1
|
Development of Cancer Immunotherapies. Cancer Treat Res 2022; 183:1-48. [PMID: 35551655 DOI: 10.1007/978-3-030-96376-7_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Cancer immunotherapy, or the utilization of components of the immune system to target and eliminate cancer, has become a highly active area of research in the past several decades and a common treatment strategy for several cancer types. The concept of harnessing the immune system for this purpose originated over 100 years ago when a physician by the name of William Coley successfully treated several of his cancer patients with a combination of live and attenuated bacteria, later known as "Coley's Toxins", after observing a subset of prior patients enter remission following their diagnosis with the common bacterial infection, erysipelas. However, it was not until late in the twentieth century that cancer immunotherapies were developed for widespread use, thereby transforming the treatment landscape of numerous cancer types. Pivotal studies elucidating molecular and cellular functions of immune cells, such as the discovery of IL-2 and production of monoclonal antibodies, fostered the development of novel techniques for studying the immune system and ultimately the development and approval of several cancer immunotherapies by the United States Food and Drug Association in the 1980s and 1990s, including the tuberculosis vaccine-Bacillus Calmette-Guérin, IL-2, and the CD20-targeting monoclonal antibody. Approval of the first therapeutic cancer vaccine, Sipuleucel-T, for the treatment of metastatic castration-resistant prostate cancer and the groundbreaking success and approval of immune checkpoint inhibitors and chimeric antigen receptor T cell therapy in the last decade, have driven an explosion of interest in and pursuit of novel cancer immunotherapy strategies. A broad range of modalities ranging from antibodies to adoptive T cell therapies is under investigation for the generalized treatment of a broad spectrum of cancers as well as personalized medicine. This chapter will focus on the recent advances, current strategies, and future outlook of immunotherapy development for the treatment of cancer.
Collapse
|
2
|
Himes BT, Geiger PA, Ayasoufi K, Bhargav AG, Brown DA, Parney IF. Immunosuppression in Glioblastoma: Current Understanding and Therapeutic Implications. Front Oncol 2021; 11:770561. [PMID: 34778089 PMCID: PMC8581618 DOI: 10.3389/fonc.2021.770561] [Citation(s) in RCA: 99] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Accepted: 09/24/2021] [Indexed: 12/12/2022] Open
Abstract
Glioblastoma (GBM) is the most common primary brain tumor in adults an carries and carries a terrible prognosis. The current regiment of surgical resection, radiation, and chemotherapy has remained largely unchanged in recent years as new therapeutic approaches have struggled to demonstrate benefit. One of the most challenging hurdles to overcome in developing novel treatments is the profound immune suppression found in many GBM patients. This limits the utility of all manner of immunotherapeutic agents, which have revolutionized the treatment of a number of cancers in recent years, but have failed to show similar benefit in GBM therapy. Understanding the mechanisms of tumor-mediated immune suppression in GBM is critical to the development of effective novel therapies, and reversal of this effect may prove key to effective immunotherapy for GBM. In this review, we discuss the current understanding of tumor-mediated immune suppression in GBM in both the local tumor microenvironment and systemically. We also discuss the effects of current GBM therapy on the immune system. We specifically explore some of the downstream effectors of tumor-driven immune suppression, particularly myeloid-derived suppressor cells (MDSCs) and other immunosuppressive monocytes, and the manner by which GBM induces their formation, with particular attention to the role of GBM-derived extracellular vesicles (EVs). Lastly, we briefly review the current state of immunotherapy for GBM and discuss additional hurdles to overcome identification and implementation of effective therapeutic strategies.
Collapse
Affiliation(s)
- Benjamin T Himes
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, United States
| | - Philipp A Geiger
- Department of Neurosurgery, University Hospital Innsbruck, Tirol, Austria
| | | | - Adip G Bhargav
- Department of Neurosurgery, University of Kansas, Kansas City, KS, United States
| | - Desmond A Brown
- Surgical Neurology Branch, National Institutes of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States
| | - Ian F Parney
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, United States.,Department of Immunology, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
3
|
Hamblin MR, Abrahamse H. Factors Affecting Photodynamic Therapy and Anti-Tumor Immune Response. Anticancer Agents Med Chem 2021; 21:123-136. [PMID: 32188394 DOI: 10.2174/1871520620666200318101037] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 01/15/2020] [Accepted: 01/29/2020] [Indexed: 11/22/2022]
Abstract
Photodynamic Therapy (PDT) is a cancer therapy involving the systemic injection of a Photosensitizer (PS) that localizes to some extent in a tumor. After an appropriate time (ranging from minutes to days), the tumor is irradiated with red or near-infrared light either as a surface spot or by interstitial optical fibers. The PS is excited by the light to form a long-lived triplet state that can react with ambient oxygen to produce Reactive Oxygen Species (ROS) such as singlet oxygen and/or hydroxyl radicals, that kill tumor cells, destroy tumor blood vessels, and lead to tumor regression and necrosis. It has long been realized that in some cases, PDT can also stimulate the host immune system, leading to a systemic anti-tumor immune response that can also destroy distant metastases and guard against tumor recurrence. The present paper aims to cover some of the factors that can affect the likelihood and efficiency of this immune response. The structure of the PS, drug-light interval, rate of light delivery, mode of cancer cell death, expression of tumor-associated antigens, and combinations of PDT with various adjuvants all can play a role in stimulating the host immune system. Considering the recent revolution in tumor immunotherapy triggered by the success of checkpoint inhibitors, it appears that the time is ripe for PDT to be investigated in combination with other approaches in clinical scenarios.
Collapse
Affiliation(s)
- Michael R Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein 2028, South Africa
| | - Heidi Abrahamse
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein 2028, South Africa
| |
Collapse
|
4
|
Abstract
Non-small cell lung cancer (NSCLC) unfortunately carries a very poor prognosis. Patients usually do not become symptomatic, and therefore do not seek treatment, until the cancer is advanced and it is too late to employ curative treatment options. New therapeutic options are urgently needed for NSCLC, because even current targeted therapies cure very few patients. Active immunotherapy is an option that is gaining more attention. A delicate and complex interplay exists between the tumor and the immune system. Solid tumors utilize a variety of mechanisms to evade immune detection. However, if the immune system can be stimulated to recognize the tumor as foreign, tumor cells can be specifically eliminated with little systemic toxicity. A number of vaccines designed to boost immunity against NSCLC are currently undergoing investigation in phase III clinical trials. Belagenpumatucel-L, an allogeneic cell vaccine that decreases transforming growth factor (TGF-β) in the tumor microenvironment, releases the immune suppression caused by the tumor and it has shown efficacy in a wide array of patients with advanced NSCLC. Melanoma-associated antigen A3 (MAGE-A3), an antigen-based vaccine, has shown promising results in MAGE-A3+ NSCLC patients who have undergone complete surgical resection. L-BLP25 and TG4010 are both antigenic vaccines that target the Mucin-1 protein (MUC-1), a proto-oncogene that is commonly mutated in solid tumors. CIMAVax is a recombinant human epidermal growth factor (EGF) vaccine that induces anti-EGF antibody production and prevents EGF from binding to its receptor. These vaccines may significantly improve survival and quality of life for patients with an otherwise dismal NSCLC prognosis. This review is intended to give an overview of the current data and the most promising studies of active immunotherapy for NSCLC.
Collapse
Affiliation(s)
- Francisco Socola
- Division of Hematology/Oncology, Sylvester Comprehensive Cancer Center, University of Miami Leonard M Miller School of Medicine, Miami, Florida, USA
| | - Naomi Scherfenberg
- University of Miami Leonard M Miller School of Medicine, Miami, Florida, USA
| | - Luis E Raez
- Thoracic Oncology Program, Memorial Cancer Institute, Memorial Health Care System, Pembroke Pines, Florida, USA
| |
Collapse
|
5
|
Gonzalez G, Diaz-Miqueli A, Crombet T, Raez LE, Lage A. Current Algorithm for Treatment of Advanced NSCLC Patients: How to Include Active Immunotherapy? ACTA ACUST UNITED AC 2013. [DOI: 10.4236/jct.2013.48a010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
6
|
De Pas T, Giovannini M, Rescigno M, Catania C, Toffalorio F, Spitaleri G, Delmonte A, Barberis M, Spaggiari L, Solli P, Veronesi G, De Braud F. Vaccines in non-small cell lung cancer: rationale, combination strategies and update on clinical trials. Crit Rev Oncol Hematol 2012; 83:432-43. [PMID: 22366114 DOI: 10.1016/j.critrevonc.2011.12.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2011] [Revised: 12/20/2011] [Accepted: 12/22/2011] [Indexed: 12/20/2022] Open
Abstract
Non-small cell lung cancer (NSCLC) remains the leading cause of cancer related mortality worldwide and despite some advances in therapy the overall prognosis remains disappointing. New therapeutic approaches like vaccination have been proposed and several clinical trials are ongoing. Many tumor antigens have been identified so far and specific tumor vaccines targeting these antigens have been developed. Even if the ideal setting for vaccine therapy might be the adjuvant one, vaccines seem to be potentially beneficial also in advanced disease and combination therapy could be a promising treatment option. In the advanced setting anti-MUC-1 vaccine (belagenpumatucel) and anti-TGF-β(2) vaccine (BPL-25) have entered in phase III trials as maintenance therapy after first line chemotherapy. In the adjuvant setting the most relevant and promising vaccines are directed against MAGE-A3 and PRAME, respectively. We will review the key points for effective active immunotherapies and combination therapies, giving an update on the most promising vaccines developed in NSCLC.
Collapse
Affiliation(s)
- Tommaso De Pas
- Medical Oncology Unit of Respiratory Tract and Sarcomas, New Drugs Development Division, European Institute of Oncology, Milan, Italy.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
7
|
Hendrickson RC, Cicinnati VR, Albers A, Dworacki G, Gambotto A, Pagliano O, Tüting T, Mayordomo JI, Visus C, Appella E, Shabanowitz J, Hunt DF, DeLeo AB. Identification of a 17beta-hydroxysteroid dehydrogenase type 12 pseudogene as the source of a highly restricted BALB/c Meth A tumor rejection peptide. Cancer Immunol Immunother 2010; 59:113-24. [PMID: 19562340 PMCID: PMC2855844 DOI: 10.1007/s00262-009-0730-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2009] [Accepted: 06/03/2009] [Indexed: 12/23/2022]
Abstract
Mass spectrometric analysis identified the peptide recognized by a cytotoxic T lymphocyte (CTL) specific for the chemically induced BALB/c Meth A sarcoma as derived from a 17beta-hydroxysteroid dehydrogenase type 12 (Hsd17b12) pseudogene present in the BALB/c genome, but only expressed in Meth A sarcoma. The sequence of the peptide is TYDKIKTGL and corresponds to Hsd17b12(114-122) with threonine instead of isoleucine at codon 114 and is designated Hsd17b12(114T). Immunization of mice with an Hsd17b12(114T) peptide-pulsed dendritic cell-based vaccine or a non-viral plasmid construct expressing the Hsd17b12(114T) peptide protected the mice from lethal Meth A tumor challenge in tumor rejection assays. A Hsd17b12(114-122) peptide-pulsed vaccine was ineffective in inducing resistance in mice to Meth A sarcoma. These results confirm the immunogenicity of the identified tumor peptide, as well as demonstrate the efficacies of these vaccine vehicles. These findings suggest that the role of the human homolog of Hsd17b12, HSD17B12, as a potential human tumor antigen be explored.
Collapse
Affiliation(s)
| | - Vito R. Cicinnati
- Division of Basic Research, Hillman Cancer Center, University of Pittsburgh Cancer Institute, Pittsburgh, PA 15213-2582 USA
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213 USA
| | - Andreas Albers
- Division of Basic Research, Hillman Cancer Center, University of Pittsburgh Cancer Institute, Pittsburgh, PA 15213-2582 USA
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213 USA
| | - Grzegorz Dworacki
- Division of Basic Research, Hillman Cancer Center, University of Pittsburgh Cancer Institute, Pittsburgh, PA 15213-2582 USA
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213 USA
| | - Andrea Gambotto
- Department of Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213 USA
| | - Ornella Pagliano
- Division of Basic Research, Hillman Cancer Center, University of Pittsburgh Cancer Institute, Pittsburgh, PA 15213-2582 USA
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213 USA
| | - Thomas Tüting
- Division of Basic Research, Hillman Cancer Center, University of Pittsburgh Cancer Institute, Pittsburgh, PA 15213-2582 USA
- Department of Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213 USA
| | - Jose I. Mayordomo
- Division of Basic Research, Hillman Cancer Center, University of Pittsburgh Cancer Institute, Pittsburgh, PA 15213-2582 USA
- Department of Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213 USA
| | - Carmen Visus
- Division of Basic Research, Hillman Cancer Center, University of Pittsburgh Cancer Institute, Pittsburgh, PA 15213-2582 USA
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213 USA
| | - Ettore Appella
- Laboratory of Cell Biology, National Cancer Institute, Bethesda, MD 20892 USA
| | - Jeffrey Shabanowitz
- Department of Chemistry, University of Virginia, Charlottesville, VA, 22901 USA
| | - Donald F. Hunt
- Department of Chemistry, University of Virginia, Charlottesville, VA, 22901 USA
- Department of Pathology, University of Virginia, Charlottesville, VA, 22901 USA
| | - Albert B. DeLeo
- Division of Basic Research, Hillman Cancer Center, University of Pittsburgh Cancer Institute, Pittsburgh, PA 15213-2582 USA
- Division of Basic Research, Hillman Cancer Center, Research Pavilion, University of Pittsburgh Cancer Institute, Pittsburgh, PA 15213-2582 USA
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213 USA
| |
Collapse
|
8
|
Kawanishi T, Ikeda-Dantsuji Y, Nagayama A. Effects of two basidiomycete species on interleukin 1 and interleukin 2 production by macrophage and T cell lines. Immunobiology 2009; 215:516-20. [PMID: 19913939 DOI: 10.1016/j.imbio.2009.10.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2009] [Accepted: 10/18/2009] [Indexed: 11/27/2022]
Abstract
Two basidiomycete species, Lentinus edodes mycelia (LEM) and Cordyceps sinensis (CS) were examined for induction of cytokines in murine macrophage cell line R309 (R309) and T cell line LBRM-33 1A5 (1A5). When lipopolysaccharide (LPS)-activated R309 were exposed to the extracts of basidiomycetes, R309 induced significant levels of interleukin 1 (IL-1). Interleukin 2 (IL-2) induction was recognized in 1A5 cultures in the presence of IL-1 and phytohemagglutinin (PHA). However, no enhancement of IL-2 production by these basidiomycetes was discerned in 1A5 cultures with IL-1 and PHA, i.e., direct action of basidiomycetes was not found on IL-2 production of 1A5. PHA-stimulated 1A5 exposed to basidiomycetes induced IL-2 without IL-1 when co-cultured with LPS-activated R309 as a source of IL-1. Effects of basidiomycetes on IL-2 production in 1A5 seemed to be caused through their action on macrophages. The induction of IL-2, Th1 type cytokine in T lymphocyte, is a significant finding since basidiomycetes, taken as a dietary supplement for immuno-suppressed patients, especially cancer patients, would be helpful in improving their immune activity against cancer.
Collapse
Affiliation(s)
- Takashi Kawanishi
- Department of Microbiology and Immunology, Fukuoka University School of Medicine, Jonan-ku, Fukuoka, Japan
| | | | | |
Collapse
|
9
|
Brichard VG, Lejeune D. Cancer immunotherapy targeting tumour-specific antigens: towards a new therapy for minimal residual disease. Expert Opin Biol Ther 2008; 8:951-68. [PMID: 18549325 DOI: 10.1517/14712598.8.7.951] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
BACKGROUND Clinical investigation of cancer immunotherapy has been very active and several approaches have been evaluated in Phase III trials. In particular, the characterisation at the molecular level of tumour-specific antigens, together with expert knowledge from GSK Biologicals in recombinant protein manufacturing and immunological Adjuvant Systems, has led the company to develop Antigen-Specific Cancer Immunotherapeutic (ASCI). OBJECTIVE/METHODS This paper reviews the different cancer immunotherapy approaches that have reached Phase III clinical development. A special attention is given to GSK's ASCI approach. CONCLUSION Based on encouraging data in a double-blind Phase II trial in non-small-cell lung cancer, the selection of the most suitable adjuvant system in melanoma and the choice of the adequate clinical setting for the clinical development of cancer immunotherapy, the ASCI approach offers the perspective that the long quest towards a new cancer treatment approach is about to succeed.
Collapse
Affiliation(s)
- Vincent G Brichard
- GlaxoSmithKline Biologicals, Cancer Immunotherapeutics R&D, Rue de l'Institut, 89, B-1330 Rixensart, Belgium.
| | | |
Collapse
|
10
|
Brichard VG, Lejeune D. GSK's antigen-specific cancer immunotherapy programme: pilot results leading to Phase III clinical development. Vaccine 2008; 25 Suppl 2:B61-71. [PMID: 17916463 DOI: 10.1016/j.vaccine.2007.06.038] [Citation(s) in RCA: 118] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2007] [Revised: 05/31/2007] [Accepted: 06/11/2007] [Indexed: 02/07/2023]
Abstract
From the first evidence that the immune system could recognize tumors, different types of tumor antigens have been identified and deeply characterized. Several different approaches aimed at targeting these antigens have already been the subject of clinical studies. In this field, the GSK Biologicals' approach relying on recombinant proteins combined with an immunological Adjuvant System in a specific clinical setting, has entertained hopes of developing a new class of well tolerated anti-cancer therapy. This methodology led to promising advances with MAGE-A3 immunotherapy in NSCLC and has the potential to be applied to all tumor types.
Collapse
Affiliation(s)
- Vincent G Brichard
- Cancer Immunotherapeutics R&D, GlaxoSmithKline Biologicals, Rue de l'Institut, Rixensart, Belgium.
| | | |
Collapse
|
11
|
López M, Aguilera R, Pérez C, Mendoza-Naranjo A, Pereda C, Ramirez M, Ferrada C, Aguillón JC, Salazar-Onfray F. The role of regulatory T lymphocytes in the induced immune response mediated by biological vaccines. Immunobiology 2006; 211:127-36. [PMID: 16446177 DOI: 10.1016/j.imbio.2005.11.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2005] [Accepted: 11/17/2005] [Indexed: 12/20/2022]
Abstract
Immunotherapy has become a novel therapeutic alternative for various kinds of tumours. Recently, we have finalized the first phase I clinical study in Chile for the treatment of advanced malignant melanoma, using dendritic cells (DCs) loaded with allogeneic melanoma cell lysate. This study included 20 patients and the obtained results, pioneer in Latin America, showed that DC-based immunotherapy is innocuous, even provided in combination with IL-2. In addition, immunological responses were detected in 50% of the treated patients, establishing a positive correlation between the delayed type hypersensitivity (DTH) reaction, which indicates induction of in vivo immunological memory, and patients surviving. Nevertheless, objective clinical responses in vaccinated patients are still insufficient. Only sporadic objective metastasis regressions have been registered and an important proportion of the treated patients did not respond, or their responses were weak. Several strategies have been described to be used by tumours to escape from the immune response. Actually, we have demonstrated that IL-10 inhibits antigen presentation in melanoma, reducing tumour sensitivity to melanoma-specific cytotoxic T lymphocytes (CTLs). Regulation of the immunological response by inhibitory cells could be another possible cause of clinical unresponsiveness. Lately, the existence of subpopulations of regulatory T lymphocytes (RTL) able to limit the immune response in a specific form has been established, specially inhibiting the proliferation and activity of CD4+ and CD8+ effector T lymphocytes. These cellular subpopulations, mostly CD4+/CD25+/Foxp3+ T lymphocytes (Treg) of thymic origin, or TR1 lymphocytes able to release IL-10, and tumour growth factor beta (TGF-beta) producing TH3 lymphocytes, would be accumulated in the body during tumour growth, inhibiting the immune response. In relation to RTL and cancer, evidence indicates that Treg cell numbers are increased in blood and other tissues in different types of cancer. Additionally, it has been demonstrated that in patients with refractory metastatic melanoma, the adoptive transference of anti-tumour CD8+ T lymphocytes after non-myeloablative chemotherapy was able to induce important tumour regressions that would be due to elimination of RTL populations. Additionally, chemotherapeutical drugs like decarbazine, besides their effect on tumour proliferation, also have an immunosuppressive effect on T lymphocyte populations, as well as on accumulated RTL. In this article, a novel strategy for the study of RTL is proposed, including potential therapeutic innovations, which is being pioneered in current clinical trials.
Collapse
Affiliation(s)
- Mercedes López
- Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Uenaka A, Nakayama E. Murine leukemia RL male 1 and sarcoma Meth A antigens recognized by cytotoxic T lymphocytes (CTL). Cancer Sci 2003; 94:931-6. [PMID: 14611667 PMCID: PMC11160269 DOI: 10.1111/j.1349-7006.2003.tb01380.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2003] [Revised: 09/02/2003] [Accepted: 09/05/2003] [Indexed: 11/29/2022] Open
Abstract
Peptide elution and expression cloning methods have been used to identify T cell-recognized antigens for which no molecular information is available. We identified a unique tumor antigen peptide pRL1a, IPGLPLSL that is recognized by CTL on BALB/c RL male 1 leukemia by peptide elution. The sequence of the peptide corresponded to the normally untranslated 5' region of akt. Cytotoxicity was generated in BALB/c spleen cells by in vivo and in vitro sensitization with pRL1a peptide in the form of multiple antigen peptide (MAP), but not the original form. pRL1a MAP immunization had a significant growth-inhibitory effect. pRL1a MAP was mostly internalized into the endosomal compartment of antigen-presenting cells, leaked to the cytosol, and degraded, and the pRL1a peptide produced was presented through the MHC class I pathway. In vivo depletion of CD4 T cells from tumor-inoculated BALB/c mice caused RL male 1 regression. Overexpression of the RLakt molecule seemed to induce CD4 immunoregulatory cells, which resulted in progressive RL male 1 growth in BALB/c mice. In vivo administration of anti-CD25 mAb (PC61) caused regression of RL male 1, suggesting that CD4(+) CD25(+) immunoregulatory cells were involved in the tumor growth. Recently, we improved the sensitivity and the efficacy of T cell antigen cloning from cDNA expression libraries by using large- and small-scale ELISPOT assays. Using the IFN-gamma ELISPOT method, we obtained a cDNA clone S35 of 937 bp recognized by AT-1 CTL on BALB/c Meth A sarcoma. S35 was a part of the retinoic acid-regulated nuclear matrix-associated protein (ramp). AT-1 CTL recognized the peptide LGAEAIFRL, which was derived from a newly created open reading frame due to the exon 14 extension.
Collapse
Affiliation(s)
- Akiko Uenaka
- Department of Immunology, Okayama University Graduate School of Medicine and Dentistry, Okayama 700-8558.
| | | |
Collapse
|
13
|
Li J, Li W, Liang S, Cai D, Kieny MP, Jacob L, Linnenbach A, Abramczuk JW, Bender H, Sproesser K, Swoboda R, Somasundaram R, Guerry D, Herlyn D. Recombinant CD63/ME491/neuroglandular/NKI/C-3 antigen inhibits growth of established tumors in transgenic mice. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2003; 171:2922-9. [PMID: 12960315 DOI: 10.4049/jimmunol.171.6.2922] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Attempts to vaccinate against tumors can be hindered by the induction of immunological tolerance to the target Ag as a result of Ag expression on normal tissues. In this study, we find that transgenic mice expressing the melanoma-associated Ag CD63/ME491/neuroglandular/NKI/C-3 on their normal tissues do, in fact, exhibit immunological tolerance to the Ag, recapitulating the conditions in cancer patients. In these mice, growth of murine melanoma cells expressing the Ag after gene transfer was inhibited by immunization with Ag-expressing recombinant vaccinia virus combined with IL-2, but not by immunization with the protein alone, anti-idiotypic Abs, or irradiated tumor cells. The effect of the recombinant virus was demonstrated both for nonestablished and established tumors. Infiltration with both CD4(+) and CD8(+) T lymphocytes was significantly more extensive in tumors from experimental mice than in tumors from control mice. MHC class I-positive, but not class I-negative, tumors were inhibited by the vaccine, suggesting that MHC class I-restricted T lymphocytes play a role in the antitumor effects. Abs did not appear to be involved in the vaccine effects. CD63 was immunogenic in 2 of 13 melanoma patients, pointing to the potential of this Ag, combined with IL-2, as a vaccine for melanoma patients.
Collapse
MESH Headings
- Animals
- Antibodies, Neoplasm/biosynthesis
- Antigens, CD/administration & dosage
- Antigens, CD/biosynthesis
- Antigens, CD/genetics
- Antigens, CD/immunology
- Antigens, Neoplasm/administration & dosage
- Antigens, Neoplasm/genetics
- Antineoplastic Agents/administration & dosage
- Antineoplastic Agents/immunology
- Antineoplastic Agents/metabolism
- Cancer Vaccines/administration & dosage
- Cancer Vaccines/biosynthesis
- Cancer Vaccines/genetics
- Cancer Vaccines/immunology
- Female
- Growth Inhibitors/administration & dosage
- Growth Inhibitors/genetics
- Hemadsorption
- Humans
- Immune Tolerance/genetics
- Immunity, Cellular/genetics
- Interleukin-2/administration & dosage
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/metabolism
- Melanoma/immunology
- Melanoma, Experimental/immunology
- Melanoma, Experimental/pathology
- Melanoma, Experimental/prevention & control
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Organ Specificity/genetics
- Organ Specificity/immunology
- Platelet Membrane Glycoproteins/administration & dosage
- Platelet Membrane Glycoproteins/biosynthesis
- Platelet Membrane Glycoproteins/genetics
- Platelet Membrane Glycoproteins/immunology
- Species Specificity
- Tetraspanin 30
- Transfection
- Tumor Cells, Cultured
- Vaccines, Synthetic/administration & dosage
- Vaccines, Synthetic/biosynthesis
- Vaccines, Synthetic/immunology
Collapse
Affiliation(s)
- Jian Li
- The Wistar Institute, Philadelphia, PA 19104, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Uenaka A, Hirano Y, Hata H, Win S, Aji T, Tanaka M, Ono T, Skipper JCA, Shimizu K, Nakayama E. Cryptic CTL epitope on a murine sarcoma Meth A generated by exon extension as a novel mechanism. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2003; 170:4862-8. [PMID: 12707369 DOI: 10.4049/jimmunol.170.9.4862] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Using the recently developed ELISPOT cloning methodology, we obtained cDNA clone S35 coding for the Ag epitope recognized by a murine sarcoma Meth A-specific CTL clone AT-1. Analysis of truncated S35 constructs and overlapping peptides revealed that the peptide epitope was LGAEAIFRL. AT-1 CTL lysed peptide-pulsed CMS8 cells at a nanomolar concentration, and the peptide strongly stimulated IFN-gamma production in AT-1 CTL. Sequence homology indicated that the S35 was derived from a mouse homologue of human retinoic acid-regulated nuclear matrix-associated protein (ramp). The ramp gene consisted of 15 exons. The majority of the ramp mRNA was the transcript normally spliced between exons 14 and 15, but a minor population of mRNA with an extended exon 14 was also present in Meth A cells. The epitope was derived from the newly created open reading frame, which resulted from extension of exon 14 after splicing of the adjacent intronic sequence.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Antigens, Neoplasm/biosynthesis
- Antigens, Neoplasm/genetics
- Antigens, Neoplasm/isolation & purification
- Antigens, Neoplasm/metabolism
- Base Sequence
- Carrier Proteins/biosynthesis
- Carrier Proteins/genetics
- Carrier Proteins/isolation & purification
- Carrier Proteins/metabolism
- Clone Cells
- Cloning, Molecular
- Cytotoxicity Tests, Immunologic
- DNA-Binding Proteins/biosynthesis
- DNA-Binding Proteins/genetics
- DNA-Binding Proteins/isolation & purification
- DNA-Binding Proteins/metabolism
- Enzyme-Linked Immunosorbent Assay
- Epitopes, T-Lymphocyte/biosynthesis
- Epitopes, T-Lymphocyte/genetics
- Epitopes, T-Lymphocyte/isolation & purification
- Epitopes, T-Lymphocyte/metabolism
- Exons/genetics
- Exons/immunology
- Gene Library
- Humans
- Interferon-gamma/analysis
- Methylcholanthrene
- Mice
- Mice, Inbred BALB C
- Mice, Inbred DBA
- Molecular Sequence Data
- Sarcoma, Experimental/chemically induced
- Sarcoma, Experimental/immunology
- T-Lymphocytes, Cytotoxic/immunology
- Transcription Factors
Collapse
Affiliation(s)
- Akiko Uenaka
- Department of Immunology, Okayama University Graduate School of Medicine and Dentistry, Okayama, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Paster W, Kalat M, Zehetner M, Schweighoffer T. Structural elements of a protein antigen determine immunogenicity of the embedded MHC class I-restricted T cell epitope. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2002; 169:2937-46. [PMID: 12218107 DOI: 10.4049/jimmunol.169.6.2937] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Substantial effort has been invested into optimization of vector structure, DNA formulation, or delivery methods to increase the effectiveness of DNA vaccines. In contrast, it has been only insufficiently explored how the higher order structure of an antigenic protein influences immunogenicity of embedded epitopes in vivo. Potent CD8+ T cell responses specific for a single immunogenic epitope are induced upon electrovaccination with plasmid DNA encoding the full-length heavy chain of the human HLA-Cw3 molecule. Contrary to expectations, a minimal construct, which provoked a substantial release of IFN-gamma from specific CTLs in vitro, did not induce a significant response in vivo. Systematically altered variants of the Cw3 molecule were thus tested both in vivo and in vitro to determine which structural parts are responsible for this discrepancy. In complementation experiments the participation of trans-acting helper epitopes was ruled out. Successive C-terminal truncations, human/mouse domain swap variants, and subdomain modifications defined the alpha3 region of the HLA heavy chain and membrane anchoring as critical elements. Based on these data, refined minimal constructs were engineered that triggered very high in vivo responses. The most advanced variant consisted only of an adenoviral leader, antigenic epitope, alpha3 domain, and 16 aa of the transmembrane domain. When a tumor Ag epitope was incorporated into one of these high performer minimal constructs, protection against melanoma metastases was attained upon vaccination. Thus, structural elements of the Ag can dominantly influence immunogenicity in vivo. These elements can also markedly improve the immunogenicity of unrelated Ags and may form the basis of a new generation of DNA vaccines.
Collapse
MESH Headings
- Animals
- Antigens, Neoplasm/administration & dosage
- Antigens, Neoplasm/chemistry
- Antigens, Neoplasm/immunology
- CD8-Positive T-Lymphocytes/immunology
- Cancer Vaccines/administration & dosage
- Cancer Vaccines/chemical synthesis
- Cancer Vaccines/immunology
- Cell Membrane/chemistry
- Cell Membrane/genetics
- Cell Membrane/immunology
- Cytotoxicity, Immunologic/genetics
- Epitopes, T-Lymphocyte/biosynthesis
- Epitopes, T-Lymphocyte/genetics
- Female
- Genetic Complementation Test
- H-2 Antigens/administration & dosage
- H-2 Antigens/chemistry
- H-2 Antigens/immunology
- HLA-C Antigens/administration & dosage
- HLA-C Antigens/chemistry
- HLA-C Antigens/genetics
- HLA-C Antigens/immunology
- Lymphocyte Activation/genetics
- Melanoma, Experimental/chemistry
- Melanoma, Experimental/immunology
- Melanoma, Experimental/prevention & control
- Mice
- Mice, Inbred C57BL
- Mice, Inbred DBA
- Peptide Fragments/administration & dosage
- Peptide Fragments/genetics
- Peptide Fragments/immunology
- Plasmids/administration & dosage
- Plasmids/chemical synthesis
- Plasmids/immunology
- Protein Engineering/methods
- Protein Structure, Tertiary/genetics
- Sequence Deletion
- Structure-Activity Relationship
- T-Lymphocytes, Cytotoxic/immunology
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- Wolfgang Paster
- Department of NBE Discovery, Boehringer Ingelheim Austria GmbH, Vienna, Austria.
| | | | | | | |
Collapse
|
16
|
Abstract
Gene transfer technology has the potential to revolutionize cancer treatment. Developments in molecular biology, genetics, genomics, stem cell technology, virology, bioengineering, and immunology are accelerating the pace of innovation and movement from the laboratory bench to the clinical arena. Pancreatic adenocarcinoma, with its particularly poor prognosis and lack of effective traditional therapy for most patients, is an area where gene transfer and immunotherapy have a maximal opportunity to demonstrate efficacy. In this review, we have discussed current preclinical and clinical investigation of gene transfer technology for pancreatic cancer. We have emphasized that the many strategies under investigation for cancer gene therapy can be classified into two major categories. The first category of therapies rely on the transduction of cells other than tumor cells, or the limited transduction of tumor tissue. These therapies, which do not require efficient gene transfer, generally lead to systemic biological effects (e.g., systemic antitumor immunity, inhibition of tumor angiogenesis, etc) and therefore the effects of limited gene transfer are biologically "amplified." The second category of gene transfer strategies requires the delivery of therapeutic genetic material to all or most tumor cells. While these elegant approaches are based on state-of-the-art advances in our understanding of the molecular biology of cancer, they suffer from the current inadequacies of gene transfer technology. At least in the short term, it is very likely that success in pancreatic cancer gene therapy will involve therapies that require only the limited transduction of cells. The time-worn surgical maxim, "Do what's easy first," certainly applies here.
Collapse
Affiliation(s)
- Jennifer F Tseng
- Division of Molecular Medicine, Children's Hospital, Department of Genetics, Harvard Medical School, Enders 861, 320 Longwood Avenue, Boston, MA 02115, USA
| | | |
Collapse
|
17
|
Bhattachary R, Bukkapatnam R, Prawoko I, Soto J, Morgan M, Salup RR. Efficacy of vaccination with plasmid DNA encoding for HER2/neu or HER2/neu-eGFP fusion protein against prostate cancer in rats. Int Immunopharmacol 2002; 2:783-96. [PMID: 12095169 DOI: 10.1016/s1567-5769(02)00017-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Despite early diagnosis and improved therapy, 31,500 men will die from prostate cancer (PC) this year. The HER2/neu oncoprotein is an important effector of cell growth found in the majority of high-grade prostatic tumors and is capable of rendering immunogenicity. The antigenicity of this oncoprotein might prove useful in the development of PC vaccines. Our goal is to prove the principle that a single DNA vaccine can provide reliable immunity against PC in the MatLyLu (MLL) translational tumor model. The parental rat MatLyLu PC cell line expresses low to moderate levels of the rat neu protein. To simulate in vivo human PC, MatLyLu cells were transfected with a truncated sequence of human HER2/neu cDNA cloned into the pCI-neo vector. This HER2/neu cDNA sequence encodes the first 433 amino acids of the extracellular domain (ECD). MatLyLu cells were also transfected with the same HER2/neu cDNA sequence cloned into the N1-terminal sequence of EGFP reporter gene to produce a fusion protein. The partial ECD sequence of HER2/neu includes five rat major histocompatibility (MHC)-II-restricted peptides with complete human-to-rat cross-species homology. The HER2/neu protein overexpression was documented by Western Blot analysis, and the expression of fusion protein was monitored by confocal microscopy and fluorimetry. Vaccination with a single injection of HER2/neu cDNA protected 50% of animals against HER2/neu-MatLyLu tumors (P < 0.01). When the tumor cells were engineered to express HER2/neu-EGFP fusion protein, the antitumor immunity was enhanced, as following vaccination with HER2/neu-EGFP cDNA, 80% of these rats rejected HER2/neu-EGFP-MatLyLu (P<0.001). Both vaccines induced HER2/neu-specific antibody titers. Rats vaccinated with EGFP-cDNA rejected 80% of EGFP-MatLyLu tumors and, interestingly, 40% of HER2/neu-MatLyLu tumors. None of the cDNA vaccines induced immunity against parental MatLyLu cells. Our data clearly demonstrate that a single injection of HER2/neu-EGFP cDNA is a very effective vaccine against PC tumors expressing the cognate tumor-associated antigen (TA). The antitumor immunity is significantly more pronounced if the tumors express xenogeneic HER2/neu-EGFP fusion protein as opposed to only the syngeneic HER2/neu oncoprotein. Our data suggests that the HER2/neu-EGFP-MatLyLu tumor is a potential animal tumor model for investigating therapeutic vaccine strategies against PC in vivo and demonstrates the limitations of a cDNA vaccine only encoding for MHC-II-restricted HER2/neu-ECD sequence peptides.
Collapse
|
18
|
Wilkinson RW, Ross EL, Poulsom R, Ilyas M, Straub J, Snary D, Bodmer WF, Mather SJ. Antibody targeting studies in a transgenic murine model of spontaneous colorectal tumors. Proc Natl Acad Sci U S A 2001; 98:10256-60. [PMID: 11517330 PMCID: PMC56948 DOI: 10.1073/pnas.181353498] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Monoclonal antibodies (mAbs) have been used to treat malignancies in humans with varying degrees of success. Progress has been hindered by the lack of suitable animal models, which would ideally consist of immunocompetent animals that are tolerant to tumor-associated antigens. Suitable models would allow the study and optimization of anti-tumor immunotherapy. We describe a murine model for the study of immunotherapy in colorectal cancers. Carcinoembryonic antigen (CEA) is a cell-surface glycoprotein that is expressed on normal human intestinal epithelium and that is overexpressed in intestinal tumors. Mice that are transgenic for the human CEA gene (CEA.Tg) were crossed with multiple intestinal neoplasia (MIN) mice. MIN mice carry a germline APC mutation and are prone to the development of intestinal adenomas. The offspring from the MIN x CEA.Tg cross developed intestinal adenomas that were shown by immunohistochemistry to overexpress CEA. Pharmacokinetic studies by using (125)I-labeled anti-CEA mAb PR1A3 showed rapid localization of antibody to tissues expressing CEA, especially the gastrointestinal tract. Macroscopic and microscopic radioautographic analysis of the gastrointestinal tracts from MIN/CEA.Tg mice indicated that PR1A3 targeted and was retained in tumors at levels higher than in areas of normal gut. These results demonstrate the utility of the MIN/CEA.Tg mouse as a model for the study of anti-CEA immunotherapy and, furthermore, demonstrate the efficiency of tumor localization by PR1A3.
Collapse
Affiliation(s)
- R W Wilkinson
- Applied Development Laboratory, Imperial Cancer Research Technology, Dominion House, 59 Bartholomew Close, London EC1A 7BE, United Kingdom
| | | | | | | | | | | | | | | |
Collapse
|
19
|
Abstract
Cancer vaccines have been extensively tested in animal models, and in humans. Initial studies focused on first generation vaccines based on whole cell preparations or tumor lysates derived from autologous or allogeneic tumors. Clinical studies conducted with such candidate vaccines contributed to establish the feasibility of immunizing cancer patients against their own tumors. Significant clinical benefits were observed, both in terms of long term survival and recurrence rate, in some of these trials. More recently, however, cancer vaccines targeting well-characterized tumor-associated antigens, i.e. molecules selectively or preferentially expressed by cancer cells but not by normal cells, have been designed and tested in humans. Results obtained as of today with these second-generation vaccines suggest that they are safe and that they can elicit humoral and cellular responses against tumor-specific antigens, without inducing unacceptable clinical signs of autoimmunity. Advances in tumor biology and tumor immunity have helped to better understand the mechanisms displayed by a number of tumors to escape host immunity. This bulk of new knowledge will be used to design future cancer vaccines, which will likely target multiple TAAs, presented by different antigen presentation platforms, in association with synthetic adjuvants and/or immunostimulatory cytokines. Lastly, specific tools allowing to assess in a qualitative and quantitative manner immune responses are critically needed in order to establish correlates between clinical and immune responses in patients receiving experimental vaccines.
Collapse
Affiliation(s)
- P Moingeon
- Aventis Pasteur, Campus Mérieux, 1541 Avenue Marcel Mérieux, 69280, Marcy l'Etoile, France.
| |
Collapse
|
20
|
Wagner SN, Wagner C, Lührs P, Weimann TK, Kutil R, Goos M, Stingl G, Schneeberger A. Intracutaneous genetic immunization with autologous melanoma-associated antigen Pmel17/gp100 induces T cell-mediated tumor protection in vivo. J Invest Dermatol 2000; 115:1082-7. [PMID: 11121145 DOI: 10.1046/j.1523-1747.2000.00157.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Using the differentiation antigen Pmel17/gp100 to genetically immunize C57BL/6 mice (H-2(b)), we and colleagues noticed that only mice that had received the human homolog but not animals injected with the murine counterpart were protected against the growth of syngeneic B16 melanoma cells. The goal of this study was to determine whether the state of nonresponsiveness to the autoantigen Pmel17/gp100 can be broken by immunization with a plasmid DNA construct encoding the autologous form of the molecule. A construct containing the murine form of Pmel17 was administered intradermally to DBA/2 mice (H-2(d)), which were then investigated for the presence of Pmel17/gp100-specific immunity. We show that administration of plasmid DNA coding for the autologous melanoma-associated antigen Pmel17/gp100 protects DBA/2 mice against the growth of Pmel17-positive M3 melanoma cells but not against Pmel17-negative M3 melanoma cells or unrelated P815 mastocytoma cells. Cell depletion experiments demonstrated that this protective effect is mediated by T lymphocytes. The notion that Pmel17/gp100 represents the biologically relevant target in this system was supported by the observations (i) that recipients of Pmel17/gp100 DNA mount an antigen-specific cytotoxic T lymphocyte response and (ii) that M3 tumors growing in mice immunized with autologous Pmel17/gp100 had lost expression of this melanoma-associated antigen whereas M3 melanomas appearing in control-vector-treated animals were still Pmel17/gp100-positive. These results indicate that intracutaneous genetic immunization with autologous melanoma-associated antigen Pmel17/gp100 encoding plasmid DNA can lead to protection against melanoma cells as a result of the induction of a melanoma-associated antigen-specific and protective T-cell-mediated immune response. J Invest Dermatol 115:1082-1087 2000
Collapse
Affiliation(s)
- S N Wagner
- Department of Dermatology, University of Essen Medical School, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Slansky JE, Rattis FM, Boyd LF, Fahmy T, Jaffee EM, Schneck JP, Margulies DH, Pardoll DM. Enhanced antigen-specific antitumor immunity with altered peptide ligands that stabilize the MHC-peptide-TCR complex. Immunity 2000; 13:529-38. [PMID: 11070171 DOI: 10.1016/s1074-7613(00)00052-2] [Citation(s) in RCA: 238] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
T cell responsiveness to an epitope is affected both by its affinity for the presenting MHC molecule and the affinity of the MHC-peptide complex for TCR. One limitation of cancer immunotherapy is that natural tumor antigens elicit relatively weak T cell responses, in part because high-affinity T cells are rendered tolerant to these antigens. We report here that amino acid substitutions in a natural MHC class I-restricted tumor antigen that increase the stability of the MHC-peptide-TCR complex are significantly more potent as tumor vaccines. The improved immunity results from enhanced in vivo expansion of T cells specific for the natural tumor epitope. These results indicate peptides that stabilize the MHC-peptide-TCR complex may provide superior antitumor immunity through enhanced stimulation of specific T cells.
Collapse
Affiliation(s)
- J E Slansky
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, USA
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Takeda J, Sato Y, Kiyosawa H, Mori T, Yokoya S, Irisawa A, Miyata M, Obara K, Fujita T, Suzuki T, Kasukawa R, Wanaka A. Anti-tumor immunity against CT26 colon tumor in mice immunized with plasmid DNA encoding beta-galactosidase fused to an envelope protein of endogenous retrovirus. Cell Immunol 2000; 204:11-8. [PMID: 11006013 DOI: 10.1006/cimm.2000.1691] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Endogenous retroviral gene products have been recognized as being expressed in human cancerous tissues. However, these products have not been shown to be antigenic targets for T-cells, possibly due to immune tolerance. Since carcinogen-induced colon tumor CT26 expresses an envelope protein, gp70, of an endogenous ecotropic murine leukemia virus that is comparable to human tumor-associated antigens, we examined whether a DNA vaccine containing the gp70 gene induces protective immunity against CT26 cells. Injection of mice with plasmid DNA (pDNA) encoding gp70 alone failed to induce anti-gp70 antibody (Ab) or anti-CT26 cytotoxic T lymphocyte (CTL) responses. However, immunization with pDNA encoding the beta-galactosidase (beta-gal)/gp70 fusion protein induced anti-gp70 Ab and anti-CT26 CTL responses and conferred protective immunity against CT26 cells. These results indicate that beta-gal acts as an immunogenic carrier protein that helps in the induction of immune responses against the poorly immunogenic gp70. Considering these results, it is possible that potential tolerance to the endogenous retroviral gene products expressed by human tumors may be overcome by DNA vaccines that contain an endogenous retroviral gene fused to genes encoding immunogenic carrier proteins.
Collapse
MESH Headings
- Animals
- Antibodies, Neoplasm/blood
- Cancer Vaccines/therapeutic use
- Colonic Neoplasms/mortality
- Colonic Neoplasms/therapy
- Cytotoxicity, Immunologic
- Female
- Leukemia Virus, Murine/genetics
- Leukemia Virus, Murine/immunology
- Mice
- Mice, Inbred BALB C
- Mice, Inbred DBA
- Neoplasm Proteins/immunology
- Recombinant Fusion Proteins/immunology
- Retroviridae Proteins, Oncogenic/genetics
- Retroviridae Proteins, Oncogenic/immunology
- T-Lymphocytes, Cytotoxic/immunology
- Vaccination
- Vaccines, DNA/therapeutic use
- Viral Envelope Proteins/genetics
- Viral Envelope Proteins/immunology
- beta-Galactosidase/genetics
- beta-Galactosidase/immunology
Collapse
Affiliation(s)
- J Takeda
- Department of Internal Medicine II, Fukushina Medical University School of Medicine, Fukushima 960-1295, Japan.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
van Hall T, van Bergen J, van Veelen PA, Kraakman M, Heukamp LC, Koning F, Melief CJ, Ossendorp F, Offringa R. Identification of a novel tumor-specific CTL epitope presented by RMA, EL-4, and MBL-2 lymphomas reveals their common origin. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2000; 165:869-77. [PMID: 10878361 DOI: 10.4049/jimmunol.165.2.869] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
C57BL/6 mice generate a vigorous H-2Db-restricted CTL response against murine leukemia virus (MuLV)-induced tumors. For many years it has been suggested that this response is directed to an MuLV-encoded peptide as well as to a nonviral tumor-associated peptide. Recently, a peptide from the leader sequence of gag was demonstrated to be the MuLV-derived epitope. Here we describe the molecular identification of the tumor-associated epitope. Furthermore, we show that the CTL response against this epitope can restrict the outgrowth of MuLV-induced tumors in vivo. The epitope is selectively presented by the MuLV-induced T cell tumors RBL-5, RMA, and MBL-2 as well as by the chemically induced T cell lymphoma EL-4. Intriguingly, these tumors share expression of the newly identified epitope because they represent variants of the same clonal tumor cell line, as evident from sequencing of the TCR alpha- and beta-chains, which proved to be identical. Our research shows that all sources of RBL-5, RMA, RMA-S, MBL-2, and EL-4 tumors are derived from a single tumor line, most likely EL-4.
Collapse
MESH Headings
- Adoptive Transfer
- Animals
- Antigen Presentation
- Binding Sites/immunology
- Cancer Vaccines/administration & dosage
- Cell Lineage/genetics
- Cell Lineage/immunology
- Epitopes, T-Lymphocyte/administration & dosage
- Epitopes, T-Lymphocyte/isolation & purification
- Epitopes, T-Lymphocyte/metabolism
- Friend murine leukemia virus
- Gene Expression Regulation/immunology
- Genes, T-Cell Receptor beta
- Leukemia, Experimental/immunology
- Leukemia, Experimental/metabolism
- Leukemia, Experimental/pathology
- Lymphoma, T-Cell/immunology
- Lymphoma, T-Cell/metabolism
- Lymphoma, T-Cell/pathology
- Mice
- Mice, Inbred C57BL
- Mice, Nude
- Moloney murine leukemia virus
- Oligopeptides/administration & dosage
- Oligopeptides/chemical synthesis
- Oligopeptides/immunology
- Oligopeptides/metabolism
- Rauscher Virus
- Sequence Analysis, Protein
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Cytotoxic/metabolism
- T-Lymphocytes, Cytotoxic/pathology
- T-Lymphocytes, Cytotoxic/transplantation
- Thymoma/immunology
- Thymoma/metabolism
- Thymoma/pathology
- Tumor Cells, Cultured/transplantation
- Tumor Virus Infections/immunology
- Tumor Virus Infections/metabolism
- Tumor Virus Infections/pathology
Collapse
Affiliation(s)
- T van Hall
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, The Netherlands
| | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Hsieh CL, Chen DS, Hwang LH. Tumor-induced immunosuppression: a barrier to immunotherapy of large tumors by cytokine-secreting tumor vaccine. Hum Gene Ther 2000; 11:681-92. [PMID: 10757348 DOI: 10.1089/10430340050015581] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
An active immunotherapy strategy with cytokine-assisted tumor vaccine, although often effective for small tumor burdens, is much less so for large tumor burdens. This study examines how large tumors might suppress the T cell functions and escape from the immune responses elicited by a granulocyte-macrophage colony-stimulating factor (GM-CSF)-secreting tumor vaccine. According to our results, the T cells isolated from the tumor-bearing mice treated late with the vaccine failed to confer protective activity on naive mice against a wild-type tumor challenge, unlike those isolated from the early-treated group. Nevertheless, the antitumor activity of the inactive T cells could be restored on in vitro stimulation. Expression of transforming growth factor beta (TGF-beta) and interleukin 10 (IL-10), the potent immunosuppressive factors, was detected in the parental tumor cell line RLmale 1 (a murine T leukemia cell line), as well as in the tumor region, the levels of which correlated with tumor progression. An in vitro assay of T cell functions revealed that the TGF-beta in the conditioned medium of RLmale 1 cells mainly affected the activation, whereas the IL-1male affected the activation to a lesser extent, but significantly affected the cytolytic activity, of tumor-specific T cells. The immunosuppressive activity of IL-10 was also signified by the findings that administration of the conditioned medium of RLmale 1 cultured in a serum-free medium, in which the TGF-beta activity was then lost while the IL-10 activity still remained, or of recombinant IL-10 to the early-treated group of mice abrogated the known efficacy of tumor vaccine on the small tumors. These data suggested that the efficacy of cytokine-secreting tumor vaccine was blocked by the immunosuppressive factors secreted from the large tumors. The results have important implications for the clinical design of immunotherapeutic strategies for advanced cancer patients.
Collapse
Affiliation(s)
- C L Hsieh
- Graduate Institute of Microbiology, College of Medicine, National Taiwan University, Taipei
| | | | | |
Collapse
|
25
|
Castelli C, Rivoltini L, Andreola G, Carrabba M, Renkvist N, Parmiani G. T-cell recognition of melanoma-associated antigens. J Cell Physiol 2000; 182:323-31. [PMID: 10653598 DOI: 10.1002/(sici)1097-4652(200003)182:3<323::aid-jcp2>3.0.co;2-#] [Citation(s) in RCA: 90] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
In this review, we summarize the significant progress that has been made in the identification of melanoma-associated antigens (MAA) recognized by cytotoxic T-lymphocytes (CTL). These antigens belong to three main groups: tumor-associated testis-specific antigens (e.g. , MAGE, BAGE, and GAGE); melanocyte differentiation antigens (e.g., tyrosinase, Melan-A/MART-1); and mutated or aberrantly expressed molecules (e.g, CDK4, MUM-1, beta-catenin). Although strong CTL activity may be induced ex vivo against most of these antigens, often in the presence of excess cytokines and antigen, a clear understanding of the functional status of CTL in vivo and their impact on tumor growth, is still lacking. Several mechanisms are described that potentially contribute to tumor cell evasion of the immune response, suggesting that any antitumor efficacy achieved by immune effectors may be offset by factors that result ultimately in tumor progression. Nevertheless, most of these MAA are currently being investigated as immunizing agents in clinical studies, the conflicting results of which are reviewed. Indeed, the therapeutic potential of MAA has still to be fully exploited and new strategies have to be found in order to achieve an effective and long-lasting in vivo immune control of melanoma growth and progression.
Collapse
Affiliation(s)
- C Castelli
- Unit of Immunotherapy of Human Tumors, Istituto Nazionale Tumori, Milan, Italy
| | | | | | | | | | | |
Collapse
|
26
|
Rosato A, Milan G, Collavo D, Zanovello P. DNA-based vaccination against tumors expressing the P1A antigen. Methods 1999; 19:187-90. [PMID: 10525455 DOI: 10.1006/meth.1999.0844] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Based on experience acquired in the last few years, we describe some technical steps and provide suggestions on how to induce an immune response against tumors expressing the weakly immunogenic antigen P1A by means of a DNA-based vaccination approach. P1A is the product of a normal mouse gene, which shares many characteristics with already identified human tumor-associated antigens, and therefore represents a useful experimental model to evaluate the efficacy of new vaccination strategies potentially applicable to the field of human tumors. Information gained with this model has been applied with success in other experimental settings, and thus we think that the procedure described herein may constitute a valid platform that can be implemented and further refined.
Collapse
Affiliation(s)
- A Rosato
- Department of Oncology and Surgical Sciences, University of Padova, Via Gattamelata 64, Padova, I-35128, Italy.
| | | | | | | |
Collapse
|
27
|
Dellabona P, Moro M, Crosti MC, Casorati G, Corti A. Vascular attack and immunotherapy: a 'two hits' approach to improve biological treatment of cancer. Gene Ther 1999; 6:153-4. [PMID: 10435097 DOI: 10.1038/sj.gt.3300829] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
28
|
Zeh HJ, Perry-Lalley D, Dudley ME, Rosenberg SA, Yang JC. High Avidity CTLs for Two Self-Antigens Demonstrate Superior In Vitro and In Vivo Antitumor Efficacy. THE JOURNAL OF IMMUNOLOGY 1999. [DOI: 10.4049/jimmunol.162.2.989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Abstract
A majority of the human tumor-associated Ags characterized to date are derived from nonmutated “self”-proteins. Little is currently understood about the nature of the self-reactive lymphocytes that recognize these Ags. We recently characterized two nonmutated tumor-associated Ags for the B16 murine melanoma: tyrosinase-related protein-2 (TRP-2) and the endogenous retroviral envelope protein, p15E. We previously reported that both TRP-2 and p15E reactive CTL could be detected in the spleens of naive animals after a single in vitro stimulation using 10−5–10−6 M of the appropriate Kb-binding 9-amino acid epitope. In this report we show that the CTL found in naive animals are low avidity lymphocytes, that respond only to high concentrations of peptide in vitro. We demonstrate that titration of in vitro-stimulating peptide to limiting concentrations distinguishes qualitative differences in the lymphocyte reactivity to these two Ags between vaccinated and unvaccinated animals. We further demonstrate that in vitro expansion of CTL in either high or low concentrations of stimulating peptide generated CTL cultures with different avidities for the relevant epitopes. CTL expanded in low concentrations demonstrated higher avidity for peptide-pulsed targets and better tumor recognition, when compared to CTL generated in the presence of high concentrations of Ag. More importantly, high avidity CTL demonstrated superior in vivo antitumor activity. These results demonstrate that qualitative differences in the CTL that recognize these two self-Ags are critically important to their in vitro and in vivo anti-tumor efficacy.
Collapse
Affiliation(s)
- Herbert J. Zeh
- Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Donna Perry-Lalley
- Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Mark E. Dudley
- Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Steven A. Rosenberg
- Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - James C. Yang
- Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| |
Collapse
|
29
|
Thomas MC, Greten TF, Pardoll DM, Jaffee EM. Enhanced tumor protection by granulocyte-macrophage colony-stimulating factor expression at the site of an allogeneic vaccine. Hum Gene Ther 1998; 9:835-43. [PMID: 9581906 DOI: 10.1089/hum.1998.9.6-835] [Citation(s) in RCA: 72] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Murine tumor models have demonstrated that whole tumor cell vaccines engineered to secrete certain cytokines in a paracrine fashion elicit systemic immune responses capable of eliminating small amounts of established tumor. In particular, autologous tumors that express the cytokine GM-CSF induce potent systemic immune responses against poorly immunogenic murine tumors. However, phase I clinical trials have demonstrated the technical difficulty of routinely expanding primary autologous human tumor cells to the numbers required for vaccination, making the generalization of autologous vaccines impractical. Dissection of the mechanism by which antitumor immunity is generated has demonstrated that GM-CSF recruits professional antigen-presenting cells that act as intermediates in presenting tumor antigen to and activating effector T cells. Furthermore, the identification of commonly recognized murine and human tumor antigens indicates that many are shared rather than unique. These findings would suggest that allogeneic as well as autologous tumor cells can be used as the vaccinating cells for activating antitumor immunity. A major concern in the application of allogeneic vaccines relates to the potential interference of allogeneic MHC expression at the vaccine site with priming of tumor-specific T cell responses. Here we describe a series of experiments that directly examines the effects of allogeneic MHC molecules on the immune-priming capabilities of a whole cell tumor vaccine engineered to secrete GM-CSF. The results demonstrate that the expression of an allogeneic MHC molecule by a vaccine cell can actually enhance the induction of systemic antitumor immunity. In addition, allogeneic MHC expression has no inhibitory effect on the ability of GM-CSF-transduced vaccines to induce systemic antitumor immunity. These findings support the design of clinical trials for testing this more feasible and generalizable allogeneic whole tumor cell vaccine approach.
Collapse
Affiliation(s)
- M C Thomas
- Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | | | | | | |
Collapse
|
30
|
Clynes R, Takechi Y, Moroi Y, Houghton A, Ravetch JV. Fc receptors are required in passive and active immunity to melanoma. Proc Natl Acad Sci U S A 1998; 95:652-6. [PMID: 9435247 PMCID: PMC18475 DOI: 10.1073/pnas.95.2.652] [Citation(s) in RCA: 249] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/1997] [Indexed: 02/05/2023] Open
Abstract
Effective tumor immunity requires recognition of tumor cells coupled with the activation of host effector responses. Fc receptor (FcR) gamma-/- mice, which lack the activating Fc gamma R types I and III, did not demonstrate protective tumor immunity in models of passive and active immunization against a relevant tumor differentiation antigen, the brown locus protein gp75. In wild-type mice, passive immunization with mAb against gp75 or active immunization against gp75 prevented the development of lung metastases. This protective response was completely abolished in FcR gamma-deficient mice. Immune responses were intact in gamma-/- mice because IgG titers against gp75 develop normally in gamma-/- mice immunized with gp75. However, uncoupling of the Fc gamma R effector pathway from antibody recognition of tumor antigens resulted in a loss of protection against tumor challenge. These data demonstrate an unexpected and critical role for FcRs in mediating tumor cytotoxicity in vivo and suggest that enhancement of Fc gamma R-mediated antibody-dependent cellular cytotoxicity by inflammatory cells is a key step in the development of effective tumor immunotherapeutics.
Collapse
Affiliation(s)
- R Clynes
- Laboratory of Molecular Genetics and Immunology, Rockefeller University, New York, NY, USA.
| | | | | | | | | |
Collapse
|
31
|
Rosato A, Zambon A, Milan G, Ciminale V, D'Agostino DM, Macino B, Zanovello P, Collavo D. CTL response and protection against P815 tumor challenge in mice immunized with DNA expressing the tumor-specific antigen P815A. Hum Gene Ther 1997; 8:1451-8. [PMID: 9287145 DOI: 10.1089/hum.1997.8.12-1451] [Citation(s) in RCA: 25] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
A DNA immunization approach was used to induce an immune response against the tumor-specific antigen P815A in DBA/2 mice. The P1A gene, which encodes the P815A antigen, was modified by the addition of a short sequence coding for a tag epitope recognized by the monoclonal antibody AU1, and cloned into the eukaryotic expression vector pBKCMV, resulting in plasmid pBKCMV-P1A. L1210 cells stably transfected with pBKCMV-P1A expressed P1A mRNA and were lysed by the syngeneic P815A-specific cytotoxic clone CTL-P1:5, thus confirming that the tag-modified P1A protein underwent correct processing and presentation. A single intramuscular injection of 100 microg of pBKCMV-P1A induced the expression of P1A mRNA for at least 4 months. Eighty percent of DBA/2 mice injected three times with 100 microg of pBKCMV-P1A generated cytotoxic T lymphocytes (CTL) that lysed P815 tumor cells, whereas mock-inoculated animals failed to show any cytotoxicity. Moreover, experiments designed to evaluate the protection of pBKCMV-P1A-immunized mice against a lethal challenge with P815 tumor cells showed that 6 of 10 immunized mice rejected the tumor, and 2 mice showed prolonged survival compared to control animals.
Collapse
Affiliation(s)
- A Rosato
- Division of Immunology, Department of Oncology and Surgical Sciences, University of Padova, Italy
| | | | | | | | | | | | | | | |
Collapse
|