1
|
Birmpilis AI, Karachaliou CE, Samara P, Ioannou K, Selemenakis P, Kostopoulos IV, Kavrochorianou N, Kalbacher H, Livaniou E, Haralambous S, Kotsinas A, Farzaneh F, Trougakos IP, Voelter W, Dimopoulos MA, Bamias A, Tsitsilonis O. Antitumor Reactive T-Cell Responses Are Enhanced In Vivo by DAMP Prothymosin Alpha and Its C-Terminal Decapeptide. Cancers (Basel) 2019; 11:cancers11111764. [PMID: 31717548 PMCID: PMC6896021 DOI: 10.3390/cancers11111764] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 11/04/2019] [Accepted: 11/06/2019] [Indexed: 12/13/2022] Open
Abstract
Prothymosin α (proTα) and its C-terminal decapeptide proTα(100-109) were shown to pleiotropically enhance innate and adaptive immune responses. Their activities have been broadly studied in vitro, focusing primarily on the restoration of the deficient immunoreactivity of cancer patients' leukocytes. Previously, we showed that proTα and proTα(100-109) act as danger-associated molecular patterns (DAMPs), ligate Toll-like receptor-4, signal through TRIF- and MyD88-dependent pathways, promote the maturation of dendritic cells and elicit T-helper type 1 (Th1) immune responses in vitro, leading to the optimal priming of tumor antigen-reactive T-cell functions. Herein, we assessed their activity in a preclinical melanoma model. Immunocompetent mice bearing B16.F1 tumors were treated with two cycles of proTα or proTα(100-109) together with a B16.F1-derived peptide vaccine. Coadministration of proTα or proTα(100-109) and the peptide vaccine suppressed melanoma-cell proliferation, as evidenced by reduced tumor-growth rates. Higher melanoma infiltration by CD3+ T cells was observed, whereas ex vivo analysis of mouse total spleen cells verified the in vivo induction of melanoma-reactive cytotoxic responses. Additionally, increased levels of proinflammatory and Th1-type cytokines were detected in mouse serum. We propose that, in the presence of tumor antigens, DAMPs proTα and proTα(100-109) induce Th1-biased immune responses in vivo. Their adjuvant ability to orchestrate antitumor immunoreactivities can eventually be exploited therapeutically in humans.
Collapse
Affiliation(s)
- Anastasios I. Birmpilis
- Department of Biology, National and Kapodistrian University of Athens, 15784 Athens, Greece; (A.I.B.); (P.S.); (K.I.); (I.V.K.); (I.P.T.)
| | - Chrysoula-Evangelia Karachaliou
- Institute of Nuclear and Radiological Sciences and Technology, Energy and Safety, NCSR “Demokritos”, Agia Paraskevi, 15310 Athens, Greece; (C.-E.K.); (E.L.)
| | - Pinelopi Samara
- Department of Biology, National and Kapodistrian University of Athens, 15784 Athens, Greece; (A.I.B.); (P.S.); (K.I.); (I.V.K.); (I.P.T.)
| | - Kyriaki Ioannou
- Department of Biology, National and Kapodistrian University of Athens, 15784 Athens, Greece; (A.I.B.); (P.S.); (K.I.); (I.V.K.); (I.P.T.)
- King’s College London, Rayne Institute, 123 Coldharbour Lane, SE5 9NU London, UK;
| | - Platon Selemenakis
- Molecular Carcinogenesis Group, Department of Histology and Embryology, School of Medicine, National and Kapodistrian University of Athens, 75 Mikras Asias Str, 11527 Athens, Greece; (P.S.); (A.K.)
| | - Ioannis V. Kostopoulos
- Department of Biology, National and Kapodistrian University of Athens, 15784 Athens, Greece; (A.I.B.); (P.S.); (K.I.); (I.V.K.); (I.P.T.)
| | - Nadia Kavrochorianou
- Inflammation Research Group, Transgenic Technology Laboratory, Hellenic Pasteur Institute, 127 Vasilissis Sofias Avenue, 11521 Athens, Greece; (N.K.); (S.H.)
| | - Hubert Kalbacher
- Interfaculty Institute of Biochemistry, University of Tübingen, 72076 Tübingen. Germany; (H.K.); (W.V.)
| | - Evangelia Livaniou
- Institute of Nuclear and Radiological Sciences and Technology, Energy and Safety, NCSR “Demokritos”, Agia Paraskevi, 15310 Athens, Greece; (C.-E.K.); (E.L.)
| | - Sylva Haralambous
- Inflammation Research Group, Transgenic Technology Laboratory, Hellenic Pasteur Institute, 127 Vasilissis Sofias Avenue, 11521 Athens, Greece; (N.K.); (S.H.)
| | - Athanasios Kotsinas
- Molecular Carcinogenesis Group, Department of Histology and Embryology, School of Medicine, National and Kapodistrian University of Athens, 75 Mikras Asias Str, 11527 Athens, Greece; (P.S.); (A.K.)
| | - Farzin Farzaneh
- King’s College London, Rayne Institute, 123 Coldharbour Lane, SE5 9NU London, UK;
| | - Ioannis P. Trougakos
- Department of Biology, National and Kapodistrian University of Athens, 15784 Athens, Greece; (A.I.B.); (P.S.); (K.I.); (I.V.K.); (I.P.T.)
| | - Wolfgang Voelter
- Interfaculty Institute of Biochemistry, University of Tübingen, 72076 Tübingen. Germany; (H.K.); (W.V.)
| | - Meletios-Athanasios Dimopoulos
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, 11528 Athens, Greece; (M.-A.D.); (A.B.)
| | - Aristotelis Bamias
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, 11528 Athens, Greece; (M.-A.D.); (A.B.)
| | - Ourania Tsitsilonis
- Department of Biology, National and Kapodistrian University of Athens, 15784 Athens, Greece; (A.I.B.); (P.S.); (K.I.); (I.V.K.); (I.P.T.)
- Correspondence: ; Tel.: +30-210-727-4215; Fax: +30-210-727-4635
| |
Collapse
|
2
|
Abstract
Vaccination against cancer-associated antigens has long held the promise of inducting potent antitumor immunity, targeted cytotoxicity while sparing normal tissues, and long-lasting immunologic memory that can provide surveillance against tumor recurrence. Evaluation of vaccination strategies in preclinical brain tumor models has borne out the capacity for the immune system to effectively and safely eradicate established tumors within the central nervous system. Early phase clinical trials have established the feasibility, safety, and immunogenicity of several vaccine platforms, predominantly in patients with glioblastoma. Definitive demonstration of clinical benefit awaits further study, but initial results have been encouraging. With increased understanding of the stimulatory and regulatory pathways that govern immunologic responses and the enhanced capacity to identify novel antigenic targets using genomic interrogation of tumor cells, vaccination platforms for patients with malignant brain tumors are advancing with increasing personalized complexity and integration into combinatorial treatment paradigms.
Collapse
Affiliation(s)
- John H Sampson
- Preston Robert Tisch Brain Tumor Center at Duke, Duke Brain Tumor Immunotherapy Program, Division of Neurosurgery, Department of Surgery, Duke University Medical Center, Durham, North Carolina (J.H.S.); Preston A. Wells, Jr. Center for Brain Tumor Therapy, UF Brain Tumor Immunotherapy Program, Department of Neurosurgery, McKnight Brain Institute, University of Florida, Gainesville, Florida (D.A.M.)
| | - Duane A Mitchell
- Preston Robert Tisch Brain Tumor Center at Duke, Duke Brain Tumor Immunotherapy Program, Division of Neurosurgery, Department of Surgery, Duke University Medical Center, Durham, North Carolina (J.H.S.); Preston A. Wells, Jr. Center for Brain Tumor Therapy, UF Brain Tumor Immunotherapy Program, Department of Neurosurgery, McKnight Brain Institute, University of Florida, Gainesville, Florida (D.A.M.)
| |
Collapse
|
3
|
Taussig D, Williams MA, Wiggins C, Feakins RM, McBride N, Newland AC, Kelsey SM. Clinical Regression and Remission of Primary Refractory Angiocentric Lymphoma Following Autologous Tumour Peptide Antigen-charged Dendritic Cells After High-dose Chemotherapy and Autologous Stem Cell Rescue. ACTA ACUST UNITED AC 2016; 3:277-89. [PMID: 27413880 DOI: 10.1080/10245332.1998.11746400] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
We report on the case of a 34-year old man with a previously refractory high-grade non-Hodgkin's Lymphoma which regressed following dendritic cell based immunotherapy after high-dose chemotherapy. Antigen presenting cells known as dendritic cells were cultured from harvested autologous peripheral blood progenitor cells. The dendritic cells were then exposed to unfractionated tumour peptides derived from a skin biopsy section inflitrated with lymphoma. These charged dendritic cells infused into the patient together with autologous T-lymphocytes after high-dose chemotherapy with peripheral blood stem cell rescue. Although the lymphoma relapsed after the high-dose chemotherapy it regressed again following the dendritic cell infusion. The patient's T-lymphocytes demonstrated in vitro reactivity to tumour peptides whereas the lymphocytes of controls did not. We propose that the lymphoma regression occurred because of a T cell mediated response against the tumour induced by the charged dendritic cells.
Collapse
Affiliation(s)
- D Taussig
- a Department of Haematology , St. Bartholomew's and The Royal London School of Medicine and Dentistry, Queen Mary and Westfield College, University of London , Whitechapel, London E1 2AD
| | - M A Williams
- a Department of Haematology , St. Bartholomew's and The Royal London School of Medicine and Dentistry, Queen Mary and Westfield College, University of London , Whitechapel, London E1 2AD
| | - C Wiggins
- a Department of Haematology , St. Bartholomew's and The Royal London School of Medicine and Dentistry, Queen Mary and Westfield College, University of London , Whitechapel, London E1 2AD
| | - R M Feakins
- b Histopathology , St. Bartholomew's and The Royal London School of Medicine and Dentistry, Queen Mary and Westfield College, University of London , Whitechapel, London E1 2AD
| | - N McBride
- a Department of Haematology , St. Bartholomew's and The Royal London School of Medicine and Dentistry, Queen Mary and Westfield College, University of London , Whitechapel, London E1 2AD
| | - A C Newland
- a Department of Haematology , St. Bartholomew's and The Royal London School of Medicine and Dentistry, Queen Mary and Westfield College, University of London , Whitechapel, London E1 2AD
| | - S M Kelsey
- a Department of Haematology , St. Bartholomew's and The Royal London School of Medicine and Dentistry, Queen Mary and Westfield College, University of London , Whitechapel, London E1 2AD
| |
Collapse
|
4
|
Ioannou K, Cheng KF, Crichlow GV, Birmpilis AI, Lolis EJ, Tsitsilonis OE, Al-Abed Y. ISO-66, a novel inhibitor of macrophage migration, shows efficacy in melanoma and colon cancer models. Int J Oncol 2014; 45:1457-68. [PMID: 25050663 PMCID: PMC4432716 DOI: 10.3892/ijo.2014.2551] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Accepted: 05/14/2014] [Indexed: 01/11/2023] Open
Abstract
Macrophage migration inhibitory factor (MIF) is a pleiotropic pro-inflammatory cytokine, which possesses a contributing role in cancer progression and metastasis and, thus, is now considered a promising anticancer drug target. Many MIF-inactivating strategies have proven successful in delaying cancer growth. Here, we report on the synthesis of ISO-66, a novel, highly stable, small-molecule MIF inhibitor, an analog of ISO-1 with improved characteristics. The MIF:ISO-66 co-crystal structure demonstrated that ISO-66 ligates the tautomerase active site of MIF, which has previously been shown to play an important role in its biological functions. In vitro, ISO-66 enhanced specific and non-specific anticancer immune responses, whereas prolonged administration of ISO-66 in mice with established syngeneic melanoma or colon cancer was non-toxic and resulted in a significant decrease in tumor burden. Subsequent ex vivo analysis of mouse splenocytes revealed that the observed decrease in tumor growth rates was likely mediated by the selective in vivo expansion of antitumor-reactive effector cells induced by ISO-66. Compared to other MIF-inactivating strategies employed in vivo, the anticancer activity of ISO-66 is demonstrated to be of equal or better efficacy. Our findings suggest that targeting MIF, via highly specific and stable compounds, such as ISO-66, may be effective for cancer treatment and stimulation of anticancer immune responses.
Collapse
Affiliation(s)
- Kyriaki Ioannou
- Department of Animal and Human Physiology, Faculty of Biology, University of Athens, Athens 15784, Greece
| | - Kai Fan Cheng
- Center for Molecular Innovation, The Feinstein Institute for Medical Research, Manhasset, NY 11030, USA
| | - Gregg V Crichlow
- Department of Pharmacology, Yale University, New Haven, CT 06510, USA
| | - Anastasios I Birmpilis
- Department of Animal and Human Physiology, Faculty of Biology, University of Athens, Athens 15784, Greece
| | - Elias J Lolis
- Department of Pharmacology, Yale University, New Haven, CT 06510, USA
| | - Ourania E Tsitsilonis
- Department of Animal and Human Physiology, Faculty of Biology, University of Athens, Athens 15784, Greece
| | - Yousef Al-Abed
- Center for Molecular Innovation, The Feinstein Institute for Medical Research, Manhasset, NY 11030, USA
| |
Collapse
|
5
|
Dermime S, Aljurf MD. Current advances, problems and prospects for vaccine-based immunotherapy in follicular non-Hodgkin's lymphoma. Leuk Lymphoma 2009; 46:497-507. [PMID: 16019477 DOI: 10.1080/104281904000025104] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Despite advances in chemotherapy, radiotherapy and combined modality treatment, a significant proportion of non-Hodgkin's lymphomas remain incurable. The disease usually responds well to chemotherapy or radiation, but relapses are observed within months to a few years, with frequent failure of subsequent therapies. High-dose chemotherapy with or without radiation and autologous or allogeneic hematopoietic stem cell transplantation provide higher cure rates and longer remissions in certain patients with aggressive lymphomas. However, the higher treatment-related morbidity and mortality of high-dose chemotherapy has driven a search for new and more tumor-specific treatment modalities, such as immunotherapy. Tumor antigens expressed by B-cell lymphomas, such as the "idiotype antigen", are seen as unique and specific target molecules for direct lymphoma immunotherapy. This review will delineate advances, problems and prospects for approaches to anti-B cell lymphoma immunotherapy where pre-clinical studies and proof of principle have been directly translated to patient care.
Collapse
Affiliation(s)
- Said Dermime
- Tumor Immunology Section, Department of Biological & Medical Research, King Faisal Specialist Hospital & Research Center, Riyadh, 11211, Saudi Arabia.
| | | |
Collapse
|
6
|
|
7
|
Cavazza A, Marini M, Spagnoli GC, Adamina M, Roda LG. Permeability of Phospholipid Vesicles to the Tumor Antigen Epitope gp100280–288. ChemMedChem 2006; 1:816-20. [PMID: 16902935 DOI: 10.1002/cmdc.200600019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Antonella Cavazza
- Dipartimento di Neuroscienze, Università degli Studi di Roma Tor Vergata, Via Montpellier 1, 00133 Roma, Italy
| | | | | | | | | |
Collapse
|
8
|
Chromik J, Schnürer E, Georg Meyer R, Wehler T, Tüting T, Wölfel T, Huber C, Herr W. Proteasome-inhibited dendritic cells demonstrate improved presentation of exogenous synthetic and natural HLA-class I peptide epitopes. J Immunol Methods 2006; 308:77-89. [PMID: 16336973 DOI: 10.1016/j.jim.2005.09.021] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2004] [Revised: 08/08/2005] [Accepted: 09/20/2005] [Indexed: 11/22/2022]
Abstract
The design and successful clinical implementation of cancer vaccines targeting the induction of T-cell mediated immunity is a rapidly evolving field that is hampered by an empirical selection of antigen and adjuvant. In particular, vaccines using defined tumor-associated peptide epitopes elicit only a restricted T-cell repertoire in a minority of patients. In this regard, vaccines comprising the whole spectrum of antigens presented by individual autologous tumors would be advantageous. In an in vitro model, we evaluated the capacity of naturally processed Epstein-Barr virus-transformed B-lymphoblastoid-cell line (LCL)-derived peptides to activate virus-specific CD8+ T cells of seropositive healthy individuals. While bulk peptides obtained by mild acid elution from LCL contained multiple T-cell epitopes, this complex mixture of peptides was poorly immunogenic, even when presented by mature dendritic cells (DC). Pretreatment of DC with proteasome inhibitors strongly enhanced the immunogenicity of single viral synthetic as well as bulk LCL peptides. This was most likely achieved by facilitating the loading of exogenous epitopes onto DC-associated HLA-class I complexes in the face of significant inter-peptide competition for such loading. Our results suggest that proteasome inhibitors may be used to increase the antigenicity of mature DC pulsed with exogenous synthetic or naturally processed peptide epitopes in vaccination trials.
Collapse
Affiliation(s)
- Jörg Chromik
- Department of Medicine III, Hematology and Oncology, Johannes Gutenberg-University of Mainz, Langenbeckstrasse 1, D-55101 Mainz, Germany
| | | | | | | | | | | | | | | |
Collapse
|
9
|
Jones LA, Salgaller ML. Therapeutic potential of dendritic-based vaccines. Expert Opin Investig Drugs 2005; 8:1007-16. [PMID: 15992102 DOI: 10.1517/13543784.8.7.1007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Involving a delicate balance of cell types, the interaction between the immune system and disease or abnormality in the human body is complex. Moreover, the mere presence of antigen and immune cells is necessary, yet insufficient to elicit immune reactivity. In order to elicit an immune response, an antigen in some form must be processed and presented to the immune system. Arguably, the most efficient antigen-presenting cell, the dendritic cell (DC), is the centre of intense investigation. The elicitation or cessation of an immune response is not a simple matter. The body must be able either to up-regulate (e.g., in the case of infectious disease) or down-regulate (e.g., in the case of transplantation) immunity to antigens located anywhere in the body. This sentinel role is capably filled via the distribution of Langerhans cells in the epidermis of the skin, and the migration of DCs throughout the lymphatic and circulatory systems. DCs are potent, as well as efficient: small numbers of cells and low levels of antigen still induce clinically relevant immunity. Lastly, they are capable of tolerance induction to self components by helping to delete self-reactive thymocytes and generating anergy in committed T-cells. Since DCs both initiate and modulate immunity, they are a component of a vast array of vaccines. This review highlights some of the intriguing basic research involving the development of DC-based therapeutics. Furthermore, whenever an area of study has progressed to human treatment, recent and on-going clinical trials are discussed.
Collapse
Affiliation(s)
- L A Jones
- Immunotherapeutics Division, Northwest Biotherapeutics, Inc. 120 Northgate Plaza, Suite 219, Seattle, WA 98125, USA
| | | |
Collapse
|
10
|
Gritzapis AD, Perez SA, Baxevanis CN, Papamichail M. Pooled peptides from HER-2/neu-overexpressing primary ovarian tumours induce CTL with potent antitumour responses in vitro and in vivo. Br J Cancer 2005; 92:72-9. [PMID: 15583693 PMCID: PMC2361747 DOI: 10.1038/sj.bjc.6602259] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Unfractionated peptides (MW: up to 10 kDa), derived from HLA-A2.1 positive (+) HER-2/neu-overexpressing primary tumour cell acid cell extracts (ACE), were successfully used to generate in vitro cytotoxic T lymphocytes (CTL). Primary tumour cells were collected from peritoneal malignant effusions of patients with ovarian cancer. Acid cell extracts-induced CTL specifically lysed in an HLA-A2-restricted manner HER-2/neu+ autologous primary tumour cells as well as HER-2/neu+ tumour cell lines. In addition, adoptive transfer of such CTL significantly prolonged the survival of SCID mice xenografted with HLA-A2.1+, HER-2/neu+ human breast and ovarian tumour cell lines. Acid cell extracts collected from HLA-A2.1+ HER-2/neu negative (-) primary ovarian tumours induced HLA-A2.1-restricted CTL with weak in vitro and in vivo antitumour capacity, suggesting that HER-2/neu peptides within ACE from HER-2/neu-overexpressing primary ovarian tumour cells are immunodominant. The results presented herein serve as a rationale for the initiation of vaccination studies in patients with HER-2/neu-overexpressing ovarian tumours utilising autologous tumour-derived ACE.
Collapse
Affiliation(s)
- A D Gritzapis
- Cancer Immunology and Immunotherapy Center, Saint Savas Cancer Hospital, Athens, Greece
| | - S A Perez
- Cancer Immunology and Immunotherapy Center, Saint Savas Cancer Hospital, Athens, Greece
| | - C N Baxevanis
- Cancer Immunology and Immunotherapy Center, Saint Savas Cancer Hospital, Athens, Greece
- Saint Savas Cancer Hospital, Cancer Immunology and Immunotherapy Center, 171 Alexandras Ave., 11522 Athens, Greece. E-mail:
| | - M Papamichail
- Cancer Immunology and Immunotherapy Center, Saint Savas Cancer Hospital, Athens, Greece
| |
Collapse
|
11
|
Morse MA, Lyerly HK. Dendritic cell-based approaches to cancer immunotherapy. Expert Opin Investig Drugs 2005; 7:1617-27. [PMID: 15991905 DOI: 10.1517/13543784.7.10.1617] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Immunologic approaches to the treatment of malignancies are currently enjoying a resurgence of enthusiasm due to the discovery of tumour-associated antigens and the requirements for stimulating a tumour antigen-specific immune response. The goal of the newer strategies is to stimulate immunity against specific tumour-associated antigens, rather than to broadly, but non-specifically, stimulate the immune system. Since dendritic cells, professional antigen-presenting cells, play a central role in stimulating immune responses in vivo, there is considerable interest in immunising patients with autologous dendritic cells loaded with tumour antigens of interest. Methods for generating large numbers of dendritic cells under clinically-applicable conditions have been developed and it has been shown that they may be loaded with antigen in many different forms including proteins or peptides, RNA or DNA and cellular extracts. Ongoing research is seeking to optimise the purity, antigen loading and maturation of the dendritic cells. Phase I clinical trials have been initiated in order to study the safety and feasibility of immunisations with dendritic cells in humans with various malignancies. Phase II studies will be performed to establish which tumours and clinical scenarios will be most relevant for dendritic cell immunotherapy. Although the commercial applicability of dendritic cell-based immunotherapy has been recognised by the biotechnology industry, commercial availability of dendritic cell vaccines await phase III studies.
Collapse
Affiliation(s)
- M A Morse
- Department of Medicine, Duke University Medical Center, Box 2606, Durham, NC 27710, USA.
| | | |
Collapse
|
12
|
Gritzapis AD, Sotiriadou NN, Papamichail M, Baxevanis CN. Generation of human tumor-specific CTLs in HLA-A2.1-transgenic mice using unfractionated peptides from eluates of human primary breast and ovarian tumors. Cancer Immunol Immunother 2004; 53:1027-40. [PMID: 15164233 PMCID: PMC11033028 DOI: 10.1007/s00262-004-0541-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2003] [Accepted: 03/16/2004] [Indexed: 11/27/2022]
Abstract
HER-2/neu oncoprotein is overexpressed in a variety of human tumors and is associated with malignant transformation and aggressive disease. Due to its overexpression in tumor cells and because it has been shown to be immunogenic, this protein represents an excellent target for T-cell immunotherapy. Peptide extracts derived from primary HLA-A*0201-positive (+) HER-2/neu+ human tumors by acid elution (acid cell extracts (ACEs)) were tested for their capacity to elicit in HLA-A*0201 transgenic mice, cytotoxic T lymphocytes (CTLs) lysing HLA-A*0201+ HER-2/neu+ tumor cells. Injections of ACE in transgenic mice induced CTLs capable of specifically lysing HER-2/neu+ tumor cell lines (also including the original HER-2/neu+ primary tumor cells from which the ACEs were derived) in an HLA-A*0201-restricted fashion. Adoptive transfer of ACE-induced CTLs was sufficient to significantly prolong survival of SCID mice inoculated with HLA-A*0201+ HER-2/ neu+ human tumor cell lines. Cytotoxicity of such ACE-induced CTL lines was directed, at least as detected herein, also against the HER-2/ neu peptides HER-2 (9(369)) and HER-2 (9(435)) demonstrating the immunodominance of these epitopes. HER-2 peptide-specific CTLs generated in the HLA-A*0201-transgenic mice, upon peptide immunization, lysed in vitro HER-2/neu+ human tumor cell lines in an HLA-A*0201-restricted manner and, when adoptively transferred, conferred sufficient protection in SCID mice inoculated with the same human tumor cell lines as above. However, CTLs induced by ACEs displayed enhanced efficacy in the therapy of xenografted SCID mice compared with the HER-2 peptide-specific CTLs (i.e., HER-2 [9(369)] or HER-2 [9(435)]). Even by administering mixtures of CTLs specific for each of these peptides, the prolongation of survival achieved was still inferior compared with that obtained with ACE-induced CTLs. This suggested that additional epitopes may contribute to the immunogenicity of such tumor-derived ACEs. Thus, immunization with ACEs from HER-2/neu+ primary tumor cells appears to be an effective approach to generate multiple and potent CTL-mediated immune responses against HER-2/neu+ tumors expressing the appropriate HLA allele(s). By screening ACE-induced CTL lines with synthetic peptides encompassing the HER-2/neu sequence, it is feasible to identify immunodominant epitopes which may be used in mixtures as vaccines with enhanced efficacy in both the prevention and therapy of HER-2/neu+ malignancies.
Collapse
Affiliation(s)
- Angelos D. Gritzapis
- Cancer Immunology and Immunotherapy Center, Saint Savas Hospital, 171 Alexandras Avenue, 11522 Athens, Greece
| | - Nectaria N. Sotiriadou
- Cancer Immunology and Immunotherapy Center, Saint Savas Hospital, 171 Alexandras Avenue, 11522 Athens, Greece
| | - Michael Papamichail
- Cancer Immunology and Immunotherapy Center, Saint Savas Hospital, 171 Alexandras Avenue, 11522 Athens, Greece
| | - Constantin N. Baxevanis
- Cancer Immunology and Immunotherapy Center, Saint Savas Hospital, 171 Alexandras Avenue, 11522 Athens, Greece
| |
Collapse
|
13
|
Thompson LW, Garbee CF, Hibbitts S, Brinckerhoff LH, Pierce RA, Chianese-Bullock KA, Deacon DH, Engelhard VH, Slingluff CL. Competition Among Peptides in Melanoma Vaccines for Binding to MHC Molecules. J Immunother 2004; 27:425-31. [PMID: 15534486 DOI: 10.1097/00002371-200411000-00002] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The effectiveness of peptide-based cancer vaccines depends on the ability of peptides to bind to MHC molecules on the surface of antigen-presenting cells, where they reconstitute epitopes for cytotoxic T lymphocytes (CTLs). Multivalent vaccines have advantages over single-peptide vaccines; however, peptides may compete for binding to the same MHC molecules. In particular, it is possible that peptides with high affinity for MHC molecules prevent the binding of lower-affinity peptides. However, only small numbers of peptide/MHC complexes per cell are required for CTL recognition. Thus, the authors hypothesized that competition of peptides for MHC binding would not significantly reduce CTL recognition of individual peptides within a multiple-peptide mixture, and this hypothesis was tested by a series of experiments performed in vitro. In multiple experiments, two peptides with different affinities for HLA-A*0201 molecules were mixed at various concentrations and pulsed onto HLA-A2 cells, which were then evaluated for susceptibility to lysis by HLA-A*0201-restricted CTLs. CTL recognition of the melanoma peptides gp100(154-162) (KTWGQYWQV), gp100(280-288) (YLEPGPVTA), and tyrosinase(369-377D) (YMDGTMSQV) was maintained even when target cells were co-pulsed with equimolar concentrations of peptides with comparable or higher affinity for HLA-A2. In some cases, CTL recognition was maintained even when the higher-affinity peptide was present at concentrations several orders of magnitude higher than the target peptide. In addition, CTLs generated by in vitro stimulation with a peptide mixture developed reactivity to three different peptides, at a level comparable to that obtained by stimulation with each individual peptide separately. These data suggest that CTLs can respond to multiple peptides presented on the same antigen-presenting cells and justify further investigation, in clinical trials, of multiple-peptide cancer vaccines.
Collapse
Affiliation(s)
- Lee W Thompson
- Department of Surgery, Division of Surgical Oncology, University of Virginia Health System, Charlottesville, Virginia, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Albo F, Cavazza A, Giardina B, Marini M, Roda LG, Schumacher R, Spagnoli GC. Degradation of the tumor antigen epitope gp100280–288 by fibroblast-associated enzymes abolishes specific immunorecognition. Biochim Biophys Acta Gen Subj 2004; 1671:59-69. [PMID: 15026146 DOI: 10.1016/j.bbagen.2004.01.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2003] [Revised: 01/21/2004] [Accepted: 01/22/2004] [Indexed: 12/16/2022]
Abstract
Degradation of the tumor antigen epitope gp100(280-288) (YLEPGPVTA) was investigated in the presence of cultured human fibroblasts, and acellular supernatants obtained from these cells; the possible effect of substrate degradation on in vitro immunorecognition was also addressed. In the presence of fibroblasts, gp100(280-288) was degraded to free amino acids with a half-life of less than 4 min; hydrolysis data support the hypothesis that substrate degradation was mainly caused by the activity of cell-expressed amino- and carboxypeptidases. Gp100(280-288) was also degraded in the presence of acellular supernatants: under these conditions, the hydrolysis pattern was similar to that observed in the presence of whole cells, but degradation kinetics was slower. As a result of these phenomena, immunorecognition of gp100(280-288)-specific cytotoxic T lymphocyte (CTL) clones was severely hampered, and was totally suppressed after 90 min. In conclusion, the high activity of fibroblast-expressed proteases, and the presence of wide-scope soluble enzymes, may explain, at least in part, the low activity of peptide-based antineoplastic vaccines, as well as the transient effectiveness of subcutaneously administered peptides in general.
Collapse
Affiliation(s)
- Federica Albo
- Dipartimento di Neuroscienze, Università degli Studi di Roma Tor Vergata, Rome, Italy
| | | | | | | | | | | | | |
Collapse
|
15
|
Afrimzon E, Zurgil N, Shafran Y, Sandbank J, Orda R, Lalchuk S, Deutsch M. Monitoring of Intracellular Enzyme Kinetic Characteristics of Peripheral Mononuclear Cells in Breast Cancer Patients. Cancer Epidemiol Biomarkers Prev 2004; 13:235-41. [PMID: 14973101 DOI: 10.1158/1055-9965.epi-03-0153] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
A new methodology for the detection of functional response of peripheral blood mononuclear cells against breast cancer (BC) antigens was developed. The method is based on cellular enzymatic activity measurements, using a fluorogenic substrate. We used this method to estimate the kinetic activity of lymphocytes derived from cancer patients and healthy donors. The aim of the study was to determine a possible correlation between the basic characteristics (K(m) and V(max)) of biochemical enzymatic reactions in live peripheral white mononuclear cells and common clinical-pathological characteristics in BC patients. Our method shows that the enzymatic activity, upon interaction with mitogen or tumor antigens, of the peripheral blood cells in BC patients is different from the enzymatic reactions in healthy individuals. This holds true in the early stages, and the difference persists throughout all of the stages of the disease. This difference is manifested, primarily, by an increase in the K(m) values after cell incubation with tumor tissue. It was also demonstrated that higher K(m) values of tumor tissue-activated peripheral blood mononuclear cells are associated with a better prognostic status of the BC patients (lymph node-negative tumors, hormone receptor preservation, and the absence of Her-2/neu protein overexpression). Thus, the present methodology may serve as an additional criterion for prognosis and monitoring, both in BC patients, and in individuals associated with high cancer risk.
Collapse
Affiliation(s)
- Elena Afrimzon
- The Biophysical Interdisciplinary Jerome Schottenstein Center for the Research and the Technology of the Cellome, Department of Physics, Bar-Ilan University, Ramat Gan, Israel
| | | | | | | | | | | | | |
Collapse
|
16
|
Morisaki T, Matsumoto K, Onishi H, Kuroki H, Baba E, Tasaki A, Kubo M, Nakamura M, Inaba S, Yamaguchi K, Tanaka M, Katano M. Dendritic cell-based combined immunotherapy with autologous tumor-pulsed dendritic cell vaccine and activated T cells for cancer patients: rationale, current progress, and perspectives. Hum Cell 2003; 16:175-82. [PMID: 15147037 DOI: 10.1111/j.1749-0774.2003.tb00151.x] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Effective adoptive cancer immunotherapy depends on an ability to generate tumor-antigen-presenting cells and tumor-reactive effector lymphocytes and to deliver these effector cells to the tumor. Dendritic cells (DCs) are the most potent antigen-presenting cells, capable of sensitizing T cells to new and recall antigens. Many studies have shown that tumors express unique proteins that can be loaded on DCs to trigger an immune response. The current experimental and clinical statuses of adoptive transfer of tumor antigen-pulsed DCs and vaccine-primed activated T cells are summarized herein. Clinical trials of antigen-pulsed DCs have been conducted in patients with various types of cancer, including non-Hodgkin lymphoma, multiple myeloma, prostate cancer, renal cell carcinoma, malignant melanoma, colorectal cancer, and non-small cell lung cancer. These studies have shown that antigen-loaded DC vaccination is safe and promising for the treatment of cancer. In addition, tumor vaccine-primed T cells have been shown to induce antitumor activity in vivo. Several clinical studies are being conducted on the use of vaccine-primed T cells such as tumor-drainage lymph node. It is reasonable to consider using both tumor antigen-pulsed DCs and vaccine-primed lymphocytes as adjuvants. We are now investigating the use of autologous whole tumor antigen-pulsed DCs and the DC vaccine-primed activated lymphocytes in patients with multiple metastasis of solid tumors.
Collapse
Affiliation(s)
- Takashi Morisaki
- Department of Cancer Therapy and Research, Station for Collaborative Research, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Broder H, Anderson A, Kremen TJ, Odesa SK, Liau LM. MART-1 adenovirus-transduced dendritic cell immunization in a murine model of metastatic central nervous system tumor. J Neurooncol 2003; 64:21-30. [PMID: 12952283 DOI: 10.1007/bf02700017] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Dendritic cells (DCs) are potent antigen-presenting cells that have been shown to play a critical role in the initiation of host immune responses against tumor antigens. In this study, a recombinant adenovirus vector encoding the melanoma-associated antigen, MART-1, was used to transduce murine DCs, which were then tested for their ability to activate cytotoxic T lymphocytes (CTLs) and induce protective immunity against B16 melanoma tumor cells implanted intracranially. Genetic modifications of murine bone marrow-derived DCs to express MART-1 was achieved through the use of an E1-deficient, recombinant adenovirus vector. Sixty-two C57BL/6 mice were immunized subcutaneously with AdVMART-1-transduced DCs (n = 23), untransduced DCs (n = 17), or sterile saline (n = 22). Using the B16 murine melanoma, which naturally expresses the MART-1 antigen, all the mice were then challenged intracranially with viable, unmodified syngeneic B16 tumor cells 7 days later. Splenocytes from representative animals in each group were harvested for standard cytotoxicity (CTL) and enzyme-linked immunospot (ELISPOT) assays. The remaining mice were followed for survival. Immunization of C57BL/6 mice with DCs transduced with an adenoviral vector encoding the MART-1 antigen elicited the development of antigen-specific CTL responses. As evidenced by a prolonged survival curve when compared to control-immunized mice with intracranial B16 tumors, AdMART-1-DC vaccination was able to elicit partial protection against central nervous system tumor challenge in vivo.
Collapse
Affiliation(s)
- Howard Broder
- Division of Neurosurgery and the Jonsson Comprehensive Cancer Center, University of California at Los Angeles School of Medicine, Los Angeles, CA 90095-6901, USA
| | | | | | | | | |
Collapse
|
18
|
Flynn S, Stockinger B. Tumor and CD4 T-cell interactions: tumor escape as result of reciprocal inactivation. Blood 2003; 101:4472-8. [PMID: 12543861 DOI: 10.1182/blood-2002-10-3030] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
This paper addresses the capacity of naive, effector, and memory CD4 T cells to control growth of a major histocompatibility complex (MHC) class II-positive B-cell lymphoma in vivo. To assess the role of T cells on their own without contributions by B cells, antibodies, or natural killer (NK) cells, we generated pure effector or memory CD4 T cells in Rag-/-gc-/- mice deficient in endogenous lymphocytes and NK cells. Lymphoma cells expressing a model antigen were injected into mice with T cells of cognate specificity that were either naive or in effector or resting memory state. Naive T cells were unable to prevent tumor growth, probably due to delay of efficient cross-presentation by dendritic cells. However, both effector and memory T cells, dependent on the amount of antigen available, controlled the tumor for a considerable period of time without the need for dendritic cell stimulation. Nevertheless, the tumor eventually grew uncontrolled in all cases. This was not because of a defect in T-cell homing to the tumor site or loss of MHC class II or costimulatory molecules by the tumor, but reflected mutual paralysis of T-cell responsiveness and antigen processing by tumor cells.
Collapse
Affiliation(s)
- Sarah Flynn
- Division of Molecular Immunology, National Institute for Medical Research, The Ridgeway, Mill Hill, London United Kingdom.
| | | |
Collapse
|
19
|
Albo F, Cavazza A, Giardina B, Lippa S, Marini M, Roda LG, Spagnoli G. Degradation of the immunogenic peptide gp100(280-288) by the monocyte-like U937 cell line. Peptides 2003; 24:371-8. [PMID: 12732334 DOI: 10.1016/s0196-9781(03)00051-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The possible degradation of the tumor antigen epitope gp100(280-288) (YLEPGPVTA) in the presence of the monocyte-like line U937, and the effect of degradation on the in vitro-measured immune recognition, were investigated by chromatographic techniques and immunological assays. Results indicate a rapid hydrolysis of the substrate in the presence of the model cells, which is consistent with the hypothesis that degradation of gp100(280-288) is caused by the activity of U937-expressed enzymes, specifically amino- and carboxypeptidases. On the other hand, these results do not support the involvement of other enzymes known to be expressed by U937 cells. From a functional point of view, these data indicate that the degradation of gp100(280-288) severely hampered recognition by specific CTL clones. The results obtained may provide a model for epitope degradation by the antigen-presenting cells found in defined anatomical compartments and may, at least in part, account for the low activity of peptide-based antineoplastic vaccines, as well as for the transience of the effects of subcutaneously administered peptides in general.
Collapse
Affiliation(s)
- Federica Albo
- Dipartimento di Neuroscienze, Università Degli Studi di Roma "Tor Vergata", Via Montpellier 1, 00133 Rome, Italy
| | | | | | | | | | | | | |
Collapse
|
20
|
Abstract
Based on their unique ability to stimulate primary immune responses, dendritic cells are the most potent antigen-presenting cells known. This ability stems from the fact that they are very efficient at the uptake and processing of antigen and they express high levels of major histocompatibility complex class I and class II, as well as costimulatory molecules, which are required to prime naive cytotoxic T-cells. Many groups of investigators have tried to take advantage of these features by developing dendritic cell-based vaccines against tumors and infectious diseases. While the basic principle in these studies is the same--dendritic cells pulsed with antigen are used to elicit cytotoxic T-cell responses--the methods used are varied. This is particularly true with respect to the nature of the antigen used and the method of antigen delivery. In this article, we will focus on the use of RNA as a form of antigen with which to load dendritic cells. We will discuss the rationale behind using RNA as an antigen source and will review recent studies in both murine and human settings that use RNA-pulsed dendritic cells as vaccines.
Collapse
Affiliation(s)
- Smita Nair
- Department of Surgery, Duke University Medical Center, Durham, NC 27710, USA.
| | | |
Collapse
|
21
|
Gatza E, Okada CY. Tumor cell lysate-pulsed dendritic cells are more effective than TCR Id protein vaccines for active immunotherapy of T cell lymphoma. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2002; 169:5227-35. [PMID: 12391241 DOI: 10.4049/jimmunol.169.9.5227] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
TCR Id protein conjugated to keyhole limpet hemocyanin (KLH) (TCR Id:KLH) and injected with a chemical adjuvant (QS-21) induces a protective, Id-specific immune response against the murine T cell lymphoma, C6VL. However, Id-based immunotherapy of C6VL has not demonstrated therapeutic efficacy in tumor-bearing mice. We report here that C6VL lysate-pulsed dendritic cells (C6VL-DC) vaccines display enhanced efficacy in both the prevention and the therapy of T cell lymphoma compared with TCR Id:KLH with QS-21 vaccines. C6VL-DC vaccines stimulated potent tumor-specific immunity that protected mice against lethal challenge with C6VL and significantly enhanced the survival of tumor-bearing mice. Tumor-specific proliferation and secretion of IFN-gamma indicative of a Th1-type immune response were observed upon ex vivo stimulation of vaccine-primed lymph node cells. Adoptive transfer of immune T cell-enriched lymphocytes was sufficient to protect naive recipients from lethal tumor challenge. Furthermore, CD8(+) T cells were absolutely required for tumor protection. Although C6VL-DC and control vaccines stimulated low levels of tumor-specific Ab production in mice, Ab levels did not correlate with the protective ability of the vaccine. Thus, tumor cell lysate-pulsed DC vaccines appear to be an effective approach to generate potent T cell-mediated immune responses against T cell malignancies without requiring identification of tumor-specific Ags or patient-specific Id protein expression.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/administration & dosage
- Antibodies, Monoclonal/immunology
- Antibodies, Neoplasm/biosynthesis
- Antigens, Neoplasm/administration & dosage
- Antigens, Neoplasm/immunology
- Antigens, Neoplasm/toxicity
- Cancer Vaccines/administration & dosage
- Cancer Vaccines/immunology
- Cancer Vaccines/toxicity
- Cell Fractionation/methods
- Cell-Free System/immunology
- Cytokines/metabolism
- Dendritic Cells/immunology
- Dendritic Cells/transplantation
- Female
- Immunization Schedule
- Immunoglobulin Idiotypes/administration & dosage
- Immunoglobulin Idiotypes/immunology
- Immunotherapy, Active/methods
- Lymph Nodes/cytology
- Lymph Nodes/immunology
- Lymph Nodes/metabolism
- Lymphocyte Activation/immunology
- Lymphoma, T-Cell/immunology
- Lymphoma, T-Cell/mortality
- Lymphoma, T-Cell/therapy
- Mice
- Mice, Inbred C57BL
- Receptors, Antigen, T-Cell, alpha-beta/immunology
- Survival Analysis
- T-Lymphocytes/immunology
- T-Lymphocytes/transplantation
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- Erin Gatza
- Graduate Program in Immunology, University of Michigan Medical School, Ann Arbor 48101, USA.
| | | |
Collapse
|
22
|
Van Meirvenne S, Straetman L, Heirman C, Dullaers M, De Greef C, Van Tendeloo V, Thielemans K. Efficient genetic modification of murine dendritic cells by electroporation with mRNA. Cancer Gene Ther 2002; 9:787-97. [PMID: 12189529 DOI: 10.1038/sj.cgt.7700499] [Citation(s) in RCA: 89] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2002] [Indexed: 11/08/2022]
Abstract
Recently, human dendritic cells (DCs) pulsed with mRNA encoding a broad range of tumor antigens have proven to be potent activators of a primary anti-tumor-specific T-cell response in vitro. The aim of this study was to improve the mRNA pulsing of murine DC. Compared to a standard lipofection protocol and passive pulsing, electroporation was, in our hands, the most efficient method. The optimal conditions to electroporate murine bone marrow-derived DCs with mRNA were determined using enhanced green fluorescent protein and a truncated form of the nerve growth factor receptor. We could obtain high transfection efficiencies around 70-80% with a mean fluorescence intensity of 100-200. A maximal expression level was reached 3 hours after electroporation. A clear dose-response effect was seen depending on the amount of mRNA used. Importantly, the electroporation process did not affect the viability nor the allostimulatory capacity or phenotype of the DC. To study the capacity of mRNA-electroporated DCs to present antigen in the context of MHC classes I and II, we made use of chimeric constructs of ovalbumin. The dose-dependent response effect and the duration of presentation were also determined. Together, these results demonstrate that mRNA electroporation is a useful method to generate genetically modified murine DC, which can be used for preclinical studies testing immunotherapeutic approaches.
Collapse
Affiliation(s)
- Sonja Van Meirvenne
- Laboratory of Physiology-Immunology of the Medical School of the Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | | | | | | | | | | | | |
Collapse
|
23
|
Zhou Y, Bosch ML, Salgaller ML. Current methods for loading dendritic cells with tumor antigen for the induction of antitumor immunity. J Immunother 2002; 25:289-303. [PMID: 12142552 DOI: 10.1097/00002371-200207000-00001] [Citation(s) in RCA: 73] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The immunotherapy of cancer is predicated on the belief that it is possible to generate a clinically meaningful antitumor response that provides patient benefit, such as improvement in the time to progression or survival. Indeed, immunotherapeutics with dendritic cells (DC) as antigen-presenting delivery vehicles for cell-based vaccines have already improved patient outcome against a wide range of tumor types (1-9). This approach stimulates the patient's own antitumor immunity through the induction or enhancement of T-cell immunity. It is generally believed that the activity of cytotoxic T lymphocytes (CTL), the cells directly responsible for killing the tumor cells in vivo, are directed by DC. Therefore, the goal of many current designs for DC-based vaccines is to induce strong tumor-specific CTL responses in patients with cancer. In practice, most studies for DC-based cancer vaccine development have focused on the development of methods that can effectively deliver exogenous tumor antigens to DC for cross-priming of CD8+ T cells through the endogenous MHC class I processing and presentation pathway (10). To date, many methods have been developed or evaluated for the delivery of defined and undefined tumor antigens to DC. This review provides a brief summary on these methods, the techniques used in these methods, as well as the advantages and disadvantages of each method.
Collapse
Affiliation(s)
- Yaling Zhou
- Northwest Biotherapeutics, Incorporated, Bothell, Washington 98021, USA
| | | | | |
Collapse
|
24
|
Graffi SJ, Dekan G, Stingl G, Epstein MM. Systemic administration of antigen-pulsed dendritic cells induces experimental allergic asthma in mice upon aerosol antigen rechallenge. Clin Immunol 2002; 103:176-84. [PMID: 12027423 DOI: 10.1006/clim.2002.5190] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Antigen-pulsed dendritic cells (DCs) have been used extensively as cellular vaccines to induce a myriad of protective immune responses. Adoptive transfer of antigen-pulsed DCs is especially effective at generating Th1 and CD8 immune responses. However, recently this strategy has been shown to induce Th2 cells when DCs are administered locally into the respiratory tract. We sought to address whether systemic rather than local antigen-pulsed DC administration could induce Th2 experimental allergic asthma. We found that OVA-pulsed splenic DCs injected intraperitoneally induced polarized Th2 allergic lung disease upon secondary OVA aerosol challenge. Disease was characterized by eosinophilic lung inflammation, excess mucus production, airway hyperresponsiveness, and OVA-specific IgG1 and IgE. In addition, unusual pathology characterized by macrophage alveolitis and multinucleated giant cells was observed. These data show that systemic administration of antigen-pulsed DCs and subsequent aeroantigen challenge induces Th2 immunity. These findings have important implications for the development of DC-based vaccines.
Collapse
Affiliation(s)
- Sebastian J Graffi
- Division of Immunology, Allergy and Infectious Diseases, Department of Dermatology, VIRCC, University of Vienna Medical School, A-1235, Austria
| | | | | | | |
Collapse
|
25
|
Baxevanis CN, Gritzapis AD, Tsitsilonis OE, Katsoulas HL, Papamichail M. HER-2/neu-derived peptide epitopes are also recognized by cytotoxic CD3(+)CD56(+) (natural killer T) lymphocytes. Int J Cancer 2002; 98:864-72. [PMID: 11948464 DOI: 10.1002/ijc.10251] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
The human HER-2/neu gene encodes a 185 kDa transmembrane glycoprotein recognized by MHC class I-restricted CTLs. Here, we report that HER-2/neu peptide CTL epitopes can also be recognized by cytotoxic NK-T lymphocytes. Unfractionated peptides derived from HLA-A2(+), HER-2/neu(+) tumor cells acid cell extract (ACE), collected from patients with metastatic ovarian cancer, were used as antigen to generate in vitro cytotoxic effectors. ACE was able to elicit from cancer patients' PBMCs both alphabetaTCR(+)CD3(+)CD56(-) and alphaTCR(+)CD3(+)CD56(+) (NK-T) CTLs that lysed ACE-sensitized T2 cells in an HLA-A2-restricted manner. The same CTL lines also recognized T2 cells pulsed with HER-2/neu-derived CTL peptide epitopes, a HER-2/neu-transfected HLA-A2(+) cell line and autologous tumor cells. alphaTCR(+)CD3(+)CD56(+) CTL lines also exhibited NK-like cytotoxicity against autologous tumor cells. CTL clones were isolated from alphaTCR(+)CD3(+)CD56(+) bulk cultures displaying both MHC- and non-MHC-restricted cytotoxicity, thus confirming the dual cytolytic function of such cells. Our data demonstrate that ACE from metastatic ovarian tumors can be used as multiepitope vaccines for generating in vitro, besides classical CTLs, NK-T cells exerting efficient MHC- and non-MHC-restricted cytotoxicity against autologous tumor targets. Such NK-T cells expressing dual cytotoxic activity may prove advantageous in cancer immunotherapy.
Collapse
MESH Headings
- Adenocarcinoma/immunology
- Antibodies, Monoclonal/immunology
- CD3 Complex/immunology
- CD56 Antigen/immunology
- Cytotoxicity Tests, Immunologic
- Epitopes/immunology
- Female
- Flow Cytometry
- Humans
- Immunotherapy
- Killer Cells, Natural/immunology
- Lymphocytes, Tumor-Infiltrating/immunology
- Monocytes/immunology
- Ovarian Neoplasms/immunology
- Receptor, ErbB-2/immunology
- Receptors, Antigen, T-Cell, alpha-beta/metabolism
- T-Lymphocytes, Cytotoxic/immunology
- Transfection
- Tumor Cells, Cultured
Collapse
|
26
|
Santin AD, Bellone S, Ravaggi A, Roman JJ, Pecorelli S, Parham GP, Cannon MJ. Induction of tumour-specific CD8(+) cytotoxic T lymphocytes by tumour lysate-pulsed autologous dendritic cells in patients with uterine serous papillary cancer. Br J Cancer 2002; 86:151-7. [PMID: 11857027 PMCID: PMC2746546 DOI: 10.1038/sj.bjc.6600026] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2001] [Revised: 10/02/2001] [Accepted: 10/16/2001] [Indexed: 11/16/2022] Open
Abstract
Uterine serous papillary carcinoma is a highly aggressive variant of endometrial cancer histologically similar to high grade ovarian cancer. Unlike ovarian cancer, however, it is a chemoresistant disease from onset, with responses to combined cisplatinum-based chemotherapy in the order of 20% and an extremely poor prognosis. In this study, we demonstrate that tumour lysate-pulsed autologous dendritic cells can elicit a specific CD8(+) cytotoxic T lymphocyte response against autologous tumour target cells in three patients with uterine serous papillary cancer. CTL from patients 1 and 2 expressed strong cytolytic activity against autologous tumour cells, did not lyse autologous lymphoblasts or autologous EBV-transformed cell lines, and were variably cytotoxic against the NK-sensitive cell line K-562. Patient 3 CD8(+) T cells expressed a modest but reproducible cytotoxicity against autologous tumour cells only at the time of the first priming. Further priming attempts with PBL collected from patient 3 after tumour progression in the lumboaortic lymph nodes were unsuccessful. Cytotoxicity against autologous tumour cells could be significantly inhibited by anti-HLA class I (W6/32) and anti-LFA-1 MAbs. Highly cytotoxic CD8(+) T cells from patients 1 and 2 showed a heterogeneous CD56 expression while CD56 was not expressed by non-cytotoxic CD8(+) T cells from patient 3. Using two colour flow cytometric analysis of intracellular cytokine expression at the single cell level, a striking dominance of IFN-gamma expressors was detectable in CTL populations of patients 1 and 2 while in patient 3 a dominant population of CD8(+) T cells expressing IL-4 and IL-10 was consistently detected. Taken together, these data demonstrate that tumour lysate-pulsed DC can be an effective tool in inducing uterine serous papillary cancer-specific CD8(+) CTL able to kill autologous tumour cells in vitro. However, high levels of tumour specific tolerance in some patients may impose a significant barrier to therapeutic vaccination. These results may have important implications for the treatment in the adjuvant setting of uterine serous papillary cancer patients with active or adoptive immunotherapy.
Collapse
Affiliation(s)
- A D Santin
- Department of Obstetrics and Gynecology, UAMS Medical Center, Division of Gynecologic Oncology, University of Arkansas, 4301 W. Markham, Little Rock, Arkansas AR 72205-7199, USA.
| | | | | | | | | | | | | |
Collapse
|
27
|
Abstract
It is now clear that many human tumour antigens can be recognised by the immune system. These tumour antigens can be classified into several groups including cancer-testis, differentiation, tissue specific, over-expressed, and viral-associated antigens. In many cases, there is a known molecular basis of carcinogenesis which provides the explanation for the differentiated expression of these antigens in tumours compared with normal cells. Improved understanding of the biology of the immune response, particularly of immune recognition and activation of T-cells, allow better design of vaccines. Pre-clinical comparative studies allow evaluation of optimal vaccine strategies which can then be delivered to the clinic. Currently, a range of cancer vaccines are being tested including those using tumour cells, proteins, peptides, viral vectors, DNA or dendritic cells. Ultimately, this research should give rise to an entirely new modality of cancer treatments.
Collapse
Affiliation(s)
- Said Dermime
- CRC Department of Medical Oncology, University of Manchester and Paterson Institute for Cancer Research, Christie Hospital NHS Trust, Manchester, UK
| | | | | | | |
Collapse
|
28
|
Clark RE, Dodi IA, Hill SC, Lill JR, Aubert G, Macintyre AR, Rojas J, Bourdon A, Bonner PL, Wang L, Christmas SE, Travers PJ, Creaser CS, Rees RC, Madrigal JA. Direct evidence that leukemic cells present HLA-associated immunogenic peptides derived from the BCR-ABL b3a2 fusion protein. Blood 2001; 98:2887-93. [PMID: 11698267 DOI: 10.1182/blood.v98.10.2887] [Citation(s) in RCA: 194] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The BCR-ABL oncogene is central in the pathogenesis of chronic myeloid leukemia (CML). Here, tandem nanospray mass spectrometry was used to demonstrate cell surface HLA-associated expression of the BCR-ABL peptide KQSSKALQR on class I-negative CML cells transfected with HLA-A*0301, and on primary CML cells from HLA-A3-positive patients. These patients mounted a cytotoxic T-lymphocyte response to KQSSKALQR that also killed autologous CML cells, and tetramer staining demonstrated the presence of circulating KQSSKALQR-specific T cells. The findings are the first demonstration that CML cells express HLA-associated leukemia-specific immunogenic peptides and provide a sound basis for immunization studies against BCR-ABL.
Collapse
MESH Headings
- Adult
- Amino Acid Sequence
- Antigen Presentation
- Antigens, Neoplasm/chemistry
- Antigens, Neoplasm/immunology
- Antigens, Surface/chemistry
- Antigens, Surface/immunology
- Female
- Fusion Proteins, bcr-abl/chemistry
- Fusion Proteins, bcr-abl/immunology
- HLA-A3 Antigen/genetics
- HLA-A3 Antigen/immunology
- Humans
- K562 Cells/immunology
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/immunology
- Male
- Middle Aged
- Neoplasm Proteins/chemistry
- Neoplasm Proteins/immunology
- Neoplastic Stem Cells/immunology
- Peptide Fragments/chemistry
- Peptide Fragments/immunology
- Recombinant Fusion Proteins/immunology
- Spectrometry, Mass, Electrospray Ionization
- T-Lymphocytes, Cytotoxic/immunology
- Transfection
Collapse
Affiliation(s)
- R E Clark
- Department of Life Sciences and Chemistry and Physics, Nottingham Trent University, Nottingham, United Kingdom
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Ni HT, Spellman SR, Jean WC, Hall WA, Low WC. Immunization with dendritic cells pulsed with tumor extract increases survival of mice bearing intracranial gliomas. J Neurooncol 2001; 51:1-9. [PMID: 11349874 DOI: 10.1023/a:1006452726391] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
The purpose of this study is to investigate the efficacy of dendritic cell-mediated immunotherapy against intracranial gliomas. Cloned DC2.4 dendritic cells originating from C57BL/6 mice were pulsed with glioma GL261 cell extracts and administered i.p. to C57BL/6 mice with intracranial GL261 gliomas. The survival of mice with and without pulsed dendritic cells was monitored after intracranial implantation of the GL261 glioma cells. Fluorescence activated cell sorting (FACS) analysis showed that DC2.4 cells express high levels of MHC class I and class II molecules, costimulatory molecules B7-1 and B7-2, and the cell adhesion molecule ICAM-1. Antigen-presenting capabilities in these dendritic cells were initially characterized in vitro by a mixed lymphocyte reaction, showing that Balb/c CD4+ and CD8+ T cells were able to generate allogeneic responses to DC2.4 cells. Tumor extract-pulsed DC2.4 dendritic cells were then used for the treatment of C57BL/6 mice with syngeneic GL261 gliomas. Animals with intracranial GL261 gliomas and vaccinated i.p. with pulsed DC2.4 dendritic cells exhibited significantly enhanced survival, relative to animals treated with saline or non-pulsed DC2.4 cells alone. In addition, cured animals showed an increased delayed-type hypersensitivity response to GL261 cells and survived when rechallenged with intracranial GL261 gliomas. In summary these results indicate that dendritic cells pulsed with tumor extract can enhance immune responses to tumor antigen and therefore represent a potential immunotherapeutic approach for treating patients with intracranial gliomas.
Collapse
Affiliation(s)
- H T Ni
- Department of Neurosurgery, University of Minnesota, Minneapolis 55455, USA
| | | | | | | | | |
Collapse
|
30
|
Okada T, Shah M, Higginbotham JN, Li Q, Wildner O, Walbridge S, Oldfield E, Blaese RM, Ramsey WJ. AV.TK-mediated killing of subcutaneous tumors in situ results in effective immunization against established secondary intracranial tumor deposits. Gene Ther 2001; 8:1315-22. [PMID: 11571568 DOI: 10.1038/sj.gt.3301526] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2000] [Accepted: 06/11/2001] [Indexed: 11/09/2022]
Abstract
Gene transfer vectors expressing herpes simplex thymidine kinase (HSVtk), in addition to direct killing of tumor cells, often have an associated local "bystander effect" mediated by metabolic coupling of tumor cells. A systemic antitumor effect mediated by the immune system, termed the distant bystander effect, has also been reported. We have observed the development of cytotoxic T-lymphocyte (CTL) populations and long-lasting antitumor immunity following treatment of subcutaneous tumors with an adenoviral vector expressing HSVtk (AV.TK) and ganciclovir (GCV) in rat glioma model. This vaccination effect seen with AV.TK/GCV treatment of subcutaneous tumor could even abrogate or retard growth of previously established secondary intracranial tumors.
Collapse
Affiliation(s)
- T Okada
- Clinical Gene Therapy Branch, National Human Genome Research Institute, Bethesda, MD, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Chang JS, Choi MJ, Cheong HS, Kim K. Development of Th1-mediated CD8+ effector T cells by vaccination with epitope peptides encapsulated in pH-sensitive liposomes. Vaccine 2001; 19:3608-14. [PMID: 11395193 DOI: 10.1016/s0264-410x(01)00104-9] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
There have been many studies for tumor therapy mediated by cytotoxic T lymphocytes (CTL) that recognize tumor-associated antigen. It is generally accepted that CTL responses are induced when antigen is delivered into the cytosol. The pH-sensitive liposomes as vehicles are well known for their capacity to deliver the antigen into the cytosol. In this work, immunization of mice with CTL epitope peptides from Hantaan nucleocapsid protein (M6) or human papilloma virus E7 encapsulated in pH-sensitive liposomes induced effective antigen-specific CTL responses. The CTL responses induced by M6 peptide encapsulated in pH-sensitive liposomes blocked the formation of tumor mass from Hantaan NP transfected B16 melanoma cells in C57BL/6 mice and delayed the growth of preinoculated melanoma cells. During the blockade of the tumor growth, the CTL response was maintained for at least approximately 6 weeks, and the mice secreted Th1 type cytokines such as IL-2 and IFN-gamma. These results suggested that the pH-sensitive liposomes might provide an effective peptide delivery system for CTL-mediated tumor therapy.
Collapse
Affiliation(s)
- J S Chang
- Drug Delivery Research Lab., Mogam Biotechnology Research Institute, 341 Pojung-ri, Koosung-Myon, Yongin City, Kyonggi-do 449-910, South Korea
| | | | | | | |
Collapse
|
32
|
Staib L, Birebent B, Somasundaram R, Purev E, Braumüller H, Leeser C, Küttner N, Li W, Zhu D, Diao J, Wunner W, Speicher D, Beger HG, Song H, Herlyn D. Immunogenicity of recombinant GA733-2E antigen (CO17-1A, EGP, KS1-4, KSA, Ep-CAM) in gastro-intestinal carcinoma patients. Int J Cancer 2001. [DOI: 10.1002/1097-0215(200102)9999:9999<::aid-ijc1164>3.0.co;2-j] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
33
|
Abstract
The characterization of tumor-associated antigens recognized by human T lymphocytes in a major histocompatibility complex (MHC)-restricted fashion has opened new possibilities for immunotherapeutic approaches to the treatment of human cancers. Dendritic cells (DC) are professional antigen presenting cells that are well suited to activate T cells toward various antigens, such as tumor-associated antigens, due to their potent costimulatory activity. The availability of large numbers of DC, generated either from hematopoietic progenitor cells or monocytes in vitro or isolated from peripheral blood, has profoundly changed pre-clinical research as well as the clinical evaluation of these cells. Accordingly, appropriately pulsed or transfected DC may be used for vaccination in the field of infectious diseases or tumor immunotherapy to induce antigen-specific T cell responses. These observations led to pilot clinical trials of DC vaccination for patients with cancer in order to investigate the feasibility, safety, as well as the immunologic and clinical effects of this approach. Initial clinical studies of human DC vaccines are generating encouraging preliminary results demonstrating induction of tumor-specific immune responses and tumor regression. Nevertheless, much work is still needed to address several variables that are critical for optimizing this approach and to determine the role of DC-based vaccines in tumor immunotherapy.
Collapse
Affiliation(s)
- N Meidenbauer
- Department of Hematology/Oncology, University of Regensburg, Germany
| | | | | |
Collapse
|
34
|
Broder H, Anderson A, Odesa SK, Kremen TJ, Liau LM. Recombinant adenovirus-transduced dendritic cell immunization in a murine model of central nervous system tumor. Neurosurg Focus 2000; 9:e6. [PMID: 16817689 DOI: 10.3171/foc.2000.9.6.7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Object
Dendritic cells (DCs) are potent antigen-presenting cells that have been shown to play a critical role in the initiation of host immune responses against tumor antigens. In this study, a recombinant adenovirus vector encoding the melanoma-associated antigen, MART-1, was used to transduce murine DCs, which were then tested for their ability to activate cytotoxic T lymphocytes (CTLs) and induce protective immunity against B16 melanoma tumor cells implanted intracranially.
Methods
Genetic modification of murine bone marrrow–derived DCs to express MART-1 was achieved through the use of an E1-deficient, recombinant adenovirus vector (AdVMART1). Sixty-two C57BL/6 mice were immunized by subcutaneous injection of AdVMART-1-transduced DCs (23 mice), untransduced DCs (17 mice), or sterile saline (22 mice). Using the B16 murine melanoma, which naturally expresses the MART-1 antigen, all the mice were then challenged intracranially with viable, unmodified syngeneic B16 tumor cells 7 days later. Splenocytes obtained from representative animals in each group were harvested for standard cytotoxicity and enzyme-linked immunospot assays. The remaining mice were followed for survival.
Immunization of C57BL/6 mice with DCs transduced with AdVMART1-DC elicited the development of antigen-specific CTL responses. As evidenced by a prolonged survival curve when compared with control-immunized mice harboring intracranial B16 tumors, AdMART1-DC vaccination was able to elicit partial protection against central nervous system (CNS) tumor challenge in vivo. However, this CNS antitumor immunity was weaker than that previously demonstrated against subcutaneous B16 tumors in which the same vaccination strategy was used.
Conclusions
These data suggest that immune responses generated against CNS tumors by DC-based vaccines may be different from those obtained against subcutaneous tumors.
Collapse
Affiliation(s)
- H Broder
- Division of Neurosurgery, University of California at Los Angeles School of Medicine, Los Angeles, California 90095-6901, USA
| | | | | | | | | |
Collapse
|
35
|
Induction of ovarian tumor-specific CD8+ cytotoxic T lymphocytes by acid-eluted peptide-pulsed autologous dendritic cells. Obstet Gynecol 2000. [PMID: 10960637 DOI: 10.1097/00006250-200009000-00019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVE To evaluate the potential of dendritic cells pulsed with acid-eluted peptides derived from autologous ovarian cancer cells for eliciting a tumor-specific cytotoxic T cell response in women with advanced ovarian cancer. METHODS CD8+ T lymphocytes derived from peripheral blood mononuclear cells stimulated in vitro with autologous ovarian tumor peptide-pulsed dendritic cells were tested for their ability to induce an HLA class I-restricted cytotoxic T lymphocyte response against autologous tumor cells. To correlate cytotoxic activity by cytotoxic T lymphocytes with T cell phenotype, we used two-color flow cytometric analysis of surface markers and intracellular cytokine expression (interferon-gamma versus interleukin-4). RESULTS CD8+ cytotoxic T lymphocyte responses against autologous ovarian tumor cells were elicited in three consecutive women who had advanced ovarian cancer. Although cytotoxic T lymphocyte populations from all women expressed strong cytolytic activity against autologous tumor cells, they did not lyse autologous lymphoblasts or Epstein-Barr virus-transformed cell lines, and they showed negligible cytotoxicity against the natural killer-sensitive cell line K-562. Cytotoxicity against the autologous tumor cells was significantly inhibited by anti-HLA class I (W6/32) and anti-HLA-A2 (BB7-2) monoclonal antibodies. CD8+ cytotoxic T lymphocytes expressed variable levels of CD56 and preferentially expressed interferon-gamma rather than interleukin-4. CONCLUSIONS Peptide-pulsed dendritic cells induced specific CD8+ cytotoxic T lymphocytes that killed autologous tumor cells from women with advanced ovarian cancer. This finding might contribute to the development of active or adoptive immunotherapy for residual or resistant ovarian cancer after standard surgery and cytotoxic treatment.
Collapse
|
36
|
Santin AD, Hermonat PL, Ravaggi A, Bellone S, Pecorelli S, Cannon MJ, Parham GP. In vitro induction of tumor-specific human lymphocyte antigen class I-restricted CD8 cytotoxic T lymphocytes by ovarian tumor antigen-pulsed autologous dendritic cells from patients with advanced ovarian cancer. Am J Obstet Gynecol 2000; 183:601-9. [PMID: 10992180 DOI: 10.1067/mob.2000.107097] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
OBJECTIVE The purpose of this study was to evaluate the potential of dendritic cells pulsed with whole-tumor extracts derived from autologous ovarian cancer cells in eliciting a tumor-specific cytotoxic T-cell response in vitro from patients with advanced ovarian cancer. STUDY DESIGN CD8(+) T lymphocytes stimulated in vitro with autologous ovarian tumor lysate-pulsed dendritic cells were tested for their ability to induce a human leukocyte antigen class I-restricted cytotoxic T-lymphocyte response able to specifically kill autologous tumor cells in standard 6-hour chromium 51 cytotoxicity assays. In addition, to correlate cytotoxic activity by cytotoxic T-lymphocytes with a particular lymphoid subset, 2-color flow cytometric analysis of intracellular cytokine expression (interferon gamma and interleukin 4) at the single-cell level was performed. RESULTS Cytotoxic T lymphocytes specific for autologous ovarian tumor cells were elicited from 3 patients with advanced ovarian cancer. Although cytotoxic T-lymphocyte populations expressed strong cytolytic activity against autologous tumor cells, they did not lyse concanavalin A-stimulated autologous lymphocytes or autologous Epstein-Barr virus-transformed lymphoblastoid cell lines and showed negligible cytotoxicity against the natural killer cell-sensitive cell line K-562. Cytotoxic effect against the autologous tumor cells was inhibited by an anti-human leukocyte antigen class I monoclonal antibody (W6/32). It is interesting that CD8(+) cytotoxic T lymphocytes expressed variable levels of CD56, a marker that may be associated with high cytotoxic activity. Finally, most of the tumor-specific CD8(+) T cells exhibited a T(H)1 cytokine bias, and a high percentage of interferon gamma expressors among cytotoxic T lymphocytes was correlated with higher cytotoxic activity. CONCLUSION These data show that tumor lysate-pulsed dendritic cells can consistently induce in vitro specific CD8(+) cytotoxic T lymphocytes able to kill autologous tumor cells from patients with advanced stage ovarian cancer. This novel approach may have important implications for the treatment of residual or resistant disease with active or adoptive immunotherapy after standard surgical and cytotoxic treatment.
Collapse
Affiliation(s)
- A D Santin
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of Arkansas, Little Rock, AR 72205-7199, USA
| | | | | | | | | | | | | |
Collapse
|
37
|
Zier K, Johnson K, Maddux JM, Sung M, Mandeli J, Eisenbach L, Schwartz M. IFNgamma secretion following stimulation with total tumor peptides from autologous human tumors. J Immunol Methods 2000; 241:61-8. [PMID: 10915849 DOI: 10.1016/s0022-1759(00)00193-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
Several issues remain to be resolved before the efficacy of various approaches to elicit anti-tumor immunity in patients can be evaluated. First, in vitro assays able to detect responses by T cells primed in vivo are needed. Second, a source of tumor antigen to stimulate patients' lymphocytes in vitro is required. The ELISPOT assay is attractive, because it can be performed with a small numbers of cells and requires only short-term culture in vitro. A source of tumor antigen is more problematic, since for most tumors, tumor-associated antigens (TAA) have not been identified and/or cloned. In this report we demonstrate that autologous antigen-presenting cells (APC) pulsed with total tumor peptides from autologous tumor tissue can stimulate IFNgamma release by patients' lymphocytes in the ELISPOT assay. Thus, this approach should be considered for monitoring immune responses in clinical immunotherapy trials.
Collapse
Affiliation(s)
- K Zier
- Immunobiology Center, The Mount Sinai School of Medicine, New York, NY 10029, USA.
| | | | | | | | | | | | | |
Collapse
|
38
|
Timmerman JM, Levy R. Linkage of foreign carrier protein to a self-tumor antigen enhances the immunogenicity of a pulsed dendritic cell vaccine. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2000; 164:4797-803. [PMID: 10779787 DOI: 10.4049/jimmunol.164.9.4797] [Citation(s) in RCA: 83] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The unique Ag-presenting capabilities of dendritic cells (DCs) make them attractive vehicles for the delivery of therapeutic cancer vaccines. While tumor Ag-pulsed DC vaccination has shown promising results in a variety of murine tumor models and early clinical trials, the optimal form of tumor Ag for use in DC pulsing has not been determined. We have studied DC vaccination using alternative forms of a soluble protein tumor Ag, the tumor-specific Ig idiotype (Id) expressed by a murine B cell lymphoma. Vaccination of mice with Id-pulsed DCs was able to induce anti-Id Abs only when the Id was modified to constitute a hapten-carrier system. DCs pulsed with Id proteins modified to include foreign constant regions, foreign constant regions plus GM-CSF, or linkage to keyhole limpet hemocyanin (KLH) carrier protein were increasingly potent in their ability to elicit anti-Id Abs. Vaccination with Id-KLH-pulsed DCs induced tumor-protective immunity superior to that obtained with Id-KLH plus a chemical adjuvant, and protection was not dependent upon effector T cells. Rather, protection was associated with the induction of high titers of anti-Id Abs of the IgG2a subclass, characteristic of a Th1 response. These findings have implications for the design of therapeutic Ag-pulsed DC vaccines for cancer immunotherapy in humans.
Collapse
MESH Headings
- Adjuvants, Immunologic/administration & dosage
- Adjuvants, Immunologic/metabolism
- Adoptive Transfer
- Animals
- Antigens, Neoplasm/administration & dosage
- Antigens, Neoplasm/immunology
- Antigens, Neoplasm/metabolism
- CD4-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/immunology
- Cancer Vaccines/administration & dosage
- Cancer Vaccines/immunology
- Cancer Vaccines/metabolism
- Carrier Proteins/administration & dosage
- Carrier Proteins/immunology
- Carrier Proteins/metabolism
- Dendritic Cells/immunology
- Dendritic Cells/transplantation
- Female
- Granulocyte-Macrophage Colony-Stimulating Factor/administration & dosage
- Granulocyte-Macrophage Colony-Stimulating Factor/immunology
- Granulocyte-Macrophage Colony-Stimulating Factor/metabolism
- Hemocyanins/administration & dosage
- Hemocyanins/immunology
- Hemocyanins/metabolism
- Humans
- Immunoglobulin Idiotypes/administration & dosage
- Immunoglobulin Idiotypes/genetics
- Immunoglobulin Idiotypes/metabolism
- Immunoglobulin Isotypes/administration & dosage
- Immunoglobulin Isotypes/biosynthesis
- Lymphoma/immunology
- Lymphoma/prevention & control
- Mice
- Mice, Inbred C3H
- Neoplasm Transplantation
- Recombinant Fusion Proteins/administration & dosage
- Recombinant Fusion Proteins/immunology
- Recombinant Proteins
- Th1 Cells/immunology
- Tumor Cells, Cultured
- Vaccines, Conjugate/administration & dosage
- Vaccines, Conjugate/immunology
- Vaccines, Conjugate/metabolism
Collapse
Affiliation(s)
- J M Timmerman
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | | |
Collapse
|
39
|
Baxevanis CN, Voutsas IF, Tsitsilonis OE, Gritzapis AD, Sotiriadou R, Papamichail M. Tumor-specific CD4+ T lymphocytes from cancer patients are required for optimal induction of cytotoxic T cells against the autologous tumor. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2000; 164:3902-12. [PMID: 10725753 DOI: 10.4049/jimmunol.164.7.3902] [Citation(s) in RCA: 129] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
This study focuses on the specific CD4+ T cell requirement for optimal induction of cytotoxicity against MHC class II negative autologous tumors (AuTu) collected from patients with various types of cancer at advanced stages. CD4+ T cells were induced in cultures of cancer patients' malignant effusion-associated mononuclear cells with irradiated AuTu (mixed lymphocyte tumor cultures (MLTC)) in the presence of recombinant IL-2 and recombinant IL-7. Tumor-specific CD4+ T cells did not directly recognize the AuTu cells, but there was an MHC class II-restricted cross-priming by autologous dendritic cells (DCs), used as APC. CD8+ CTL, also induced during the MLTC, lysed specifically AuTu cells or DCs pulsed with AuTu peptide extracts (acid wash extracts (AWE)) in an MHC class I-restricted manner. Removal of CD4+ T cells or DCs from the MLTC drastically reduced the CD8+ CTL-mediated cytotoxic response against the AuTu. AWE-pulsed DCs preincubated with autologous CD4+ T cells were able, in the absence of CD4+ T cells, to stimulate CD8+ T cells to lyse autologous tumor targets. Such activated CD8+ T cells produced IL-2, IFN-gamma, TNF-alpha, and GM-CSF. The process of the activation of AWE-pulsed DCs by CD4+ T cells could be inhibited with anti-CD40 ligand mAb. Moreover, the role of CD4+ T cells in activating AWE-pulsed DCs was undertaken by anti-CD40 mAb. Our data demonstrate for the first time in patients with metastatic cancer the essential role of CD4+ Th cell-activated DCs for optimal CD8+ T cell-mediated killing of autologous tumors and provide the basis for the design of novel protocols in cellular adoptive immunotherapy of cancer, utilizing synthetic peptides capable of inducing T cell help in vivo.
Collapse
Affiliation(s)
- C N Baxevanis
- Cancer Immunology and Immunotherapy Center, Saint Savas Cancer Hospital, Athens, Greece.
| | | | | | | | | | | |
Collapse
|
40
|
Bremers AJ, Parmiani G. Immunology and immunotherapy of human cancer: present concepts and clinical developments. Crit Rev Oncol Hematol 2000; 34:1-25. [PMID: 10781746 DOI: 10.1016/s1040-8428(99)00059-1] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Immunotherapy of cancer is entering into a new phase of active investigation both at the pre-clinical and clinical level. This is due to the exciting developments in basic immunology and tumour biology that have allowed a tremendous increase in our understanding of mechanisms of interactions between the immune system and tumour cells. This review briefly summarizes the state of the art in basic tumour immunology before discussing the clinical applications of the new concepts in the clinical setting. Clinical approaches are diverse but can now be based on strong scientific rationales. The analysis of the available clinical results suggests that, despite some disappointments, there is room for optimism that both active immunotherapy (vaccination) and adoptive immunotherapy may soon become part of the therapeutic arsenal to combat cancer in a more efficient way.
Collapse
Affiliation(s)
- A J Bremers
- Unit of Immunotherapy of Human Tumours, Istituto Nazionale per lo Studio e la Cura dei Tumori, Via Venezian 1, 20133, Milan, Italy
| | | |
Collapse
|
41
|
Heimberger AB, Crotty LE, Archer GE, McLendon RE, Friedman A, Dranoff G, Bigner DD, Sampson JH. Bone marrow-derived dendritic cells pulsed with tumor homogenate induce immunity against syngeneic intracerebral glioma. J Neuroimmunol 2000; 103:16-25. [PMID: 10674985 DOI: 10.1016/s0165-5728(99)00172-1] [Citation(s) in RCA: 98] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
To evaluate the efficacy and toxicity of dendritic cell (DC) based therapy for intracerebral gliomas, we utilized a cell line derived from an astrocytoma that arose spontaneously in a VM/Dk mouse. This astrocytoma mirrors human gliomas phenotypically, morphologically and secretes transforming growth factor (TGF)-betas, immunosuppressive cytokines secreted by human gliomas. Systemic vaccination of mice with DCs pulsed with tumor homogenate followed by intracranial tumor challenge produced a > 160% increase in median survival (p = 0.016) compared with mice vaccinated with PBS or unpulsed DCs (p = 0.083). Fifty percent of mice treated with pulsed DCs survived long-term. Immunologic memory was demonstrated by survival of mice rechallenged with tumor. Both cell-mediated and humoral immunity was induced. On histological examination only focal areas of demyelination at the tumor implantation site were present. There was no evidence that autoimmune encephalomyelitis was induced by DC vaccination. Therefore, in a murine model, vaccination with DCs pulsed with glioma tumor homogenate is a safe and effective therapy against a syngeneic glioma located in the immunologically privileged central nervous system (CNS).
Collapse
Affiliation(s)
- A B Heimberger
- Division of Neurosurgery, Duke University Medical Center, Durham, NC 27710, USA
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Seo N, Tokura Y, Nishijima T, Hashizume H, Furukawa F, Takigawa M. Percutaneous peptide immunization via corneum barrier-disrupted murine skin for experimental tumor immunoprophylaxis. Proc Natl Acad Sci U S A 2000; 97:371-6. [PMID: 10618425 PMCID: PMC26670 DOI: 10.1073/pnas.97.1.371] [Citation(s) in RCA: 83] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/1999] [Accepted: 10/01/1999] [Indexed: 12/19/2022] Open
Abstract
H-2K(b)-restricted tumor epitope peptides, including tyrosinase-related protein 2 residues 181-188 (TRP-2) and connexin 37 residues 52-59 (MUT1), were applied to permeability barrier-disrupted C57BL/6 (B6) mouse skin from which the stratum corneum of the epidermis had been removed by tape-stripping. This procedure primed tumor-specific cytotoxic T lymphocytes (CTLs) in the lymph nodes and spleen, protected mice against subsequent challenge with corresponding tumor cells, and suppressed the growth of established tumors. Preventive and therapeutic effectiveness was correlated with the frequency of tumor-specific CTL precursors. MHC class II Ia(b+) cells separated from tape-stripped skin, compared with those from intact skin, exhibited a strong antigen-presenting capacity for CTL, suggesting that CTL expansion after peptide application is primarily mediated by epidermal Langerhans cells. Thus, percutaneous peptide immunization via barrier-disrupted skin provides a simple and noninvasive means of inducing potent anti-tumor immunity which may be exploited for cancer immunotherapy.
Collapse
Affiliation(s)
- N Seo
- Department of Dermatology, Hamamatsu University School of Medicine, 3600 Handa-cho, Hamamatsu 431-3192, Japan.
| | | | | | | | | | | |
Collapse
|
43
|
Brinckerhoff LH, Kalashnikov VV, Thompson LW, Yamshchikov GV, Pierce RA, Galavotti HS, Engelhard VH, Slingluff CL. Terminal modifications inhibit proteolytic degradation of an immunogenic MART-1(27-35) peptide: implications for peptide vaccines. Int J Cancer 1999; 83:326-34. [PMID: 10495424 DOI: 10.1002/(sici)1097-0215(19991029)83:3<326::aid-ijc7>3.0.co;2-x] [Citation(s) in RCA: 125] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Peptide epitopes for tumor-reactive cytotoxic T-lymphocytes (CTL) have been identified on human cancers and are being used in tumor vaccine trials. However, the pharmacokinetics and pharmacodynamics of such peptides have been inadequately studied. It is predicted that immunogenic tumor peptides would have short half-lives in vivo. The goal of the present work was to evaluate the stability of the immunogenic peptide MART-1(27-35) in fresh normal human plasma (NHP) and to identify modifications that convey protection against enzymatic destruction without loss of immunogenicity. We evaluated the stability of the MART-1(27-35) peptide (AAGIGILTV) and modified forms of that peptide for stability and immune recognition in an in vitro model. The peptides were incubated in plasma for varied time intervals and evaluated for their ability to reconstitute the epitope for MART-1(27-35)-reactive CTL. Loss of CTL reactivity signaled loss of immunoreactive peptide. When 1 microM MART-1(27-35) peptide was incubated in plasma prior to pulsing on target cells, CTL reactivity was lost within 3 hr, and the calculated half-life of this peptide was 22 sec. This degradation was mediated by peptidases. The stability of MART-1(27-35) was markedly prolonged by C-terminal amidation and/or N-terminal acetylation (peptide capping), or by polyethylene-glycol modification (PEGylation) of the C-terminus. These modified peptides were recognized by CTL. The MART-1(27-35) peptide is very unstable in plasma. It is probable that it and other immunogenic peptides will be similarly unstable in vivo. Immunogenicity of these peptides might be enhanced by creating modifications that enhance stability.
Collapse
Affiliation(s)
- L H Brinckerhoff
- Department of Surgery, University of Virginia, Health Sciences Center, Charlottesville, VA, 22906, USA
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Bellone M, Iezzi G, Imro MA, Protti MP. Cancer immunotherapy: synthetic and natural peptides in the balance. IMMUNOLOGY TODAY 1999; 20:457-62. [PMID: 10500293 DOI: 10.1016/s0167-5699(99)01503-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
The identification of human tumor-associated antigens has opened new avenues for immune intervention in cancer. Clinical trials using synthetic peptides that match segments of known tumor-associated proteins are ongoing. Alternatively, naturally processed peptides, obtained by acid treatment of tumor cells can be used. Here, Matteo Bellone and colleagues discuss the advantages and disadvantages of synthetic versus natural tumor peptides in cancer immunotherapy.
Collapse
Affiliation(s)
- M Bellone
- Laboratory of Tumor Immunology, and the Cancer Immunotherapy and Gene Therapy Program, Instituto Scientifico H San Raffaele, Via Olgettina 60, 20132 Milan, Italy.
| | | | | | | |
Collapse
|
45
|
Abstract
There is increasing evidence that tumors express putative target molecules for a therapeutic immune reaction. Yet, tumor cells lack the prerequisites for appropriate antigen presentation and--hence--the immune system does not respond. This difficulty can probably be circumvented when tumor antigens are processed by conventional antigen presenting cells. Thus, the identification of immunogenic tumor-associated antigens may allow new modes of vaccination with the hope of adding a fourth and hopefully powerful weapon to surgery, radiation and chemotherapy in the fight against cancer.
Collapse
Affiliation(s)
- M Zöller
- Department of Tumor Progression and Immune Defense, German Cancer Research Center, Heidelberg.
| | | |
Collapse
|
46
|
Men Y, Miconnet I, Valmori D, Rimoldi D, Cerottini JC, Romero P. Assessment of Immunogenicity of Human Melan-A Peptide Analogues in HLA-A*0201/Kb Transgenic Mice. THE JOURNAL OF IMMUNOLOGY 1999. [DOI: 10.4049/jimmunol.162.6.3566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Abstract
Previous studies have shown that substitution of single amino acid residues in human Melan-A immunodominant peptides Melan-A27–35 and Melan-A26–35 greatly improved their binding and the stability of peptide/HLA-A*0201 complexes. In particular, one Melan-A peptide analogue was more efficient in the generation of Melan-A peptide-specific and melanoma-reactive CTL than its parental peptide in vitro from human PBL. In this study, we analyzed the in vivo immunogenicity of Melan-A natural peptides and their analogues in HLA-A*0201/Kb transgenic mice. We found that two human Melan-A natural peptides, Melan-A26–35 and Melan-A27–35, were relatively weak immunogens, whereas several Melan-A peptide analogues were potent immunogens for in vivo CTL priming. In addition, induced Melan-A peptide-specific mouse CTL cross-recognized natural Melan-A peptides and their analogues. More interestingly, these mouse CTL were also able to lyse human melanoma cell lines in vitro in a HLA-A*0201-restricted, Melan-A-specific manner. Our results indicate that the HLA-A*0201/Kb transgenic mouse is a useful animal model to perform preclinical testing of potential cancer vaccines, and that Melan-A peptide analogues are attractive candidates for melanoma immunotherapy.
Collapse
Affiliation(s)
- Ying Men
- *Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne, Epalinges, Switzerland; and
| | - Isabelle Miconnet
- *Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne, Epalinges, Switzerland; and
| | - Danila Valmori
- †Division of Clinical Onco-Immunology, Ludwig Institute for Cancer Research, Lausanne Branch, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Donata Rimoldi
- *Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne, Epalinges, Switzerland; and
| | - Jean-Charles Cerottini
- *Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne, Epalinges, Switzerland; and
- †Division of Clinical Onco-Immunology, Ludwig Institute for Cancer Research, Lausanne Branch, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Pedro Romero
- *Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne, Epalinges, Switzerland; and
- †Division of Clinical Onco-Immunology, Ludwig Institute for Cancer Research, Lausanne Branch, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| |
Collapse
|
47
|
De Veerman M, Heirman C, Van Meirvenne S, Devos S, Corthals J, Moser M, Thielemans K. Retrovirally Transduced Bone Marrow-Derived Dendritic Cells Require CD4+ T Cell Help to Elicit Protective and Therapeutic Antitumor Immunity. THE JOURNAL OF IMMUNOLOGY 1999. [DOI: 10.4049/jimmunol.162.1.144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Abstract
It has been extensively documented that murine dendritic cells loaded with tumor-associated Ag (TAA)-derived peptides or protein can prime Ag-specific CD8+ cytotoxic T cells in vivo and can elicit Ag-specific immunity. Optimal presentation of TAA might be achieved by retroviral transduction of DCs allowing long term and stable expression of the TAA-peptides as well as the presentation of multiple epitopes in the context of MHC class I and/or class II molecules. Here we show that retroviral transduction of bone marrow-derived dendritic cells (DCs) with chicken OVA cDNA or the reporter gene green fluorescent protein retained their potent stimulatory capacity and that the transduced DCs could process and present the endogenously expressed OVA protein. The DCs transduced with cDNA encoding native OVA protein presented OVA-derived peptides in the context of MHC class I as well as MHC class II and induced a strong Ag-specific CTL response. DCs expressing a cytosolic form of OVA presented OVA peptides only in the context of MHC class I and failed to induce an OVA-specific CTL response in vivo when they had been cultured in the absence of exogenous protein. Immunization with retrovirally transduced DCs resulted in an Ag-specific immunity and rejection of a tumor cell challenge and a significant survival advantage in tumor-bearing mice. These results obtained in this rapidly lethal tumor model suggest that DCs transduced with TAA may be useful for tumor immunotherapy and underscore the importance of the simultaneous delivery of T cell help in the development of Ag-specific cytotoxic T-cells.
Collapse
Affiliation(s)
- Marijke De Veerman
- *Laboratory of Physiology, Medical School of Vrije Universiteit Brussel, Brussels, Belgium; and
| | - Carlo Heirman
- *Laboratory of Physiology, Medical School of Vrije Universiteit Brussel, Brussels, Belgium; and
| | - Sonja Van Meirvenne
- *Laboratory of Physiology, Medical School of Vrije Universiteit Brussel, Brussels, Belgium; and
| | - Sophie Devos
- *Laboratory of Physiology, Medical School of Vrije Universiteit Brussel, Brussels, Belgium; and
| | - Jurgen Corthals
- *Laboratory of Physiology, Medical School of Vrije Universiteit Brussel, Brussels, Belgium; and
| | - Muriel Moser
- †Department of Molecular Biology, Université Libre de Bruxelles, Rhode-St-Genèse, Belgium
| | - Kris Thielemans
- *Laboratory of Physiology, Medical School of Vrije Universiteit Brussel, Brussels, Belgium; and
| |
Collapse
|
48
|
Luykx-de Bakker SA, de Gruijl TD, Scheper RJ, Wagstaff J, Pinedo HM. Dendritic cells: a novel therapeutic modality. Ann Oncol 1999; 10:21-7. [PMID: 10076717 DOI: 10.1023/a:1008349920664] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- S A Luykx-de Bakker
- Department of Medical Oncology, Free University Hospital, Amsterdam, The Netherlands
| | | | | | | | | |
Collapse
|
49
|
Abstract
Recently, cancer immunotherapy has emerged as a therapeutic option for the management of cancer patients. This is based on the fact that our immune system, once activated, is capable of developing specific immunity against neoplastic but not normal cells. Increasing evidence suggests that cell-mediated immunity, particularly T-cell-mediated immunity, is important for the control of tumor cells. Several experimental vaccine strategies have been developed to enhance cell-mediated immunity against tumors. Some of these tumor vaccines have generated promising results in murine tumor systems. In addition, several phase I/II clinical trials using these vaccine strategies have shown extremely encouraging results in patients. In this review, we will discuss many of these promising cancer vaccine strategies. We will pay particular attention to the strategies employing dendritic cells, the central player for tumor vaccine development.
Collapse
Affiliation(s)
- C H Chen
- Department of Internal Medicine, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan
| | | |
Collapse
|
50
|
Antileukemic HLA-Restricted T-Cell Clones Generated With Naturally Processed Peptides Eluted From Acute Myeloblastic Leukemia Blasts. Blood 1998. [DOI: 10.1182/blood.v92.1.19.413a40_19_24] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Recent studies have shown that transfusions of HLA-compatible donor lymphocytes may induce complete remission in marrow-grafted patients with relapses of acute myeloblastic leukemia (AML). We investigated the in vitro generation of antileukemia T-cell clones obtained from the peripheral blood mononuclear cells of a partially HLA-compatible donor (HLA-A2 and B7 molecules in common with the leukemic blasts) after stimulation with a pool of naturally processed peptides extracted from leukemic blast cells collected at diagnosis from a patient with hyperleucocytosis AML. We recovered a significant quantity of peptides that bound to the HLA-A2 or HLA-B7 molecules that were able to induce cytolytic T-lymphocyte (CTL) lines and clones specific for the eluted AML peptides and restricted to the HLA-A2 or B7 molecules. Such CTL line did not recognize the patient's nonleukemic cells, and one clone was able to interact with the leukemic blasts from which the naturally processed peptides had been eluted. Such T-cell clones might provide a rationale for the development of adoptive immunotherapy and could be used to improve the efficiency of HLA-compatible T-lymphocyte transfusions and the graft-versus-leukemia response in patients with AML.
Collapse
|