1
|
Benli-Hoppe T, Göl Öztürk Ş, Öztürk Ö, Berger S, Wagner E, Yazdi M. Transferrin Receptor Targeted Polyplexes Completely Comprised of Sequence-Defined Components. Macromol Rapid Commun 2021; 43:e2100602. [PMID: 34713524 DOI: 10.1002/marc.202100602] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Revised: 10/20/2021] [Indexed: 11/08/2022]
Abstract
Human transferrin protein (Tf) modified polyplexes have already displayed encouraging potential for receptor-mediated nucleic acid delivery into tumors. The use of a blood-derived targeting protein and polydisperse macromolecular cationic subunits however presents a practical challenge for pharmaceutical grade production. Here, Tf receptor (TfR) targeted small interfering RNA (siRNA) polyplexes are designed that are completely composed of synthetic, monodisperse, and sequence-defined subunits generated by solid-phase supported synthesis. An optimized cationizable lipo-oligoaminoamide (lipo-OAA) is used for siRNA core polyplex formation, and a retro-enantio peptide (reTfR) attached via a monodisperse polyethylene glycol (PEG) spacer via click chemistry is applied for targeting. Improved gene silencing is demonstrated in TfR-expressing KB and DU145 cells. Analogous plasmid DNA (pDNA) polyplexes are successfully used for receptor-mediated gene delivery in TfR-rich K562 cells and Neuro2a cells. Six lipo-OAAs differing in their lipidic domain and redox-sensitive attachment of lipid residues are tested in order to evaluate the impact of core polyplex stability on receptor-dependent gene transfer.
Collapse
Affiliation(s)
- Teoman Benli-Hoppe
- Pharmaceutical Biotechnology, Center for Drug Research, and Center for Nanoscience, Ludwig-Maximilians-Universität Munich, Butenandtstrasse 5-13, Munich, 81377, Germany
| | - Şurhan Göl Öztürk
- Pharmaceutical Biotechnology, Center for Drug Research, and Center for Nanoscience, Ludwig-Maximilians-Universität Munich, Butenandtstrasse 5-13, Munich, 81377, Germany
| | - Özgür Öztürk
- Pharmaceutical Biotechnology, Center for Drug Research, and Center for Nanoscience, Ludwig-Maximilians-Universität Munich, Butenandtstrasse 5-13, Munich, 81377, Germany
| | - Simone Berger
- Pharmaceutical Biotechnology, Center for Drug Research, and Center for Nanoscience, Ludwig-Maximilians-Universität Munich, Butenandtstrasse 5-13, Munich, 81377, Germany
| | - Ernst Wagner
- Pharmaceutical Biotechnology, Center for Drug Research, and Center for Nanoscience, Ludwig-Maximilians-Universität Munich, Butenandtstrasse 5-13, Munich, 81377, Germany
| | - Mina Yazdi
- Pharmaceutical Biotechnology, Center for Drug Research, and Center for Nanoscience, Ludwig-Maximilians-Universität Munich, Butenandtstrasse 5-13, Munich, 81377, Germany
| |
Collapse
|
2
|
Büning H, Fehse B, Ivics Z, Kochanek S, Koehl U, Kupatt C, Mussolino C, Nettelbeck DM, Schambach A, Uckert W, Wagner E, Cathomen T. Gene Therapy "Made in Germany": A Historical Perspective, Analysis of the Status Quo, and Recommendations for Action by the German Society for Gene Therapy. Hum Gene Ther 2021; 32:987-996. [PMID: 34662229 DOI: 10.1089/hum.2021.29178.hbu] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Gene therapies have been successfully applied to treat severe inherited and acquired disorders. Although research and development are sufficiently well funded in Germany and while the output of scientific publications and patents is comparable with the leading nations in gene therapy, the country lags noticeably behind with regard to the number of both clinical studies and commercialized gene therapy products. In this article, we give a historical perspective on the development of gene therapy in Germany, analyze the current situation from the standpoint of the German Society for Gene Therapy (DG-GT), and define recommendations for action that would enable our country to generate biomedical and economic advantages from innovations in this sector, instead of merely importing advanced therapy medicinal products. Inter alia, we propose (1) to harmonize and simplify regulatory licensing processes to enable faster access to advanced therapies, and (2) to establish novel coordination, support and funding structures that facilitate networking of the key players. Such a center would provide the necessary infrastructure and know-how to translate cell and gene therapies to patients on the one hand, and pave the way for commercialization of these promising and innovative technologies on the other. Hence, these courses of action would not only benefit the German biotech and pharma landscape but also the society and the patients in need of new treatment options.
Collapse
Affiliation(s)
- Hildegard Büning
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
| | - Boris Fehse
- Research Department Cell and Gene Therapy, Department of Stem Cell Transplantation, University Medical Centre Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Zoltán Ivics
- Division of Medical Biotechnology, Paul Ehrlich Institute, Langen, Germany
| | | | - Ulrike Koehl
- Fraunhofer Institute for Cell Therapy and Immunology (IZI) and Institute of Clinical Immunology, University of Leipzig, Leipzig, Germany.,Institute for Cellular Therapeutics, Hannover Medical School, Hannover, Germany
| | - Christian Kupatt
- Klinik und Poliklinik für Innere Medizin I, Klinikum rechts der Isar, Technical University Munich, Munich, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Claudio Mussolino
- Institute for Transfusion Medicine and Gene Therapy, Medical Center-University of Freiburg, Freiburg, Germany.,Center for Chronic Immunodeficiency (CCI), Medical Faculty, University of Freiburg, Freiburg, Germany
| | - Dirk M Nettelbeck
- Clinical Cooperation Unit Virotherapy, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Axel Schambach
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
| | - Wolfgang Uckert
- Department of Molecular Cell Biology and Gene Therapy, Max-Delbrück-Center for Molecular Medicine (MDC), Berlin, Germany
| | - Ernst Wagner
- Pharmaceutical Biotechnology, Center for System-based Drug Research, Center for NanoScience (CeNS), Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Toni Cathomen
- Institute for Transfusion Medicine and Gene Therapy, Medical Center-University of Freiburg, Freiburg, Germany.,Center for Chronic Immunodeficiency (CCI), Medical Faculty, University of Freiburg, Freiburg, Germany
| |
Collapse
|
3
|
Abstract
It has been known for decades that the immune system can be spontaneously activated against melanoma. The presence of tumor infiltrating lymphocytes in tumor deposits is a positive prognostic factor. Cancer vaccination includes approaches to generate, amplify, or skew antitumor immunity. To accomplish this goal, tested approaches involve administration of tumor antigens, antigen presenting cells or other immune modulators, or direct modulation of the tumor. Because the success of checkpoint blockade can depend in part on an existing antitumor response, cancer vaccination may play an important role in future combination therapies. In this review, we discuss a variety of melanoma vaccine approaches and methods to determine the biological impact of vaccination.
Collapse
|
4
|
Zhang P, Wagner E. History of Polymeric Gene Delivery Systems. Top Curr Chem (Cham) 2017; 375:26. [PMID: 28181193 DOI: 10.1007/s41061-017-0112-0] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2016] [Accepted: 01/24/2017] [Indexed: 12/16/2022]
Abstract
As an option for genetic disease treatment and an alternative for traditional cancer chemotherapy, gene therapy achieves significant attention. Nucleic acid delivery, however, remains a main challenge in human gene therapy. Polymer-based delivery systems offer a safer and promising route for therapeutic gene delivery. Over the past five decades, various cationic polymers have been optimized for increasingly effective nucleic acid transfer. This resulted in a chemical evolution of cationic polymers from the first-generation polycations towards bioinspired multifunctional sequence-defined polymers and nanocomposites. With the increasing of knowledge in molecular biological processes and rapid progress of macromolecular chemistry, further improvement of polymeric nucleic acid delivery systems will provide effective tool for gene-based therapy in the near future.
Collapse
Affiliation(s)
- Peng Zhang
- Pharmaceutical Biotechnology, Center for System-Based Drug Research Ludwig-Maximilians-Universität, 81377, Munich, Germany. .,Nanosystems Initiative Munich (NIM), 80799, Munich, Germany.
| | - Ernst Wagner
- Pharmaceutical Biotechnology, Center for System-Based Drug Research Ludwig-Maximilians-Universität, 81377, Munich, Germany.,Nanosystems Initiative Munich (NIM), 80799, Munich, Germany.,Center for NanoScience (CeNS), Ludwig-Maximilians-Universität, 80799, Munich, Germany
| |
Collapse
|
5
|
Lächelt U, Wagner E. Nucleic Acid Therapeutics Using Polyplexes: A Journey of 50 Years (and Beyond). Chem Rev 2015; 115:11043-78. [DOI: 10.1021/cr5006793] [Citation(s) in RCA: 469] [Impact Index Per Article: 46.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Ulrich Lächelt
- Pharmaceutical
Biotechnology, Department of Pharmacy, Ludwig Maximilians Universität, 81377 Munich, Germany
- Nanosystems
Initiative
Munich (NIM), 80799 Munich, Germany
| | - Ernst Wagner
- Pharmaceutical
Biotechnology, Department of Pharmacy, Ludwig Maximilians Universität, 81377 Munich, Germany
- Nanosystems
Initiative
Munich (NIM), 80799 Munich, Germany
| |
Collapse
|
6
|
Agorio C, Schreiber F, Sheppard M, Mastroeni P, Fernandez M, Martinez MA, Chabalgoity JA. Live attenuatedSalmonella as a vector for oral cytokine gene therapy in melanoma. J Gene Med 2007; 9:416-23. [PMID: 17410612 DOI: 10.1002/jgm.1023] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Systemic administration of cytokines has shown therapeutic benefits in cancer patients; however, serious adverse effects associated with direct protein administration prevent the wide use of this approach. We have assessed the capacity of live attenuated Salmonella to act as a vector for oral cytokine-gene therapy. Salmonella orally administered to melanoma-bearing mice was found to accumulate within the tumor, reaching up to 10(5) bacteria per gram of tumor by day 21 after bacterial inoculation. Numbers of bacteria recovered from tumor did not differ from those recovered from liver or spleen at any time point. Recombinant bacteria carrying eukaryotic expression vectors encoding the murine IL-4 or IL-18 genes were administered to groups of mice with established subcutaneous melanoma tumors. We found that a single oral dose of Salmonella carrying any of the cytokine-encoding plasmids resulted in significantly increased survival time, as compared with mice that received Salmonella carrying the parental plasmid or PBS. Increased levels of IFNgamma were found in sera of animals receiving either of the cytokine-encoding bacteria, but not in mice receiving Salmonella alone or PBS. Co-administration of both recombinant bacteria maximized the production of IFNgamma. Overall these results suggest that cytokine-encoding Salmonella can be an effective and safer alternative to systemic administration of cytokines for immunotherapy of cancer.
Collapse
Affiliation(s)
- Caroline Agorio
- Laboratory for Vaccine Research, Departamento de Desarrollo Biotecnológico, Instituto de Higiene, Facultad de Medicina, Av. Navarro 3051, Montevideo, Uruguay
| | | | | | | | | | | | | |
Collapse
|
7
|
Couvreur P, Vauthier C. Nanotechnology: intelligent design to treat complex disease. Pharm Res 2006; 23:1417-50. [PMID: 16779701 DOI: 10.1007/s11095-006-0284-8] [Citation(s) in RCA: 523] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2005] [Accepted: 03/01/2006] [Indexed: 01/19/2023]
Abstract
The purpose of this expert review is to discuss the impact of nanotechnology in the treatment of the major health threats including cancer, infections, metabolic diseases, autoimmune diseases, and inflammations. Indeed, during the past 30 years, the explosive growth of nanotechnology has burst into challenging innovations in pharmacology, the main input being the ability to perform temporal and spatial site-specific delivery. This has led to some marketed compounds through the last decade. Although the introduction of nanotechnology obviously permitted to step over numerous milestones toward the development of the "magic bullet" proposed a century ago by the immunologist Paul Ehrlich, there are, however, unresolved delivery problems to be still addressed. These scientific and technological locks are discussed along this review together with an analysis of the current situation concerning the industrial development.
Collapse
Affiliation(s)
- Patrick Couvreur
- Laboratoire de Physico-chimie, Pharmacotechnie et Biopharmacie, UMR CNRS 8612, Université de Paris Sud, 5 Rue J.B. Clément, 92 296, Chatenay-Malabry Cedex, France
| | | |
Collapse
|
8
|
Yamano T, Kaneda Y, Huang S, Hiramatsu SH, Hoon DSB. Enhancement of immunity by a DNA melanoma vaccine against TRP2 with CCL21 as an adjuvant. Mol Ther 2005; 13:194-202. [PMID: 16112911 DOI: 10.1016/j.ymthe.2005.05.018] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2005] [Revised: 05/04/2005] [Accepted: 05/19/2005] [Indexed: 10/25/2022] Open
Abstract
Tyrosinase-related protein-2 (TRP2) is a weak antigen expressed in murine and human melanomas. Induction of antibody (Ab) response and T-cell immunity toward TRP2 with DNA plasmid vaccines has not been efficient to date. Recent studies have suggested that a chemokine ligand for the CCR7 (CCL21) present on T-cells and dendritic cells is important in activating and regulating immunity. We investigated the effectiveness of CCL21 as an adjuvant with an HVJ anionic liposomal TRP2 DNA (plasmid) vaccine to enhance anti-TRP2 Ab, cytokines, delayed-type hypersensitivity, T-cell responses, and tumor protection against B16 melanoma cells. Induction of anti-TRP2 immunity depended mainly on cell-mediated immunity, which was regulated by timing and route of CCL21 administration with DNA vaccine. The optimum protocol was to administer CCL21 im 24 h before DNA vaccine at the same vaccination site. Two vaccinations (prime/boost) were essential for induction of strong anti-TRP2 cell-mediated immunity. CCL21 administration 3 days before or 24 h after DNA vaccine, simultaneous with DNA vaccine, or at different sites (iv, opposite leg) was not effective. This study demonstrated that CCL21 was an effective adjuvant to enhance TRP2-specific immunity induced by a plasmid DNA cancer vaccine.
Collapse
MESH Headings
- Adjuvants, Immunologic/administration & dosage
- Adjuvants, Immunologic/therapeutic use
- Animals
- Antibody Formation
- Cancer Vaccines/administration & dosage
- Cancer Vaccines/immunology
- Cancer Vaccines/therapeutic use
- Cell Line, Tumor
- Chemokine CCL21
- Chemokines, CC/administration & dosage
- Chemokines, CC/immunology
- Chemokines, CC/therapeutic use
- Female
- Immunity, Cellular
- Intramolecular Oxidoreductases/immunology
- Liposomes
- Lymph Nodes/immunology
- Lymph Nodes/pathology
- Melanoma, Experimental/immunology
- Melanoma, Experimental/therapy
- Mice
- Mice, Inbred C57BL
- Neoplasm Transplantation
- Spleen/immunology
- Spleen/pathology
- Transplantation, Heterologous
- Vaccination
- Vaccines, DNA/administration & dosage
- Vaccines, DNA/immunology
- Vaccines, DNA/therapeutic use
Collapse
Affiliation(s)
- Tomoki Yamano
- Department of Molecular Oncology, John Wayne Cancer Institute, Saint John's Health Center, 2200 Santa Monica Boulevard, Santa Monica, CA 90404, USA
| | | | | | | | | |
Collapse
|
9
|
Larin SS, Korobko EV, Kustikova OS, Borodulina OR, Raikhlin NT, Brisgalov IP, Georgiev GP, Kiselev SL. Immunotherapy with autologous tumor cells engineered to secrete Tag7/PGRP, an innate immunity recognition molecule. J Gene Med 2004; 6:798-808. [PMID: 15241787 DOI: 10.1002/jgm.560] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Recent studies indicate that the innate component of immune defense plays an important role in the establishment of antigen-specific immune response. We have previously isolated a novel mouse gene tag7/PGRP that was shown to be involved in the innate component of the immune system, and its insect homologue is an upstream mediator of Toll signaling in Drosophila. METHODS Transiently or stably genetically modified mouse tumor cell lines expressing Tag7 were used. Tumor growth rate and animal survival were analyzed. Possible effector cells involved in tumor suppression were detected immunohistochemically. RESULTS Transfection of mammary gland adenocarcinoma cells with the tag7 cDNA did not alter their growth rate in vitro but diminished their tumorogenicity in vivo in syngeneic and immunodeficient animals. Increased incidence of apoptosis was registered in the modified tumors. Transient expression of Tag7 by mouse melanoma M3 cells elicited protective immunity against parental tumor cells. Immunohistochemical analysis revealed that tumors after immunization with the genetically modified cells were infiltrated with Mac1(+) cells, B220(+) cells, and NK cells. Using nude mice we observed rejection of modified cells, but did not detect memory formation. CONCLUSIONS We can conclude that secretion of the Tag7 protein by genetically modified cells can induce mobilization of antigen-presenting cells and innate effectors. Memory mechanisms are mediated by T cell response. For the first time our results demonstrate that local secretion of Tag7-the molecule involved in innate immunity-may play an important role in the induction of effective antitumor response in mice.
Collapse
Affiliation(s)
- Sergey S Larin
- Institute of Gene Biology, Russian Academy of Sciences, Moscow, Russia
| | | | | | | | | | | | | | | |
Collapse
|
10
|
Staib L, Harel W, Mitchell MS. Optimization of intracerebral tumour protection by active-specific immunization against murine melanoma B16/G3.12. Melanoma Res 2001; 11:325-35. [PMID: 11479420 DOI: 10.1097/00008390-200108000-00002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Development of brain metastases despite extracerebral response to systemic immunotherapy is a common problem in melanoma patients. We have previously described a murine melanoma vaccine of interferon-gamma (IFNgamma)-treated, irradiated syngeneic B16/G3.12 and allogeneic (Cloudman) melanoma cells, plus the adjuvant DETOX, that is protective against subcutaneous (93%) or intracerebral (69%) syngeneic challenge. This study aimed to optimize this vaccine. Groups of nine or 10 mice were immunized five times in 5 weeks with: (i) complete vaccine +/- IFNgamma (VAC+, VAC-); (ii) syngeneic 2 x 106 G3.12 cells plus DETOX (Syn+D), (iii) 2 x 106 allogeneic Cloudman cells plus DETOX (Allo+D); (iv) VAC+ without DETOX (no DETOX); (v) DETOX alone (DETOX); or (vi) phosphate buffered saline (PBS). Mice were challenged subcutaneously with 104 viable G3.12 (or Cloudman cells) and after 35 days intracerebrally with 104 G3.12 cells. Expression of H-2 antigens (measured using fluorescence-activated cell sorting), splenocyte cytotoxicity (measured using 51Cr release) and median overall survival (OAS) were analysed using the log-rank test. VAC+, VAC- and G3.12 mice were equally protected from subcutaneous (s.c.) and intracerebral (i.c.) melanoma challenge (OAS 65 days for s.c., 30 days for i.c.). Protection was less (P < 0.05) in DETOX mice (48 days for s.c.), PBS mice (47 days for s.c., 21 days for i.c.) or no DETOX mice (51 days for s.c.). Allo+D mice showed s.c. (59 days) but not i.c. protection (20 days). IFNgamma incubation did not increase the effect in either the challenge cells or the vaccine cells (P > 0.05). Specific cytotoxicity was seen with G3.12 targets in VAC+ (27%) but not PBS (2%; P < 0.05) mice with equal NK (YAC-1) lysis (10% versus 7%; P< 0.05). Optimal protection against s.c./i.c. experimental murine melanoma was yielded by irradiated syngeneic cells plus DETOX. DETOX alone was not active. Upregulation of H-2 antigens with IFNgamma under these conditions does not augment protection.
Collapse
Affiliation(s)
- L Staib
- Department of Medicine and the Kenneth Norris Jr Comprehensive Cancer Center, University of Southern California School of Medicine, Los Angeles, California 90033, USA.
| | | | | |
Collapse
|
11
|
Abstract
Our knowledge of the immune system has grown tremendously in the 50 years since Coley used bacteria in an attempt to create a vaccine for cancer. The strategy for cancer vaccines has developed in that time as well. Both clinical and laboratory evidence suggests that melanoma is the more immunogenic of solid tumors. If treated early, melanoma can be controlled with surgery, but many patients continue to die from it. With our increased understanding of the immune system's interaction with melanoma, many clinical trials of melanoma vaccines are now underway. Vaccines designed to treat metastatic melanoma have shown some evidence of clinical effectiveness. This article outlines the current status of melanoma vaccination.
Collapse
Affiliation(s)
- L W Thompson
- Department of Surgery, University of Virginia Health Science Center, PO Box 10005, Charlottesville, VA 22906-0005, USA
| | | | | |
Collapse
|
12
|
Abstract
Remarkable advances in tumor vaccination have been made since Coley first deliberately infected cancer patients with both live and heat-killed bacteria. Melanoma is the most immunogenic solid tumor and, as such, has served as the major model for tumor vaccine investigation in both the laboratory and the clinic. Many advances in the field of melanoma vaccination have been based on an improved understanding of the cellular interaction required to induce a specific antitumor immune response. As a result of this new knowledge, many clinical trials of melanoma vaccines are now under way, and vaccines for metastatic melanoma have shown evidence of clinical effectiveness. This paper outlines the current status of melanoma vaccination.
Collapse
Affiliation(s)
- L H Brinckerhoff
- University of Virginia Health System, Department of Surgery, Charlottesville 22908, USA
| | | | | |
Collapse
|
13
|
Abstract
Gene transfer to humans requires carriers for the plasmid DNA which can efficiently and safely carry the gene into the nucleus of the desired cells. A series of chemically different cationic polymers are currently being investigated for these purposes. Although many cationic polymers indeed condense DNA spontaneously, which is a requirement for gene transfer in most types of cells, the physicochemical and biopharmaceutical behavior of the current generation of polyplexes severely limits an efficient gene transfer in vitro and especially in vivo. This paper summarizes recent physicochemical and biological information on polyplexes and aims to provide new insights with respect to this type of gene delivery system. Firstly, the chemical structure of frequently studied cationic polymers is represented. Secondly, the parameters influencing condensation of DNA by cationic polymers are described. Thirdly, the surface properties, solubility, aggregration behavior, degradation and dissociation of polyplexes are considered. The review ends by describing the in vitro and in vivo gene transfection behavior of polyplexes.
Collapse
|
14
|
Abstract
Early studies demonstrated the role of cytotoxic T cells as an immune defence mechanism against tumour cells. The demonstration of tumour antigen peptides and their presentation to T cells on major histocompatibility complex class I molecules highlighted the importance of these molecules in effective anti-tumour responses. It is well established that many tumours escape T cell recognition by loss or down regulation of class I molecule expression on the cell surface of tumour cells. Tumours which have lost class I expression are immunoselected and as a result have a propensity for growth and metastatic spread. With the development of cancer vaccine strategies for clinical use, there will be a future role for histocompatibility laboratories in determining class I expression on tumour cells in individual patients. These studies of expression will require not just the demonstration of total class I expression but the demonstration of locus and allele specific class I molecules involved in the relevant tumour peptide presentation. These studies will be pivotal in tailoring individual patient therapies. The identification of appropriate monoclonal antibody reagents for class I expression and techniques used on different kinds of tissue sections will be a component of the forthcoming 13th International Histocompatibility Workshop.
Collapse
Affiliation(s)
- B D Tait
- Victorian Transplantation and Immunogenetics Service-Australian Red Cross Blood Services, Parkville, Victoria, Australia.
| |
Collapse
|
15
|
Sprinzl GM, Hussl B, Obrist P, Yoneda K, Thumfart WF, Romani N, Schrott-Fischer A. Dendritic cells in precancerous lesions of the larynx. Laryngoscope 2000; 110:13-8. [PMID: 10646708 DOI: 10.1097/00005537-200001000-00003] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVES Hyperplastic lesions of the laryngeal mucosa can eventually develop into squamous cell carcinoma The relationship between dendritic cell infiltration of head and neck cancers and prognosis is well known. Surprisingly, data regarding dendritic cell infiltration in precancerous lesions are not available today. It was the purpose of our study to extend these observations and to investigate in more detail the density and distribution of dendritic cells in pre-cancerous lesions. STUDY DESIGN Retrospective survey by immunohistochemistry. METHODS For this study we investigated paraffin-embedded tissue sections of 41 specimens. Histological diagnosis disclosed precancerous lesions of the larynx in 34 cases and in 7 cases, squamous cell carcinoma Immunohistochemical study was performed using antibodies against the cell surface markers S-100, HLA-DR, CD20, CD45 RO, CD45 RA, and Lag. Typical dendritic cell distributions of the immunostained specimens were photographed and measured on a quantitative basis. The medical histories of the patients were then analyzed retrospectively. RESULTS HLA-DR+ cells could be detected in 14 of 16 cases in mild dysplastic lesions. The infiltration of the dysplastic lesions was sparse compared to cases with higher-graded dysplastic lesions. The distribution patterns of the dendritic cells in specimens with severe dysplastic lesions, but squamous cell carcinoma were extremely similar and markedly different from those in grades I and II specimens. Memory T lymphocytes (CD45 RO+) were detected more often in the group with severe dysplastic lesions (8 of 9 cases) than in the group with squamous cell carcinoma (3 of 8 cases). The inverse became evident for CD20 and CD45 RA immunolabeling. CONCLUSIONS Few dendritic cells were found in the precancerous lesions. This may suggest that these early lesions (grades I and H) are not efficiently monitored by the immune system. Therefore they may develop into carcinomas unimpaired by cytotoxic T cells. As the degree of malignancy rises (grade III), more dendritic cells infiltrate the tumor.
Collapse
Affiliation(s)
- G M Sprinzl
- Department of Otorhinolaryngology--Head and Neck Surgery, University ofInnsbruck, Austria
| | | | | | | | | | | | | |
Collapse
|
16
|
Schreiber S, Kämpgen E, Wagner E, Pirkhammer D, Trcka J, Korschan H, Lindemann A, Dorffner R, Kittler H, Kasteliz F, Küpcü Z, Sinski A, Zatloukal K, Buschle M, Schmidt W, Birnstiel M, Kempe RE, Voigt T, Weber HA, Pehamberger H, Mertelsmann R, Bröcker EB, Wolff K, Stingl G. Immunotherapy of metastatic malignant melanoma by a vaccine consisting of autologous interleukin 2-transfected cancer cells: outcome of a phase I study. Hum Gene Ther 1999; 10:983-93. [PMID: 10223732 DOI: 10.1089/10430349950018382] [Citation(s) in RCA: 94] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
We performed a phase I trial to evaluate the safety and tolerability of repeated skin injections of IL-2-transfected autologous melanoma cells into patients with advanced disease. Cell suspensions, propagated from excised metastases, were IL-2 gene transfected by adenovirus-enhanced transferrinfection and X-irradiated prior to injection. Vaccine production was successful in 54% of the patients. Fifteen patients (37%) received two to eight skin vaccinations of either 3 x 10(6) (intradermal) or 1 x 10(7) (half intradermal, half subcutaneous) transfected melanoma cells per vaccination (secreting 140-17,060 biological response modifier program units of IL-2/10(6) cells/24 hr). Analyses of safety and efficacy were carried out in 15 and 14 patients, respectively. Overall, the vaccine was well tolerated. All patients displayed modest local reactions (erythema, induration, and pruritus) and some experienced flu-like symptoms. Apart from newly appearing (4 of 14) and increasing (5 of 14) anti-adenovirus and newly detectable anti-nuclear antibody titers (1 of 15), recipients developed de novo or exhibited increased melanoma cell-specific delayed-type hypersensitivity (DTH) reactions (8 of 15) and vitiligo (3 of 15) and showed signs of tumor regression (3 of 15). This supports the idea of a vaccine-induced or -amplified anti-cancer immune response. None of the patients exhibited complete or partial regressions, but five of them experienced periods of disease stabilization. Three of these individuals received more than the four planned vaccinations and their mean survival time was 15.7 +/- 3.5 months as compared to 7.8 +/- 4.6 months for the entire patient cohort. These data indicate that IL-2-producing, autologous cancer cells can be safely administered to stage IV melanoma patients and could conceivably be of benefit to patients with less advanced disease.
Collapse
Affiliation(s)
- S Schreiber
- Department of Dermatology, University of Vienna Medical School, Austria
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Abstract
It has been more than 100 years since the first reported attempts to activate a patient's immune system to eradicate developing cancers. Although a few of the subsequent vaccine studies demonstrated clinically significant treatment effects, active immunotherapy has not yet become an established cancer treatment modality. Two recent advances have allowed the design of more specific cancer vaccine approaches: improved molecular biology techniques and a greater understanding of the mechanisms involved in the activation of T cells. These advances have resulted in improved systemic antitumor immune responses in animal models. Because most tumor antigens recognized by T cells are still not known, the tumor cell itself is the best source of immunizing antigens. For this reason, most vaccine approaches currently being tested in the clinics use whole cancer cells that have been genetically modified to express genes that are now known to be critical mediators of immune system activation. In the future, the molecular definition of tumor-specific antigens that are recognized by activated T cells will allow the development of targeted antigen-specific vaccines for the treatment of patients with cancer.
Collapse
Affiliation(s)
- T F Greten
- Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, MD 21205-2196, USA
| | | |
Collapse
|
18
|
Hengge UR, Pfützner W, Williams M, Goos M, Vogel JC. Efficient expression of naked plasmid DNA in mucosal epithelium: prospective for the treatment of skin lesions. J Invest Dermatol 1998; 111:605-8. [PMID: 9764840 DOI: 10.1046/j.1523-1747.1998.00353.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Mucocutaneous gene therapy offers exciting new treatment modalities for skin lesions. Transient expression of naked plasmid DNA could be used as a local treatment of various skin lesions where the corresponding gene product (protein) has therapeutic or immunization potential. We analyzed the time course, magnitude, and histologic expression of the indicator plasmid DNA (pCMV:beta-Gal) in mucosal epithelium and papilloma lesions. Upon direct injection of naked plasmid DNA (20 microg) into oral mucosa, expression occurred at high local concentrations, up to 35-fold higher than in comparable injections into the epidermis. Due to the accelerated turnover of mucosal epithelium beta-galactosidase positive epithelial cells were detected in the basal and suprabasal layers as early as 3 h after injection, whereas only the most superficial mucosal layers demonstrated beta-galactosidase staining at 24 h post-injection. These biologic characteristics need to be taken into consideration when clinical applications of expressing naked plasmid DNA in epithelial tissues are considered.
Collapse
Affiliation(s)
- U R Hengge
- Department of Dermatology and Venerology, University of Essen, Germany
| | | | | | | | | |
Collapse
|
19
|
Ollila DW, Kelley MC, Gammon G, Morton DL. Overview of melanoma vaccines: active specific immunotherapy for melanoma patients. SEMINARS IN SURGICAL ONCOLOGY 1998; 14:328-36. [PMID: 9588726 DOI: 10.1002/(sici)1098-2388(199806)14:4<328::aid-ssu9>3.0.co;2-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Although a phase III trial has yet to show a statistically significant improvement in the disease-free or overall survival of melanoma patients receiving vaccine therapy, several phase II trials have shown enhanced disease-free and overall survival of patients who develop a humoral and/or cellular response to a melanoma vaccine. The challenge of active specific immunotherapy research is to determine which combination of humoral and cellular immune responses optimizes clinical outcome and how to monitor the immune response effectively. This review identifies key components of a successful melanoma vaccine, discusses new ways to modulate and stimulate the immune system, and summarizes some of the important clinical trials of active specific immunotherapy for patients with melanoma.
Collapse
Affiliation(s)
- D W Ollila
- The Roy E. Coats Research Laboratories, John Wayne Cancer Institute at Saint John's Health Center, Santa Monica, California 90404, USA
| | | | | | | |
Collapse
|
20
|
Abstract
As melanoma evolves, it interacts with the immune system. Based on this immunobiology, there are now a number of rationally designed attempts to develop genetically modified melanoma vaccines. This article outlines immunologic and other strategies in gene therapy for melanoma and provides an overview of current clinical trials.
Collapse
Affiliation(s)
- R Gutzmer
- Division of Hematology/Oncology, University of Pennsylvania School of Medicine, Philadelphia, USA
| | | |
Collapse
|
21
|
Abstract
Receptor-mediated gene transfer is a promising gene delivery technique. It employs a DNA-binding polycation, such as polylysine, to compact plasmid DNA to a size that can be taken up by cells (<100-200 nm). To allow internalization by receptor-mediated endocytosis, cell binding ligands, such as asialoglycoproteins or galactose for hepatocytes, anti-CD3 and anti-CD5 for T-cells, and transferrin, have been covalently attached to polylysine. Intracellular barriers for successful gene transfer include release of DNA complexes from endosomes or lysosomes, nuclear import of DNA complexes, and disassembly of the DNA-polylysine particles. Release of particles from internal vesicles has been achieved by the addition of lysosomotropic agents or glycerol to the transfection medium, or by the incorporation of endosomolytic compounds, such as viruses or membrane active peptides. This technique has already been used to transfect certain organs in vivo, including liver and lung.
Collapse
|
22
|
Bennett MJ, Aberle AM, Balasubramaniam RP, Malone JG, Malone RW, Nantz MH. Cationic lipid-mediated gene delivery to murine lung: correlation of lipid hydration with in vivo transfection activity. J Med Chem 1997; 40:4069-78. [PMID: 9406597 DOI: 10.1021/jm970155q] [Citation(s) in RCA: 86] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
A panel of lipidic tetraalkylammonium chlorides has been prepared and screened in studies of both lipid hydration and in vivo mouse transfection. The effect of cationic lipid structure on liposome surface hydration was determined using differential scanning calorimetry. Increases in headgroup steric bulk and the inclusion of cis-unsaturation in the hydrophobic domain led to greater lipid hydration, indicative of a decrease in lipid polar domain associations. Cationic lipids containing hydrogen-bonding functionality in the polar domain exhibited a corresponding decrease in observed lipid hydration, indicative of an increase in lipid polar domain associations. To explore a potential correlation of the hydration data with transfection activity, we examined the in vivo transfection activity of the lipid panel by direct intratracheal instillation of cationic liposome-DNA complexes into BALB/c mice. The more active transfection agents were the lipids that featured headgroup structures promoting close polar domain association in combination with fatty acyl cis-unsaturation. The hydration data suggest that the more effective transfection lipids for mouse lung delivery are those possessing the greatest imbalance between the cross-sectional areas occupied by the polar and hydrophobic domains.
Collapse
Affiliation(s)
- M J Bennett
- Department of Chemistry, University of California, Davis 95616, USA
| | | | | | | | | | | |
Collapse
|
23
|
Abstract
Although a randomized clinical trial has yet to show a statistically significant improvement in the survival of patients receiving vaccine therapy for malignant melanoma, several studies have shown enhanced survival of patients developing an immune response to a melanoma vaccine. The knowledge and techniques of modern molecular biology and immunology suggest multiple strategies to augment this response. The challenge of immunotherapy research is to determine which combination of approaches leads to a favorable clinical response and how to monitor that response effectively. This review identifies components of a successful vaccine, discusses new ways to modulate and stimulate the immune system, and summarizes some of the more interesting clinical trials of melanoma vaccine immunotherapy.
Collapse
Affiliation(s)
- A M Conforti
- Roy E. Coats Research Laboratories, John Wayne Cancer Institute, Saint John's Health Center, Santa Monica, CA 90404, USA
| | | | | | | | | |
Collapse
|
24
|
Dwenger A, Lindemann A, Mertelsmann R. Minimal residual disease: detection, clinical relevance, and treatment strategies. JOURNAL OF HEMATOTHERAPY 1996; 5:537-48. [PMID: 8938526 DOI: 10.1089/scd.1.1996.5.537] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Improvement of established treatment strategies for cancer has resulted in increased survival times for patients with malignancies. However, success of surgery, chemotherapy, and radiotherapy is limited, as even combined and repeated therapy regimens and high-dose chemotherapy can only reduce tumor burden by several logarithmic steps and are not able to completely eradicate all neoplastic cells. If clinically complete remission is achieved--that is, if no sign of the tumor is detectable by standard diagnostic procedures, remaining minimal residual disease (MRD) can eventually give rise to clinically manifest relapse. More sensitive methods are, therefore, necessary to detect single tumor cells for exact staging, to assess the metastatic potential of an individual tumor, to evaluate the sensitivity to prior therapy, and to detect MRD-positive patients with remaining malignant cells who are at higher risk for relapse. Novel treatment approaches must be created to eradicate minimal residual disease after conventional therapy.
Collapse
Affiliation(s)
- A Dwenger
- University Medical Center Freiburg, Department of Internal Medicine I (Hematology/Oncology), Germany
| | | | | |
Collapse
|