CA; Sarah Cannon Cancer Center, Nashville, TN; Madigan Army Medical Center, Tacoma WA; Atlanta Cancer Care, Roswell, GA; University of South Alabama, Mitchell Cancer Institute, Mobile, AL; Cancer Center of the Carolinas, Greenville, SC; and Duke University Medical Center, Durham, NC
| | - David McCune
- From the Mary Crowley Cancer Research Centers; Baylor Sammons Cancer Center; Gradalis Inc; and Texas Oncology Physicians Association, Dallas; Tyler Cancer Center, Tyler, TX; IDM Pharma Inc, Irvine; Pharmexa-Epimmune, San Diego, CA; Sarah Cannon Cancer Center, Nashville, TN; Madigan Army Medical Center, Tacoma WA; Atlanta Cancer Care, Roswell, GA; University of South Alabama, Mitchell Cancer Institute, Mobile, AL; Cancer Center of the Carolinas, Greenville, SC; and Duke University Medical Center, Durham, NC
| | - Ronald Steis
- From the Mary Crowley Cancer Research Centers; Baylor Sammons Cancer Center; Gradalis Inc; and Texas Oncology Physicians Association, Dallas; Tyler Cancer Center, Tyler, TX; IDM Pharma Inc, Irvine; Pharmexa-Epimmune, San Diego, CA; Sarah Cannon Cancer Center, Nashville, TN; Madigan Army Medical Center, Tacoma WA; Atlanta Cancer Care, Roswell, GA; University of South Alabama, Mitchell Cancer Institute, Mobile, AL; Cancer Center of the Carolinas, Greenville, SC; and Duke University Medical Center, Durham, NC
| | - Hung Khong
- From the Mary Crowley Cancer Research Centers; Baylor Sammons Cancer Center; Gradalis Inc; and Texas Oncology Physicians Association, Dallas; Tyler Cancer Center, Tyler, TX; IDM Pharma Inc, Irvine; Pharmexa-Epimmune, San Diego, CA; Sarah Cannon Cancer Center, Nashville, TN; Madigan Army Medical Center, Tacoma WA; Atlanta Cancer Care, Roswell, GA; University of South Alabama, Mitchell Cancer Institute, Mobile, AL; Cancer Center of the Carolinas, Greenville, SC; and Duke University Medical Center, Durham, NC
| | - Donald Richards
- From the Mary Crowley Cancer Research Centers; Baylor Sammons Cancer Center; Gradalis Inc; and Texas Oncology Physicians Association, Dallas; Tyler Cancer Center, Tyler, TX; IDM Pharma Inc, Irvine; Pharmexa-Epimmune, San Diego, CA; Sarah Cannon Cancer Center, Nashville, TN; Madigan Army Medical Center, Tacoma WA; Atlanta Cancer Care, Roswell, GA; University of South Alabama, Mitchell Cancer Institute, Mobile, AL; Cancer Center of the Carolinas, Greenville, SC; and Duke University Medical Center, Durham, NC
| | - Joe Stephenson
- From the Mary Crowley Cancer Research Centers; Baylor Sammons Cancer Center; Gradalis Inc; and Texas Oncology Physicians Association, Dallas; Tyler Cancer Center, Tyler, TX; IDM Pharma Inc, Irvine; Pharmexa-Epimmune, San Diego, CA; Sarah Cannon Cancer Center, Nashville, TN; Madigan Army Medical Center, Tacoma WA; Atlanta Cancer Care, Roswell, GA; University of South Alabama, Mitchell Cancer Institute, Mobile, AL; Cancer Center of the Carolinas, Greenville, SC; and Duke University Medical Center, Durham, NC
| | - Prasanthi Ganesa
- From the Mary Crowley Cancer Research Centers; Baylor Sammons Cancer Center; Gradalis Inc; and Texas Oncology Physicians Association, Dallas; Tyler Cancer Center, Tyler, TX; IDM Pharma Inc, Irvine; Pharmexa-Epimmune, San Diego, CA; Sarah Cannon Cancer Center, Nashville, TN; Madigan Army Medical Center, Tacoma WA; Atlanta Cancer Care, Roswell, GA; University of South Alabama, Mitchell Cancer Institute, Mobile, AL; Cancer Center of the Carolinas, Greenville, SC; and Duke University Medical Center, Durham, NC
| | - Jackie Nemunaitis
- From the Mary Crowley Cancer Research Centers; Baylor Sammons Cancer Center; Gradalis Inc; and Texas Oncology Physicians Association, Dallas; Tyler Cancer Center, Tyler, TX; IDM Pharma Inc, Irvine; Pharmexa-Epimmune, San Diego, CA; Sarah Cannon Cancer Center, Nashville, TN; Madigan Army Medical Center, Tacoma WA; Atlanta Cancer Care, Roswell, GA; University of South Alabama, Mitchell Cancer Institute, Mobile, AL; Cancer Center of the Carolinas, Greenville, SC; and Duke University Medical Center, Durham, NC
| | - Glenn Ishioka
- From the Mary Crowley Cancer Research Centers; Baylor Sammons Cancer Center; Gradalis Inc; and Texas Oncology Physicians Association, Dallas; Tyler Cancer Center, Tyler, TX; IDM Pharma Inc, Irvine; Pharmexa-Epimmune, San Diego, CA; Sarah Cannon Cancer Center, Nashville, TN; Madigan Army Medical Center, Tacoma WA; Atlanta Cancer Care, Roswell, GA; University of South Alabama, Mitchell Cancer Institute, Mobile, AL; Cancer Center of the Carolinas, Greenville, SC; and Duke University Medical Center, Durham, NC
| | - Beena Pappen
- From the Mary Crowley Cancer Research Centers; Baylor Sammons Cancer Center; Gradalis Inc; and Texas Oncology Physicians Association, Dallas; Tyler Cancer Center, Tyler, TX; IDM Pharma Inc, Irvine; Pharmexa-Epimmune, San Diego, CA; Sarah Cannon Cancer Center, Nashville, TN; Madigan Army Medical Center, Tacoma WA; Atlanta Cancer Care, Roswell, GA; University of South Alabama, Mitchell Cancer Institute, Mobile, AL; Cancer Center of the Carolinas, Greenville, SC; and Duke University Medical Center, Durham, NC
| | - Michael Nemunaitis
- From the Mary Crowley Cancer Research Centers; Baylor Sammons Cancer Center; Gradalis Inc; and Texas Oncology Physicians Association, Dallas; Tyler Cancer Center, Tyler, TX; IDM Pharma Inc, Irvine; Pharmexa-Epimmune, San Diego, CA; Sarah Cannon Cancer Center, Nashville, TN; Madigan Army Medical Center, Tacoma WA; Atlanta Cancer Care, Roswell, GA; University of South Alabama, Mitchell Cancer Institute, Mobile, AL; Cancer Center of the Carolinas, Greenville, SC; and Duke University Medical Center, Durham, NC
| | - Michael Morse
- From the Mary Crowley Cancer Research Centers; Baylor Sammons Cancer Center; Gradalis Inc; and Texas Oncology Physicians Association, Dallas; Tyler Cancer Center, Tyler, TX; IDM Pharma Inc, Irvine; Pharmexa-Epimmune, San Diego, CA; Sarah Cannon Cancer Center, Nashville, TN; Madigan Army Medical Center, Tacoma WA; Atlanta Cancer Care, Roswell, GA; University of South Alabama, Mitchell Cancer Institute, Mobile, AL; Cancer Center of the Carolinas, Greenville, SC; and Duke University Medical Center, Durham, NC
| | - Bonnie Mills
- From the Mary Crowley Cancer Research Centers; Baylor Sammons Cancer Center; Gradalis Inc; and Texas Oncology Physicians Association, Dallas; Tyler Cancer Center, Tyler, TX; IDM Pharma Inc, Irvine; Pharmexa-Epimmune, San Diego, CA; Sarah Cannon Cancer Center, Nashville, TN; Madigan Army Medical Center, Tacoma WA; Atlanta Cancer Care, Roswell, GA; University of South Alabama, Mitchell Cancer Institute, Mobile, AL; Cancer Center of the Carolinas, Greenville, SC; and Duke University Medical Center, Durham, NC
| | - Phillip B. Maples
- From the Mary Crowley Cancer Research Centers; Baylor Sammons Cancer Center; Gradalis Inc; and Texas Oncology Physicians Association, Dallas; Tyler Cancer Center, Tyler, TX; IDM Pharma Inc, Irvine; Pharmexa-Epimmune, San Diego, CA; Sarah Cannon Cancer Center, Nashville, TN; Madigan Army Medical Center, Tacoma WA; Atlanta Cancer Care, Roswell, GA; University of South Alabama, Mitchell Cancer Institute, Mobile, AL; Cancer Center of the Carolinas, Greenville, SC; and Duke University Medical Center, Durham, NC
| | - Jeffrey Sherman
- From the Mary Crowley Cancer Research Centers; Baylor Sammons Cancer Center; Gradalis Inc; and Texas Oncology Physicians Association, Dallas; Tyler Cancer Center, Tyler, TX; IDM Pharma Inc, Irvine; Pharmexa-Epimmune, San Diego, CA; Sarah Cannon Cancer Center, Nashville, TN; Madigan Army Medical Center, Tacoma WA; Atlanta Cancer Care, Roswell, GA; University of South Alabama, Mitchell Cancer Institute, Mobile, AL; Cancer Center of the Carolinas, Greenville, SC; and Duke University Medical Center, Durham, NC
| | - John J. Nemunaitis
- From the Mary Crowley Cancer Research Centers; Baylor Sammons Cancer Center; Gradalis Inc; and Texas Oncology Physicians Association, Dallas; Tyler Cancer Center, Tyler, TX; IDM Pharma Inc, Irvine; Pharmexa-Epimmune, San Diego, CA; Sarah Cannon Cancer Center, Nashville, TN; Madigan Army Medical Center, Tacoma WA; Atlanta Cancer Care, Roswell, GA; University of South Alabama, Mitchell Cancer Institute, Mobile, AL; Cancer Center of the Carolinas, Greenville, SC; and Duke University Medical Center, Durham, NC
| |
Collapse
|
40
|
Lauwen MM, Zwaveling S, de Quartel L, Ferreira Mota SC, Grashorn JAC, Melief CJM, van der Burg SH, Offringa R. Self-tolerance does not restrict the CD4+ T-helper response against the p53 tumor antigen. Cancer Res 2008; 68:893-900. [PMID: 18245492 DOI: 10.1158/0008-5472.can-07-3166] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Tumorigenesis is frequently associated with mutation and overexpression of p53, which makes it an attractive target antigen for T cell-mediated immunotherapy of cancer. However, the magnitude and breadth of the p53-specific T-cell repertoire may be restricted due to the ubiquitous expression of wild-type p53 in normal somatic tissues. In view of the importance of the CD4+ T-helper cell responses in effective antitumor immunity, we have analyzed and compared the p53-specific reactivity of this T cell subset in p53+/+ and p53-/- C57Bl/6 mice. This response was found to be directed against the same three immunodominant epitopes in both mouse types. Fine-specificity, magnitude, and avidity were not affected by self-tolerance. Immunization of p53-/- and p53+/+ mice with synthetic peptide vaccines comprising the identified epitopes induced equal levels of Th1 immunity. Our findings imply that the p53-specific CD4+ T-cell repertoire is not restricted by self-tolerance and is fully available for the targeting of cancer.
Collapse
Affiliation(s)
- Marjolein M Lauwen
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, the Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Bos R, van Duikeren S, van Hall T, Lauwen MM, Parrington M, Berinstein NL, McNeil B, Melief CJM, Verbeek JS, van der Burg SH, Offringa R. Characterization of antigen-specific immune responses induced by canarypox virus vaccines. THE JOURNAL OF IMMUNOLOGY 2007; 179:6115-22. [PMID: 17947686 DOI: 10.4049/jimmunol.179.9.6115] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Avipoxvirus-based vectors, such as recombinant canarypox virus ALVAC, are studied extensively as delivery vehicles for vaccines against cancer and infectious diseases. Effective use of such vaccines is expected to benefit from proper understanding of the interaction between these viral vectors and the host immune system. We performed preclinical vaccination experiments in a murine tumor model to analyze the immunogenic properties of an ALVAC-based vaccine against carcinoembryonic Ag (ALVAC-CEA), a tumor-associated autoantigen commonly overexpressed in colorectal cancers. The protective CEA-specific immunity induced by this vaccine consisted of CD4(+) T cell responses with a mixed Th1/Th2 cytokine profile that were accompanied by potent humoral responses, but not by CEA-specific CD8(+) CTL immunity. In contrast, protective immunity induced by a CEA-specific DNA vaccine (DNA-CEA) consisted of Th1 and CTL responses. Modification of the ALVAC-CEA vaccine through coinjection of DNA-CEA, admixture with CpG oligodeoxynucleotides, or supplementation with additional transgenes encoding a triad of costimulatory molecules (TRICOM) did not result in induction of CEA-specific CTL responses. Even though these results suggested that ALVAC does not elicit Ag-specific CTLs, immunization with ALVAC vaccines against other Ags efficiently induced CTL responses. Our data show that the capacity of ALVAC vaccines to elicit CTL immunity against transgene-encoded Ags critically depends on the presence of highly immunogenic CTL epitopes in these Ags. This consideration needs to be taken into account with respect to the design and evaluation of vaccination strategies that use ALVAC-based vaccine.
Collapse
Affiliation(s)
- Rinke Bos
- Department of Immunohematology and Blood Transfusion, Tumor Immunology Group, Leiden University Medical Center, Leiden, The Netherlands
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Weinhold M, Sommermeyer D, Uckert W, Blankenstein T. Dual T cell receptor expressing CD8+ T cells with tumor- and self-specificity can inhibit tumor growth without causing severe autoimmunity. THE JOURNAL OF IMMUNOLOGY 2007; 179:5534-42. [PMID: 17911640 DOI: 10.4049/jimmunol.179.8.5534] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The engineering of Ag-specific T cells by expression of TCR genes is a convenient method for adoptive T cell immunotherapy. A potential problem is the TCR gene transfer into self-reactive T cells that survived tolerance mechanisms. We have developed an experimental system with T cells that express two TCRs with defined Ag-specificities, one recognizing a tumor-specific Ag (LCMV-gp(33)), the other recognizing a self-Ag in the pancreas (OVA). By using tumor cells expressing high and low amounts of Ag and mice expressing high and low levels of self-Ag in the pancreas (RIP-OVA-Hi and RIP-OVA-Lo), we show that 1) tumor rejection requires high amount of tumor Ag, 2) severe autoimmunity requires high amount of self-Ag, and 3) if Ag expression on tumor cells is sufficient and low in the pancreas, successful adoptive T cell therapy can be obtained in the absence of severe autoimmunity. These results are shown with T cells from dual TCR transgenic mice or T cells that were redirected by TCR gene transfer. Our data demonstrate that the approach of adoptively transferring TCR redirected T cells can be effective without severe side effects, even when high numbers of T cells with self-reactivity were transferred.
Collapse
MESH Headings
- Animals
- Antigens, Neoplasm/biosynthesis
- Antigens, Neoplasm/physiology
- Autoantigens/physiology
- Autoimmune Diseases/immunology
- Autoimmune Diseases/metabolism
- Autoimmune Diseases/prevention & control
- CD8-Positive T-Lymphocytes/classification
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- Cell Line, Tumor
- Chickens
- Epitopes, T-Lymphocyte/physiology
- Growth Inhibitors/physiology
- Melanoma, Experimental/immunology
- Melanoma, Experimental/metabolism
- Melanoma, Experimental/prevention & control
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Receptors, Antigen, T-Cell/biosynthesis
- Receptors, Antigen, T-Cell/genetics
Collapse
Affiliation(s)
- Monika Weinhold
- Institute of Immunology, Charité, Hindenburgdamm 30, Berlin, Germany
| | | | | | | |
Collapse
|
43
|
Lambeck A, Leffers N, Hoogeboom BN, Sluiter W, Hamming I, Klip H, ten Hoor K, Esajas M, van Oven M, Drijfhout JW, Platteel I, Offringa R, Hollema H, Melief K, van der Burg S, van der Zee A, Daemen T, Nijman H. P53-specific T cell responses in patients with malignant and benign ovarian tumors: implications for p53 based immunotherapy. Int J Cancer 2007; 121:606-14. [PMID: 17415711 DOI: 10.1002/ijc.22710] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Despite intensive treatment, 70% of the ovarian cancer patients will develop recurrent disease, emphasizing the need for new approaches such as immunotherapy. A promising antigenic target for immunotherapy in ovarian cancer is the frequently overexpressed p53 protein. The aim of the study was to evaluate the nature and magnitude of the baseline anti-p53 immune response in ovarian cancer patients. P53-specific T cell responses were detected in both half of the ovarian cancer patients as in the group of control subjects, consisting of women with benign ovarian tumors and healthy controls. Importantly, while in the control group p53-specific immunity was detected among the CD45RA(+) naïve subset of T cells only, the p53-specific T-cell responses in ovarian cancer patients were also present in the CD45RO(+) memory T-cell subset, suggesting that in the cancer patients sufficient amounts of cancer-derived p53 was presented to induce the formation of a p53-specific memory T-cell response. Further characterization of the p53-specific memory T-cell responses revealed that in addition to the type 1 cytokine IFN-gamma also the type 2 cytokines IL-4 and IL-5, as well as the immunosuppressive cytokine IL-10 were produced. Notably, p53-specific T cells were not only detected in the peripheral blood, but also among tumor infiltrating lymphocytes and in tumor-draining lymph nodes. In conclusion, the existence of a weak mixed T-helper type 1 and 2 p53-specific T-cell repertoire supports the rationale of using p53 long peptides in vaccination strategies aiming at the induction of p53-specific Th1/CTL immunity.
Collapse
Affiliation(s)
- Annechien Lambeck
- Department of Gynaecology, University Medical Center Groningen, University of Groningen, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Song GY, Gibson G, Haq W, Huang ECC, Srivasta T, Hollstein M, Daftarian P, Wang Z, Diamond D, Ellenhorn JDI. An MVA vaccine overcomes tolerance to human p53 in mice and humans. Cancer Immunol Immunother 2007; 56:1193-205. [PMID: 17219151 PMCID: PMC11030254 DOI: 10.1007/s00262-006-0270-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2006] [Accepted: 11/28/2006] [Indexed: 10/23/2022]
Abstract
BACKGROUND The cellular regulatory protein p53 is overexpressed by almost 50% of all malignancies making it an attractive target for a vaccine approach to cancer. A number of immunotherapy approaches targeting p53 have been evaluated successfully in murine models, but translation of these preclinical findings to the clinic has been unsuccessful. Prior studies in our laboratory employing murine models demonstrated that a modified vaccinia virus Ankara (MVA) vaccine expressing murine p53 could stimulate p53 specific immunity. Systemic administration of the MVA vaccine was able to effect the rejection of established tumors. To better understand the immunologic mechanisms that underlie the vaccine function of human p53, we utilized a murine model in which the murine germ line copy of p53 was replaced with a modified human one. These mice, referred to as Hupki, were evaluated as a tolerant model to explore the capacity of MVA expressing human p53 to overcome tolerance and reject human p53-expressing tumors. RESULTS MVAp53 immunization of Hupki mice resulted in the generation of p53-specific CD8(+) T cells and the rejection of a highly aggressive murine mammary carcinoma cell line 4T1(H-2d) transfected with human p53 (4T1p53). An immunologic correlate of tumor protection was evaluated utilizing an overlapping peptide library spanning the full length of human p53. This reagent was also used in combination with MVAp53 to stimulate p53-specific CD8(+) T cell responses in cancer patients. CONCLUSION These studies demonstrate the potential of MVAp53 to overcome tolerance to p53 for cancer immunotherapy.
Collapse
MESH Headings
- Animals
- Antigens, Neoplasm/chemistry
- Antigens, Neoplasm/genetics
- Antigens, Neoplasm/immunology
- CD8-Positive T-Lymphocytes/immunology
- Cancer Vaccines/immunology
- Cancer Vaccines/pharmacology
- Cancer Vaccines/therapeutic use
- Carcinoma, Squamous Cell/immunology
- Cells, Cultured/immunology
- Cytotoxicity, Immunologic
- Drug Screening Assays, Antitumor
- Female
- Head and Neck Neoplasms/immunology
- Humans
- Immune Tolerance
- Immunotherapy, Active
- Interferon-gamma/metabolism
- Lymphocyte Activation
- Lymphocytes, Tumor-Infiltrating/immunology
- Mammary Neoplasms, Experimental/immunology
- Mammary Neoplasms, Experimental/therapy
- Mice
- Mice, Inbred BALB C
- Neoplasm Transplantation
- Peptide Fragments/chemical synthesis
- Peptide Fragments/immunology
- Transfection
- Tumor Suppressor Protein p53/chemistry
- Tumor Suppressor Protein p53/genetics
- Tumor Suppressor Protein p53/immunology
Collapse
Affiliation(s)
- Guang-Yun Song
- Department of General and Oncologic Surgery, City of Hope National Medical Center, Duarte, CA USA
| | - Glen Gibson
- Department of General and Oncologic Surgery, City of Hope National Medical Center, Duarte, CA USA
| | - Wahajul Haq
- Laboratory of Vaccine Research, City of Hope National Medical Center, Duarte, CA USA
| | - Eric C. C. Huang
- Laboratory of Vaccine Research, City of Hope National Medical Center, Duarte, CA USA
| | - Tumul Srivasta
- Laboratory of Vaccine Research, City of Hope National Medical Center, Duarte, CA USA
| | - Monica Hollstein
- Division of Genetic Alterations in Carcinogenesis, German Cancer Research Center, Heidelberg, Germany
| | - Pirouz Daftarian
- Laboratory of Vaccine Research, City of Hope National Medical Center, Duarte, CA USA
| | - Zhongde Wang
- Laboratory of Vaccine Research, City of Hope National Medical Center, Duarte, CA USA
| | - Don Diamond
- Laboratory of Vaccine Research, City of Hope National Medical Center, Duarte, CA USA
| | - Joshua D. I. Ellenhorn
- Department of General and Oncologic Surgery, City of Hope National Medical Center, Duarte, CA USA
- Division of Surgery, City of Hope Comprehensive Cancer Center, 1500 E. Duarte Road, Duarte, CA 91010 USA
| |
Collapse
|
45
|
Mansour M, Pohajdak B, Kast WM, Fuentes-Ortega A, Korets-Smith E, Weir GM, Brown RG, Daftarian P. Therapy of established B16-F10 melanoma tumors by a single vaccination of CTL/T helper peptides in VacciMax. J Transl Med 2007; 5:20. [PMID: 17451606 PMCID: PMC1867806 DOI: 10.1186/1479-5876-5-20] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2007] [Accepted: 04/23/2007] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Melanoma tumors are known to express antigens that usually induce weak immune responses of short duration. Expression of both tumor-associated antigens p53 and TRP2 by melanoma cells raises the possibility of simultaneously targeting more than one antigen in a therapeutic vaccine. In this report, we show that VacciMax (VM), a novel liposome-based vaccine delivery platform, can increase the immunogenicity of melanoma associated antigens, resulting in tumor elimination. METHODS C57BL/6 mice bearing B16-F10 melanoma tumors were vaccinated subcutaneously 6 days post tumor implantation with a mixture of synthetic peptides (modified p53: 232-240, TRP-2: 181-188 and PADRE) and CpG. Tumor growth was monitored and antigen-specific splenocyte responses were assayed by ELISPOT. RESULTS Vaccine formulated in VM increased the number of both TRP2- and p53-specific IFN-gamma producing splenocytes following a single vaccination. Vaccine formulated without VM resulted only in enhanced IFN-gamma producing splenocytes to one CTL epitopes (TRP2:180-188), suggesting that VM overcomes antigen dominance and enhances immunogenicity of multiple epitopes. Vaccination of mice bearing 6-day old B16-F10 tumors with both TRP2 and p53-peptides formulated in VM successfully eradicated tumors in all mice. A control vaccine which contained all ingredients except liposomes resulted in eradication of tumors in no more than 20% of mice. CONCLUSION A single administration of VM is capable of inducing an effective CTL response to multiple tumor-associated antigens. The responses generated were able to reject 6-day old B16-F10 tumors.
Collapse
Affiliation(s)
- Marc Mansour
- ImmunoVaccine Technologies Inc., Halifax, NS, Canada
| | - Bill Pohajdak
- ImmunoVaccine Technologies Inc., Halifax, NS, Canada
| | - W Martin Kast
- Dept. of Molecular Microbiology & Immunology and Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, USA
| | | | | | | | | | - Pirouz Daftarian
- ImmunoVaccine Technologies Inc., Halifax, NS, Canada
- Department of Microbiology & Immunology, Dalhousie University, Halifax, NS, Canada
| |
Collapse
|
46
|
Lazoura E, Lodding J, Farrugia W, Ramsland PA, Stevens J, Wilson IA, Pietersz GA, Apostolopoulos V. Enhanced major histocompatibility complex class I binding and immune responses through anchor modification of the non-canonical tumour-associated mucin 1-8 peptide. Immunology 2007; 119:306-16. [PMID: 17067310 PMCID: PMC1819580 DOI: 10.1111/j.1365-2567.2006.02434.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
Abstract
Designing peptide-based vaccines for therapeutic applications in cancer immunotherapy requires detailed knowledge of the interactions between the antigenic peptide and major histocompatibility complex (MHC) in addition to that between the peptide-MHC complex and the T-cell receptor. Past efforts to immunize with high-affinity tumour-associated antigenic peptides have not been very immunogenic, which may be attributed to the lack of T cells to these peptides, having been deleted during thymic development. For this reason, low-to-medium affinity non-canonical peptides represent more suitable candidates. However, in addition to the difficulty in identifying such antigens, peptide binding to MHC, and hence its ability to induce a strong immune response, is limited. Therefore, to enhance binding to MHC and improve immune responses, anchor modifications of non-canonical tumour-associated peptides would be advantageous. In this study, the non-canonical tumour-associated peptide from MUC1, MUC1-8 (SAPDTRPA), was modified at the MHC anchor residues to SAPDFRPL (MUC1-8-5F8L) and showed enhanced binding to H-2Kb and improved immune responses. Furthermore, the crystal structure of MUC1-8-5F8L in complex with H-2Kb was determined and it revealed that binding of the peptide to MHC is similar to that of the canonical peptide OVA8 (SIINFEKL).
Collapse
Affiliation(s)
- Eliada Lazoura
- Burnet Institute at Austin, Immunology and Vaccine LaboratoryHeidelberg, VIC, Australia
| | - Jodie Lodding
- Burnet Institute at Austin, Immunology and Vaccine LaboratoryHeidelberg, VIC, Australia
| | - William Farrugia
- Burnet Institute at Austin, Structural Immunology LaboratoryHeidelberg, VIC, Australia
| | - Paul A Ramsland
- Burnet Institute at Austin, Structural Immunology LaboratoryHeidelberg, VIC, Australia
| | - James Stevens
- Department of Molecular Biology, The Scripps Research InstituteLa Jolla, CA, USA
| | - Ian A Wilson
- Department of Molecular Biology, The Scripps Research InstituteLa Jolla, CA, USA
- Skaggs Institute for Chemical Biology, The Scripps Research InstituteLa Jolla, CA, USA
| | - Geoffrey A Pietersz
- Burnet Institute at Austin, Bio-Organic and Medicinal Chemistry LaboratoryHeidelberg VIC Australia
| | - Vasso Apostolopoulos
- Burnet Institute at Austin, Immunology and Vaccine LaboratoryHeidelberg, VIC, Australia
| |
Collapse
|
47
|
Ternovoi VV, Curiel DT, Smith BF, Siegal GP. Adenovirus-mediated p53 tumor suppressor gene therapy of osteosarcoma. J Transl Med 2006; 86:748-66. [PMID: 16751779 DOI: 10.1038/labinvest.3700444] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
The clinical outcome for osteosarcoma (OS) remains discouraging despite efforts to optimize treatment using conventional modalities including surgery, radiotherapy and chemotherapy. Novel therapeutic approaches based on our expanding understanding of the mechanisms of tumor cell killing have the potential to alter this situation. Tumor suppressor gene therapy aims to restore the function of a tumor suppressor gene lost or functionally inactivated in cancer cells. One such molecule, the p53 tumor suppressor gene plays a critical role in safeguarding the integrity of the genome and preventing tumorigenesis. Introduction of wild-type (wt) p53 into transformed cells has been shown to be lethal for most cancer cells in vitro, but clinical trials of p53 gene replacement have had limited success. Analysis of these clinical trials highlighted the insufficient efficacy of current vectors and low proapoptotic activity of wt p53 as a single agent in vivo. In this review, a contemporary summarization of the current status of adenovirus-mediated p53 gene therapy of OS is presented. Advancement in our understanding of p53 tumor suppressor activity, the molecular biology of chemoresistant OS, and recent advances in tumor targeting with adenoviral vectors are also addressed. Based on these parameters, prospects for future investigations are proposed.
Collapse
Affiliation(s)
- Vladimir V Ternovoi
- Division of Human Gene Therapy, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | | | | | | |
Collapse
|
48
|
Voss RH, Kuball J, Engel R, Guillaume P, Romero P, Huber C, Theobald M. Redirection of T cells by delivering a transgenic mouse-derived MDM2 tumor antigen-specific TCR and its humanized derivative is governed by the CD8 coreceptor and affects natural human TCR expression. Immunol Res 2006; 34:67-87. [PMID: 16720899 DOI: 10.1385/ir:34:1:67] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/1999] [Revised: 11/30/1999] [Accepted: 11/30/1999] [Indexed: 11/11/2022]
Abstract
Retroviral transfer of T cell antigen receptor (TCR) genes selected by circumventing tolerance to broad tumor- and leukemia-associated antigens in human leukocyte antigen (HLA)-A*0201 (A2.1) transgenic (Tg) mice allows the therapeutic reprogramming of human T lymphocytes. Using a human CD8 x A2.1/Kb mouse derived TCR specific for natural peptide-A2.1 (pA2.1) complexes comprising residues 81-88 of the human homolog of the murine double-minute 2 oncoprotein, MDM2(81-88), we found that the heterodimeric CD8 alpha beta coreceptor, but not normally expressed homodimeric CD8 alpha alpha, is required for tetramer binding and functional redirection of TCR- transduced human T cells. CD8+T cells that received a humanized derivative of the MDM2 TCR bound pA2.1 tetramers only in the presence of an anti-human-CD8 anti-body and required more peptide than wild-type (WT) MDM2 TCR+T cells to mount equivalent cytotoxicity. They were, however, sufficiently effective in recognizing malignant targets including fresh leukemia cells. Most efficient expression of transduced TCR in human T lymphocytes was governed by mouse as compared to human constant (C) alphabeta domains, as demonstrated with partially humanized and murinized TCR of primary mouse and human origin, respectively. We further observed a reciprocal relationship between the level of Tg WT mouse relative to natural human TCR expression, resulting in T cells with decreased normal human cell surface TCR. In contrast, natural human TCR display remained unaffected after delivery of the humanized MDM2 TCR. These results provide important insights into the molecular basis of TCR gene therapy of malignant disease.
Collapse
Affiliation(s)
- Ralf-Holger Voss
- Department of Hematology and Oncology, Johannes Gutenberg-University, Mainz, Germany
| | | | | | | | | | | | | |
Collapse
|
49
|
Wysocki PJ, Kwiatkowska EP, Kazimierczak U, Suchorska W, Kowalczyk DW, Mackiewicz A. Captopril, an Angiotensin-Converting Enzyme Inhibitor, Promotes Growth of Immunogenic Tumors in Mice. Clin Cancer Res 2006; 12:4095-102. [PMID: 16818710 DOI: 10.1158/1078-0432.ccr-05-2489] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Antitumor potential of angiotensin-converting enzyme inhibitors has been shown in different preclinical settings, which always involved immunocompromised organisms or nonimmunogenic tumor models. In our study, we wanted to evaluate the effect of captopril on growth of immunogenic tumors in immunocompetent animals. EXPERIMENTAL DESIGN We used different murine tumor models to evaluate the effect of captopril on tumor take and survival of tumor-bearing immunocompetent and immunocompromised mice. We used an orthotopic renal cell cancer model and highly immunogenic tumor model, which were based on kidney subcapsular injection of RenCa cells or s.c. injection of MethA cells, respectively. To show the influence of captopril on antigen-specific immune responses, we have used two model antigens (green fluorescent protein and beta-galactosidase). RESULTS Captopril decreased survival of RenCa-bearing, immunocompetent mice in a dose-dependent manner and in adjuvant setting. In nephrectomized mice, captopril shortened their survival. Captopril promoted formation of immunogenic MethA sarcoma tumors but had no effect on nonimmunogenic melanoma cells (B78-H1). Treatment of immunocompromised mice bearing MethA tumors or RenCa kidney tumors with captopril did not affect tumor formation nor survival, respectively. Captopril-treated mice immunized with AdLacZ or AdGFP vectors did not generate or generated decreased numbers of antigen-specific CD8+ T cells, respectively. However, they showed B-cell responses represented by infiltration of MethA tumors with activated B cells and dramatically increased serum level of beta-galactosidase-specific antibodies. CONCLUSIONS Our results show a novel role of captopril in tumor biology and the tumor-promoting properties of captopril seem to be associated with its immunomodulatory potential.
Collapse
MESH Headings
- Angiotensin-Converting Enzyme Inhibitors/pharmacology
- Animals
- CD8-Positive T-Lymphocytes/drug effects
- CD8-Positive T-Lymphocytes/immunology
- Captopril/pharmacology
- Cell Line, Tumor
- Cell Survival/drug effects
- Disease Models, Animal
- Dose-Response Relationship, Drug
- Drug Screening Assays, Antitumor
- Female
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, SCID
- Sarcoma, Experimental/immunology
- Sarcoma, Experimental/pathology
- Transplantation, Heterologous
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Piotr J Wysocki
- Department of Cancer Immunology, University of Medical Sciences at GreatPoland Cancer Center, Poznan, Poland.
| | | | | | | | | | | |
Collapse
|
50
|
Antonia SJ, Mirza N, Fricke I, Chiappori A, Thompson P, Williams N, Bepler G, Simon G, Janssen W, Lee JH, Menander K, Chada S, Gabrilovich DI. Combination of p53 cancer vaccine with chemotherapy in patients with extensive stage small cell lung cancer. Clin Cancer Res 2006; 12:878-87. [PMID: 16467102 DOI: 10.1158/1078-0432.ccr-05-2013] [Citation(s) in RCA: 307] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
PURPOSE The initial goal of this study was to test the immunologic and clinical effects of a new cancer vaccine consisting of dendritic cells (DC) transduced with the full-length wild-type p53 gene delivered via an adenoviral vector in patients with extensive stage small cell lung cancer. EXPERIMENTAL DESIGN Twenty-nine patients with extensive stage small cell lung cancer were vaccinated repeatedly at 2-week intervals. Most of the patients received three immunizations. p53-specific responses were evaluated, and phenotype and function of T cells, DCs, and immature myeloid cells were analyzed and correlated with antigen-specific immune responses. Objective clinical response to vaccination as well as subsequent chemotherapy was evaluated. RESULTS p53-specific T cell responses to vaccination were observed in 57.1% of patients. Immunologic responses to vaccination were positively associated with a moderate increase in the titer of antiadenovirus antibodies, and negatively with an accumulation of immature myeloid cells. One patient showed a clinical response to vaccination whereas most of the patients had disease progression. However, we observed a high rate of objective clinical responses to chemotherapy (61.9%) that immediately followed vaccination. Clinical response to subsequent chemotherapy was closely associated with induction of immunologic response to vaccination. CONCLUSIONS This study provides clinical support for an emerging paradigm in cancer immunotherapy, wherein optimal use of vaccination might be more effective, not as a separate modality, but in direct combination with chemotherapy.
Collapse
Affiliation(s)
- Scott J Antonia
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida 33612, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|