1
|
Ohe G, Kudo Y, Kamada K, Mouri Y, Takamaru N, Kudoh K, Kurio N, Miyamoto Y. The Soluble Factor from Oral Cancer Cell Lines Inhibits Interferon-γ Production by OK-432 via the CD40/CD40 Ligand Pathway. Cancers (Basel) 2021; 13:cancers13133301. [PMID: 34209347 PMCID: PMC8269085 DOI: 10.3390/cancers13133301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 06/20/2021] [Accepted: 06/28/2021] [Indexed: 11/16/2022] Open
Abstract
(1) Background: OK-432 is a penicillin-killed, lyophilized formulation of a low-toxicity strain (Su) of Streptococcus pyogenes (Group A). It is a potent immunotherapy agent for several types of cancer, including oral cancer. We previously showed that (i) OK-432 treatment induces a high amount of IFN-? production from peripheral blood mononuclear cells (PBMCs), and (ii) conditioned medium (CM) from oral cancer cells suppresses both the IFN-? production and cytotoxic activity of PBMCs driven by OK-432. The aim of this study was to determine the inhibitory mechanism of OK-432-induced IFN-? production from PBMCs by CM. (2) Methods: We performed cDNA microarray analysis, quantitative RT-PCR, and ELISA to reveal the inhibitory mechanism of CM. (3) Results: We found that CD40 plays a key role in IFN-? production via IL-12 production. Although OK-432 treatment upregulated the expression levels of the IL-12p40, p35, and CD40 genes, CM from oral cancer cells downregulate these genes. The amount of IFN-? production by OK-432 treatment was decreased by an anti-CD40 neutralizing antibody. (4) Conclusions: Our study suggests that uncertain soluble factor(s) produced from oral cancer cells may inhibit IFN-? production from PBMCs via suppressing the CD40/CD40L-IL-12 axis.
Collapse
Affiliation(s)
- Go Ohe
- Department of Oral Surgery, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima 770-8504, Japan; (K.K.); (N.T.); (K.K.); (N.K.); (Y.M.)
- Dentistry and Oral Surgery, Takamatsu Municipal Hospital, 847-1 Ko Busshozan-cho, Takamatsu 761-8538, Japan
- Correspondence:
| | - Yasusei Kudo
- Department of Oral Bioscience, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima 770-8504, Japan; (Y.K.); (Y.M.)
| | - Kumiko Kamada
- Department of Oral Surgery, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima 770-8504, Japan; (K.K.); (N.T.); (K.K.); (N.K.); (Y.M.)
| | - Yasuhiro Mouri
- Department of Oral Bioscience, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima 770-8504, Japan; (Y.K.); (Y.M.)
| | - Natsumi Takamaru
- Department of Oral Surgery, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima 770-8504, Japan; (K.K.); (N.T.); (K.K.); (N.K.); (Y.M.)
| | - Keiko Kudoh
- Department of Oral Surgery, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima 770-8504, Japan; (K.K.); (N.T.); (K.K.); (N.K.); (Y.M.)
| | - Naito Kurio
- Department of Oral Surgery, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima 770-8504, Japan; (K.K.); (N.T.); (K.K.); (N.K.); (Y.M.)
| | - Youji Miyamoto
- Department of Oral Surgery, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima 770-8504, Japan; (K.K.); (N.T.); (K.K.); (N.K.); (Y.M.)
| |
Collapse
|
2
|
Vidovic D, Giacomantonio C. Insights into the Molecular Mechanisms Behind Intralesional Immunotherapies for Advanced Melanoma. Cancers (Basel) 2020; 12:cancers12051321. [PMID: 32455916 PMCID: PMC7281646 DOI: 10.3390/cancers12051321] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 05/17/2020] [Accepted: 05/20/2020] [Indexed: 12/12/2022] Open
Abstract
The incidence of cutaneous melanoma, a highly malignant skin cancer, is increasing yearly. While surgical removal of the tumor is the mainstay of treatment for patients with locally confined disease, those with metastases face uncertainty when it comes to their treatment. As melanoma is a relatively immunogenic cancer, current guidelines suggest using immunotherapies that can rewire the host immune response to target melanoma tumor cells. Intralesional therapy, where immunomodulatory agents are injected directly into the tumor, are an emerging aspect of treatment for in-transit melanoma because of their ability to mitigate severe off-target immune-related adverse events. However, their immunomodulatory mechanisms are poorly understood. In this review, we will summarize and discuss the different intralesional therapies for metastatic melanoma with respect to their clinical outcomes and immune molecular mechanisms.
Collapse
|
3
|
Remic T, Sersa G, Ursic K, Cemazar M, Kamensek U. Development of Tumor Cell-Based Vaccine with IL-12 Gene Electrotransfer as Adjuvant. Vaccines (Basel) 2020; 8:vaccines8010111. [PMID: 32121641 PMCID: PMC7157224 DOI: 10.3390/vaccines8010111] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 02/25/2020] [Accepted: 02/27/2020] [Indexed: 02/01/2023] Open
Abstract
Tumor cell-based vaccines use tumor cells as a source of tumor-associated antigens. In our study, we aimed to develop and test a tumor vaccine composed of tumor cells killed by irradiation combined with in vivo interleukin-12 gene electrotransfer as an adjuvant. Vaccination was performed in the skin of B16-F10 malignant melanoma or CT26 colorectal carcinoma tumor-bearing mice, distant from the tumor site and combined with concurrent tumor irradiation. Vaccination was also performed before tumor inoculation in both tumor models and tumor outgrowth was followed. The antitumor efficacy of vaccination in combination with tumor irradiation or preventative vaccination varied between the tumor models. A synergistic effect between vaccination and irradiation was observed in the B16-F10, but not in the CT26 tumor model. In contrast, up to 56% of mice were protected from tumor outgrowth in the CT26 tumor model and none were protected in the B16-F10 tumor model. The results suggest a greater contribution of the therapeutic vaccination to tumor irradiation in a less immunogenic B16-F10 tumor model, in contrast to preventative vaccination, which has shown greater efficacy in a more immunogenic CT26 tumor model. Upon further optimization of the vaccination and irradiation regimen, our vaccine could present an alternative tumor cell-based vaccine.
Collapse
Affiliation(s)
- Tinkara Remic
- Department of Experimental Oncology, Institute of Oncology Ljubljana, Zaloska cesta 2, SI-1000 Ljubljana, Slovenia; (T.R.); (G.S.); (K.U.); (M.C.)
- Faculty of Medicine, University of Ljubljana, Vrazov Trg 2, SI-1000 Ljubljana, Slovenia
| | - Gregor Sersa
- Department of Experimental Oncology, Institute of Oncology Ljubljana, Zaloska cesta 2, SI-1000 Ljubljana, Slovenia; (T.R.); (G.S.); (K.U.); (M.C.)
- Faculty of Health Sciences, University of Ljubljana, Zdravstvena pot 5, SI-1000 Ljubljana, Slovenia
| | - Katja Ursic
- Department of Experimental Oncology, Institute of Oncology Ljubljana, Zaloska cesta 2, SI-1000 Ljubljana, Slovenia; (T.R.); (G.S.); (K.U.); (M.C.)
| | - Maja Cemazar
- Department of Experimental Oncology, Institute of Oncology Ljubljana, Zaloska cesta 2, SI-1000 Ljubljana, Slovenia; (T.R.); (G.S.); (K.U.); (M.C.)
- Faculty of Health Sciences, University of Primorska, Polje 42, SI-6310 Izola, Slovenia
| | - Urska Kamensek
- Department of Experimental Oncology, Institute of Oncology Ljubljana, Zaloska cesta 2, SI-1000 Ljubljana, Slovenia; (T.R.); (G.S.); (K.U.); (M.C.)
- Correspondence:
| |
Collapse
|
4
|
|
5
|
Khammari A, Nguyen JM, Saint-Jean M, Knol AC, Pandolfino MC, Quereux G, Brocard A, Peuvrel L, Saiagh S, Bataille V, Limacher JM, Dreno B. Adoptive T cell therapy combined with intralesional administrations of TG1042 (adenovirus expressing interferon-γ) in metastatic melanoma patients. Cancer Immunol Immunother 2015; 64:805-15. [PMID: 25846669 PMCID: PMC11029588 DOI: 10.1007/s00262-015-1691-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Accepted: 03/23/2015] [Indexed: 01/08/2023]
Abstract
Tumor immune escape has recently been shown to be related to the development of an immune tolerance state of the microenvironment. Cytokines activating the immune system such as IFN-γ can be used to reverse the immune escape and thus to potentiate the efficacy of immunotherapy. A clinical study was conducted in 18 stage IIIc/IV melanoma patients treated with tumor-infiltrating lymphocytes (TILs) in combination with intratumoral TG1042 injection (adenovirus expressing IFN-γ). The primary objective was to investigate the safety of treatment. Secondary objectives were to study the clinical response and translational research. The treatment was well tolerated. Among the 13 patients evaluable for tumor response, 38.5% had an overall objective response (OOR = CR + PR) and disease control rate (DCR = CR + PR + S) of 46%. The clinical response of the 37 targeted lesions led to an OOR of 51% and a DCR of 75%. Translational research on predictive markers did not significantly differ between responder and non-responder patients. However, specifically regarding injected lesions, the clinical response correlated with CD3-/CD56+ NK cells which could be activated by TG1042. Further larger studies of this combined immunotherapy are needed to confirm our findings. Intralesional TG1042 combined with antigen-selected TILs should be discussed.
Collapse
Affiliation(s)
- Amir Khammari
- Dermato-Oncology Department, Nantes Hospital, 1 Place Alexis Ricordeau, 44093 Nantes Cedex 1, France
- Cancer Research Center Nantes-Angers (Inserm U892, CNRS 6299), 9 Quai Moncousu, 44093 Nantes Cedex 1, France
| | - Jean-Michel Nguyen
- Cancer Research Center Nantes-Angers (Inserm U892, CNRS 6299), 9 Quai Moncousu, 44093 Nantes Cedex 1, France
- Biostatistic Department (PIMESP), Hôpital St Jacques - CHU Nantes, 44093 Nantes, France
| | - Melanie Saint-Jean
- Dermato-Oncology Department, Nantes Hospital, 1 Place Alexis Ricordeau, 44093 Nantes Cedex 1, France
- Cancer Research Center Nantes-Angers (Inserm U892, CNRS 6299), 9 Quai Moncousu, 44093 Nantes Cedex 1, France
| | - Anne-Chantal Knol
- Cancer Research Center Nantes-Angers (Inserm U892, CNRS 6299), 9 Quai Moncousu, 44093 Nantes Cedex 1, France
| | - Marie-Christine Pandolfino
- Cancer Research Center Nantes-Angers (Inserm U892, CNRS 6299), 9 Quai Moncousu, 44093 Nantes Cedex 1, France
- Cell and Gene Therapy Unit (UTCG), Nantes Hospital, 1 Place Alexis Ricordeau, 44093 Nantes Cedex 1, France
| | - Gaelle Quereux
- Dermato-Oncology Department, Nantes Hospital, 1 Place Alexis Ricordeau, 44093 Nantes Cedex 1, France
- Cancer Research Center Nantes-Angers (Inserm U892, CNRS 6299), 9 Quai Moncousu, 44093 Nantes Cedex 1, France
| | - Anabelle Brocard
- Dermato-Oncology Department, Nantes Hospital, 1 Place Alexis Ricordeau, 44093 Nantes Cedex 1, France
- Cancer Research Center Nantes-Angers (Inserm U892, CNRS 6299), 9 Quai Moncousu, 44093 Nantes Cedex 1, France
| | - Lucie Peuvrel
- Dermato-Oncology Department, Nantes Hospital, 1 Place Alexis Ricordeau, 44093 Nantes Cedex 1, France
- Cancer Research Center Nantes-Angers (Inserm U892, CNRS 6299), 9 Quai Moncousu, 44093 Nantes Cedex 1, France
| | - Soraya Saiagh
- Cell and Gene Therapy Unit (UTCG), Nantes Hospital, 1 Place Alexis Ricordeau, 44093 Nantes Cedex 1, France
| | - Vincent Bataille
- Transgene SA, Boulevard Gonthier d’Andernach, CS80166, 67405 Illkirch-Graffenstaden, France
| | - Jean-Marc Limacher
- Transgene SA, Boulevard Gonthier d’Andernach, CS80166, 67405 Illkirch-Graffenstaden, France
| | - Brigitte Dreno
- Dermato-Oncology Department, Nantes Hospital, 1 Place Alexis Ricordeau, 44093 Nantes Cedex 1, France
- Cancer Research Center Nantes-Angers (Inserm U892, CNRS 6299), 9 Quai Moncousu, 44093 Nantes Cedex 1, France
- Cell and Gene Therapy Unit (UTCG), Nantes Hospital, 1 Place Alexis Ricordeau, 44093 Nantes Cedex 1, France
| |
Collapse
|
6
|
Bahrambeigi V, Ahmadi N, Moisyadi S, Urschitz J, Salehi R, Haghjooy Javanmard S. PhiC31/PiggyBac modified stromal stem cells: effect of interferon γ and/or tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) on murine melanoma. Mol Cancer 2014; 13:255. [PMID: 25428727 PMCID: PMC4258801 DOI: 10.1186/1476-4598-13-255] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Accepted: 11/14/2014] [Indexed: 01/13/2023] Open
Abstract
Background TRAIL and IFNγ are promising anti-cancer cytokines and it has been shown that IFNγ may sensitize cancer cells to TRAIL. Adipose derived mesenchymal stem cells (ADSCs) are attractive vehicles for delivering anti-cancer agents. In this study, we evaluated the therapeutic potential of PhiC31 (φC31) recombinase and/or piggyBac transposase (pBt) modified ADSCs expressing either TRAIL, IFNγ, or co-expressing TRAIL/IFNγ in mouse models of melanoma. Methods The expression and bioactivity of mouse IFNγ and TRAIL in φC31 and pBt modified cells were confirmed. We examined the effects of modified ADSCs on signal intensity of red fluorescence protein expressed by melanoma cells in subcutaneous tumors or established lung metastases and on survival (6 mice per group). We also conducted a flow cytometric analysis of systemic CD4+CD25+FOXP3+ T regulatory cells (Tregs) and histological analysis of melanoma tumors. Data were analyzed by Student t test, ANOVA, and log-rank tests. All statistical tests were two-sided. Results We demonstrated non-viral DNA-integrating vectors can be used for stable transgene expression. IFNγ inhibited melanoma cell growth in vitro probably via IFNγ-induced JAK/STAT1 signaling pathway activation. Murine TRAIL induced apoptosis in the human cell lines CAOV-4 and Ej-138, while MCF7 and B16F10 cells appeared to be insensitive to TRAIL. Treatment of melanoma cells with IFNγ did not influence their response to TRAIL. In contrast, results from in vivo studies showed that IFNγ-expressing ADSCs, engrafted into tumor stroma, inhibited tumor growth and angiogenesis, prevented systemic increase of Tregs, increased PD-L1 expression and CD8+ infiltration (but not interleukin-2+ cells), and prolonged the survival of mice (68 days, 95% confidence interval [CI] =52 to 86 days compared to 36 days, 95% CI =29 to 39 days for control, P < .001). Conclusions For the first time, we employed DNA integrating vectors for safe and stable modification of MSCs. Our data indicate potential of non-virally modified IFNγ-expressing ADSCs for treatment of melanoma through direct effects of IFNγ. This study may have a significant role in the management of cancer in the future. Electronic supplementary material The online version of this article (doi:10.1186/1476-4598-13-255) contains supplementary material, which is available to authorized users.
Collapse
|
7
|
Senovilla L, Vacchelli E, Garcia P, Eggermont A, Fridman WH, Galon J, Zitvogel L, Kroemer G, Galluzzi L. Trial watch: DNA vaccines for cancer therapy. Oncoimmunology 2014; 2:e23803. [PMID: 23734328 PMCID: PMC3654598 DOI: 10.4161/onci.23803] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2013] [Accepted: 01/28/2013] [Indexed: 12/22/2022] Open
Abstract
The foundation of modern vaccinology dates back to the 1790s, when the English physician Edward Jenner uncovered the tremendous medical potential of prophylactic vaccination. Jenner’s work ignited a wave of nationwide vaccination campaigns abating the incidence of multiple life-threatening infectious diseases and culminating with the eradication of natural smallpox virus, which was definitively certified by the WHO in 1980. The possibility of using vaccines against cancer was first proposed at the end of the 19th century by Paul Ehrlich and William Coley. However, it was not until the 1990s that such a hypothesis began to be intensively investigated, following the realization that the immune system is not completely unresponsive to tumors and that neoplastic cells express immunogenic tumor-associated antigens (TAAs). Nowadays, anticancer vaccines are rapidly moving from the bench to the bedside, and a few prophylactic and therapeutic preparations have already been approved by FDA for use in humans. In this setting, one interesting approach is constituted by DNA vaccines, i.e., TAA-encoding circularized DNA constructs, often of bacterial origin, that are delivered to patients as such or by means of specific vectors, including (but not limited to) liposomal preparations, nanoparticles, bacteria and viruses. The administration of DNA vaccines is most often performed via the intramuscular or subcutaneous route and is expected to cause (1) the endogenous synthesis of the TAA by myocytes and/or resident antigen-presenting cells; (2) the presentation of TAA-derived peptides on the cell surface, in association with MHC class I molecules; and (3) the activation of potentially therapeutic tumor-specific immune responses. In this Trial Watch, we will summarize the results of recent clinical trials that have evaluated/are evaluating DNA vaccines as therapeutic interventions against cancer.
Collapse
Affiliation(s)
- Laura Senovilla
- Institut Gustave Roussy; Villejuif, France ; INSERM; U848; Villejuif, France ; INSERM; U1015 labelisée par la Ligue Nationale contre le Cancer; CICBT507; Villejuif, France
| | | | | | | | | | | | | | | | | |
Collapse
|
8
|
Pol J, Bloy N, Obrist F, Eggermont A, Galon J, Hervé Fridman W, Cremer I, Zitvogel L, Kroemer G, Galluzzi L. Trial Watch: DNA vaccines for cancer therapy. Oncoimmunology 2014; 3:e28185. [PMID: 24800178 PMCID: PMC4008456 DOI: 10.4161/onci.28185] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2014] [Accepted: 02/10/2014] [Indexed: 12/13/2022] Open
Abstract
During the past 2 decades, the possibility that preparations capable of eliciting tumor-specific immune responses would mediate robust therapeutic effects in cancer patients has received renovated interest. In this context, several approaches to vaccinate cancer patients against their own malignancies have been conceived, including the administration of DNA constructs coding for one or more tumor-associated antigens (TAAs). Such DNA-based vaccines conceptually differ from other types of gene therapy in that they are not devised to directly kill cancer cells or sensitize them to the cytotoxic activity of a drug, but rather to elicit a tumor-specific immune response. In spite of an intense wave of preclinical development, the introduction of this immunotherapeutic paradigm into the clinical practice is facing difficulties. Indeed, while most DNA-based anticancer vaccines are well tolerated by cancer patients, they often fail to generate therapeutically relevant clinical responses. In this Trial Watch, we discuss the latest advances on the use of DNA-based vaccines in cancer therapy, discussing the literature that has been produced around this topic during the last 13 months as well as clinical studies that have been launched in the same time frame to assess the actual therapeutic potential of this intervention.
Collapse
Affiliation(s)
- Jonathan Pol
- Gustave Roussy; Villejuif, France ; INSERM, U848; Villejuif, France ; Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers; Paris, France ; Université Paris-Sud/Paris XI; Paris, France
| | - Norma Bloy
- Gustave Roussy; Villejuif, France ; INSERM, U848; Villejuif, France ; Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers; Paris, France ; Université Paris-Sud/Paris XI; Paris, France
| | - Florine Obrist
- Gustave Roussy; Villejuif, France ; INSERM, U848; Villejuif, France ; Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers; Paris, France ; Université Paris-Sud/Paris XI; Paris, France
| | | | - Jérôme Galon
- Université Paris Descartes/Paris V; Sorbonne Paris Cité; Paris, France ; Université Pierre et Marie Curie/Paris VI; Paris, France ; INSERM, UMRS1138; Paris, France ; Laboratory of Integrative Cancer Immunology; Centre de Recherche des Cordeliers; Paris, France
| | - Wolf Hervé Fridman
- Université Pierre et Marie Curie/Paris VI; Paris, France ; INSERM, UMRS1138; Paris, France ; Equipe 13, Centre de Recherche des Cordeliers; Paris, France
| | - Isabelle Cremer
- Université Pierre et Marie Curie/Paris VI; Paris, France ; INSERM, UMRS1138; Paris, France ; Equipe 13, Centre de Recherche des Cordeliers; Paris, France
| | - Laurence Zitvogel
- Gustave Roussy; Villejuif, France ; INSERM, U1015; CICBT507; Villejuif, France
| | - Guido Kroemer
- Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP; Paris, France ; Metabolomics and Cell Biology Platforms, Gustave Roussy; Villejuif, France ; INSERM, U848; Villejuif, France ; Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers; Paris, France ; Université Paris Descartes/Paris V; Sorbonne Paris Cité; Paris, France
| | - Lorenzo Galluzzi
- Gustave Roussy; Villejuif, France ; Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers; Paris, France ; Université Paris Descartes/Paris V; Sorbonne Paris Cité; Paris, France
| |
Collapse
|
9
|
Gene therapy in interventional pulmonology: Interferon gene delivery with focus on thoracic malignancies. ACTA ACUST UNITED AC 2012. [DOI: 10.1007/s13665-011-0008-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
10
|
Yi Y, Noh MJ, Lee KH. Current advances in retroviral gene therapy. Curr Gene Ther 2011; 11:218-28. [PMID: 21453283 PMCID: PMC3182074 DOI: 10.2174/156652311795684740] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2011] [Accepted: 03/15/2011] [Indexed: 12/25/2022]
Abstract
There have been major changes since the incidents of leukemia development in X-SCID patients after the treatments using retroviral gene therapy. Due to the risk of oncogenesis caused by retroviral insertional activation of host genes, most of the efforts focused on the lentiviral therapies. However, a relative clonal dominance was detected in a patient with β-thalassemia Major, two years after the subject received genetically modified hematopoietic stem cells using lentiviral vectors. This disappointing result of the recent clinical trial using lentiviral vector tells us that the current and most advanced vector systems does not have enough safety. In this review, various safety features that have been tried for the retroviral gene therapy are introduced and the possible new ways of improvements are discussed. Additional feature of chromatin insulators, co-transduction of a suicidal gene under the control of an inducible promoter, conditional expression of the transgene only in appropriate target cells, targeted transduction, cell type-specific expression, targeted local administration, splitting of the viral genome, and site specific insertion of retroviral vector are discussed here.
Collapse
|
11
|
Nagane K, Jo JI, Tabata Y. Promoted Adipogenesis of Rat Mesenchymal Stem Cells by Transfection of Small Interfering RNA Complexed with a Cationized Dextran. Tissue Eng Part A 2010; 16:21-31. [DOI: 10.1089/ten.tea.2009.0170] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Affiliation(s)
- Kentaro Nagane
- Department of Biomaterials, Field of Tissue Engineering, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan
| | - Jun-ichiro Jo
- Department of Biomaterials, Field of Tissue Engineering, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan
| | - Yasuhiko Tabata
- Department of Biomaterials, Field of Tissue Engineering, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan
| |
Collapse
|
12
|
|
13
|
|
14
|
Jo JI, Tabata Y. Non-viral gene transfection technologies for genetic engineering of stem cells. Eur J Pharm Biopharm 2008; 68:90-104. [PMID: 17870447 DOI: 10.1016/j.ejpb.2007.04.021] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2007] [Revised: 04/20/2007] [Accepted: 04/20/2007] [Indexed: 10/23/2022]
Abstract
The recent rapid progress of molecular biology together with the steady progress of genome projects has given us some essential and revolutionary information about DNA and RNA to elucidate various biological phenomena at a genetic level. Under these circumstances, the technology and methodology of gene transfection have become more and more important to enhance the efficacy of gene therapy for several diseases. In addition, gene transfection is a fundamental technology indispensable to the further research development of basic biology and medicine regarding stem cells. Stem cells genetically manipulated will enhance the therapeutic efficacy of cell transplantation. In this paper, the carrier and technology of gene delivery are briefly overviewed while the applications to the basic researches of biology and medicine as well as regenerative medical therapy are introduced. A new non-viral carrier and the cell culture system are described to efficiently manipulate stem cells.
Collapse
Affiliation(s)
- Jun-ichiro Jo
- Department of Biomaterials, Field of Tissue Engineering, Institute for Frontier Medical Sciences, Kyoto University, 53 Kawara-cho Shogoin, Sakyo-ku, Kyoto, Japan
| | | |
Collapse
|
15
|
Nashan D, Müller ML, Grabbe S, Wustlich S, Enk A. Systemic therapy of disseminated malignant melanoma: an evidence-based overview of the state-of-the-art in daily routine. J Eur Acad Dermatol Venereol 2007; 21:1305-18. [DOI: 10.1111/j.1468-3083.2007.02475.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
16
|
Minuzzo S, Moserle L, Indraccolo S, Amadori A. Angiogenesis meets immunology: Cytokine gene therapy of cancer. Mol Aspects Med 2007; 28:59-86. [PMID: 17306360 DOI: 10.1016/j.mam.2006.12.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2006] [Accepted: 12/29/2006] [Indexed: 01/19/2023]
Abstract
Delivery of cytokine genes at the tumor site in pre-clinical models has been shown to recruit host inflammatory cells followed by inhibition of tumor growth. This local effect is often accompanied by systemic protection mediated by the immune system, mainly by CD8(+) T and NK cells. On this basis, cytokine gene-transduced tumor cells have widely been used as vaccines in clinical trials, which have shown good safety profiles and some local responses but substantial lack of systemic efficacy. Are these findings the end of the story? Possibly not, if major improvements will be attained in the coming years. These should be directed at the level of gene selection and delivery, in order to identify the optimal cytokine and achieve efficient and durable cytokine expression, and at the level of improving immune stimulation, i.e. by co-administration of co-stimulatory molecules including B7 and CD40, or boosting the expression of tumor antigens or MHC class I molecules. Interestingly, some of the cytokines which have shown encouraging anti-tumor activity, including IFNs, IL-4, IL-12 and TNF-alpha, are endowed with anti-angiogenic or vasculotoxic effects, which may significantly contribute to local tumor control. Therapeutic exploitation of this property may result in the design of novel approaches which, by maximizing immune-stimulating and anti-angiogenic effects, could possibly lead to starvation of established tumors in patients.
Collapse
Affiliation(s)
- Sonia Minuzzo
- Department of Oncology and Surgical Sciences, University of Padova, via Gattamelata 64, 35128 Padova, Italy
| | | | | | | |
Collapse
|
17
|
Rodrigues T, Carrondo MJT, Alves PM, Cruz PE. Purification of retroviral vectors for clinical application: Biological implications and technological challenges. J Biotechnol 2007; 127:520-41. [PMID: 16950534 DOI: 10.1016/j.jbiotec.2006.07.028] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2006] [Revised: 07/12/2006] [Accepted: 07/25/2006] [Indexed: 11/16/2022]
Abstract
For centuries mankind led a difficult battle against viruses, the smallest infectious agents at the surface of the earth. Nowadays it is possible to use viruses for our benefit, both at a prophylactic level in the production of vaccines and at a therapeutic level in the promising field of gene therapy. Retroviruses were discovered at the end of the 19th century and constitute one of the most effective entities for gene transfer and insertion into the genome of mammalian cells. This attractive feature has intensified research in retroviral vectors development and production over the past years, mainly due to the expectations raised by the concept of gene therapy. The demand for high quality retroviral vectors that meet standard requisites from the regulatory agencies (FDA and EMEA) is therefore increasing, as the technology has moved into clinical trials. The development of safer producer cell lines that can be used in large-scale production will result in the production of large quantities of retroviral stocks. Cost-efficient and scalable purification processes are essential for production of injectable-grade preparations to achieve final implementation of these vectors as therapeutics. Several preparative purification steps already established for proteins can certainly be applied to retroviral vectors, in particular membrane filtration and chromatographic methods. Nevertheless, the special properties of these complex products require technological improvement of the existing purification steps and/or development of particular purification steps to increase productivity and throughput, while maintaining biological activity of the final product. This review focuses on downstream process development in relation to the retroviral vectors characteristics and quality assessment of retroviral stocks for intended use in gene therapy.
Collapse
Affiliation(s)
- Teresa Rodrigues
- IBET/ITQB, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal
| | | | | | | |
Collapse
|
18
|
Dietrich A, Becherer L, Brinckmann U, Hauss J, Liebert UG, Gütz A, Aust G. Particle-mediated cytokine gene therapy leads to antitumor and antimetastatic effects in mouse carcinoma models. Cancer Biother Radiopharm 2006; 21:333-41. [PMID: 16999599 DOI: 10.1089/cbr.2006.21.333] [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] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND We investigated the effects of continuous cancer gene therapy including (antigen-presenting cell) (APC) engineering and local stimulation of the immune system. MATERIALS AND METHODS Lewis lung carcinomas and B16 melanomas, intradermally established on C57/Bl6 mice, were shot using a gene gun every 4th day with a combination of plasmids. The first therapy group received plasmids coding the genes for interleukin (IL)-12 and IL-2. The second therapy group was treated with plasmids coding for B7.1 interferon-gamma (IFN-gamma)/IL-12 alternated by a plasmid coding IL-2. Control were mice without any therapy or treatment with the empty plasmid. RESULTS Gene therapy led to reduced tumor sizes in the therapy groups of both models (significant for the Lewis lung carcinoma). We found an enhanced survival and reduced tumor growth rate in the therapy groups; however, the effects were not significant. IL- 12/IL-2 therapy was more effective, compared to B7.1/IFN-gamma/IL-12 and IL-2. Cytokine gene transfer let to a significantly lower metastasis rate in Lewis lung carcinoma. CONCLUSIONS Continuous particle-mediated gene transfer is easy to handle and shows good results. Gene therapy combining the genes coding for IL-12 and IL-2 was superior to additional IFN-gamma/B7.1. APC engineering does not appear to be sufficient in these poorly antigenic tumors.
Collapse
Affiliation(s)
- Arne Dietrich
- Clinic for Visceral, Thoracic, Vascular and Transplant Surgery, University of Leipzig, Leipzig, Germany.
| | | | | | | | | | | | | |
Collapse
|
19
|
Abstract
GVAX is a granulocyte-macrophage colony-stimulating factor (GM-CSF) gene-transfected tumor cell vaccine. Original work with GM-CSF as a recombinant DNA protein (Leukine) involved proliferative stimulation of macrophages and neutrophils for the purpose of reducing hematopoietic toxicity related to dose-intensive chemotherapy. Following US Food and Drug Administration approval of Leukine several years ago, extensive preclinical results have demonstrated an immunostimulatory effect related to GM-CSF gene when transfected into tumor cells and used as a vaccine (GVAX). Tumor regression and prolonged survival was demonstrated in animal models. Toxicology with GVAX indicated no adverse effects, which enabled further testing in cancer patients. A small number of responses were demonstrated in Phase I trials in immunosensitive cancer patients (renal cell carcinoma and melanoma). However, a series of dramatic complete and durable responses in advanced non-small cell lung cancer patients, demonstrated in recent clinical trials, have generated interest in further development of this vaccine in nontraditional cancer disease types. The rationale of GVAX development and a summary of clinical results are reviewed.
Collapse
|
20
|
Rodrigues T, Carvalho A, Roldão A, Carrondo MJT, Alves PM, Cruz PE. Screening anion-exchange chromatographic matrices for isolation of onco-retroviral vectors. J Chromatogr B Analyt Technol Biomed Life Sci 2006; 837:59-68. [PMID: 16697280 DOI: 10.1016/j.jchromb.2006.03.061] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2005] [Revised: 03/01/2006] [Accepted: 03/30/2006] [Indexed: 11/25/2022]
Abstract
The adsorption kinetics of retroviral vectors to several chromatographic media, DEAE FF, Streamlinetrade mark Q XL and CHTtrade mark Ceramic Hydroxyapatite, in batch mode was investigated. The effects of buffer type, pH and operational temperature were studied. A mathematical model describing viral adsorption kinetics that considers viral degradation in solution was developed. The best results, either in terms of speed and extent of adsorbed infectious particles, were obtained with DEAE FF and Streamlinetrade mark Q XL. Fixed-bed chromatography was further investigated using DEAE FF, Q XL and Q FF, for validation of the batch adsorption process. Fixed-bed DEAE FF and Q XL proved to be good candidates for purification of MoMLV derived vectors due to resulting high yields, 53+/-13% and 51+/-7%, respectively, while removing more than 99% of protein and 90% of the DNA contaminants.
Collapse
|
21
|
Tan PH, Chan CLH, Chan C, George AJT. The evolving role of gene-based treatment in surgery. Br J Surg 2005; 92:1466-80. [PMID: 16273530 DOI: 10.1002/bjs.5181] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Abstract
Background
The completion of the sequencing of the human genome in 2003 marked the dawn of a new era of human biology and medicine. Although these remarkable scientific advances improve the understanding of human biology, the question remains how this rapidly expanding knowledge of functional genomics affects the role of surgeons. This article reviews the potential therapeutic application of gene therapy for various surgical conditions.
Methods
The core of this review was derived from a Medline database literature search.
Results and conclusion
The currently available vectors in the field of gene therapy and their limitations for clinical applications were analysed. The achievements of gene therapy in clinical trials and the future ramifications for surgery were also explored. Whether gene therapy takes a major role in surgical practice will depend greatly on the success of future vector development. Advances in viral vector technology to reduce the inflammatory effect, and improvements in the efficiency of gene delivery using non-viral vector technology, would allow this form of therapy to become more clinically applicable.
Collapse
Affiliation(s)
- P H Tan
- Department of Surgery, Stoke Mandeville Hospital, South Buckinghamshire NHS Trust, Aylesbury, UK.
| | | | | | | |
Collapse
|
22
|
Nemunaitis J, Nemunaitis J. Granulocyte-macrophage colony-stimulating factor gene-transfected autologous tumor cell vaccine: focus[correction to fcous] on non-small-cell lung cancer. Clin Lung Cancer 2004; 5:148-57. [PMID: 14667270 DOI: 10.3816/clc.2003.n.027] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Traditionally, non-small-cell lung cancer (NSCLC) is not thought of as an immunosensitive malignancy. However, recent clinical results with GVAX, a granulocyte-macrophage colony-stimulating factor (GM-CSF) gene-transduced autologous tumor vaccine, may suggest otherwise. This review summarizes immune-induced activity caused by GM-CSF protein and GM-CSF gene-transfected vaccines. Initial indication of use for GM-CSF protein (sargramostim) was to improve neutrophil recovery following cytotoxic chemotherapy. However, several trials involving patients with hematologic malignancy demonstrated improvement in survival related to delayed disease progression in patients receiving sargramostim in combination with chemotherapy. Subsequently, others explored potential antitumor activity with sargramostim in a variety of trials. Results did not consistently demonstrate sufficient antitumor activity to justify routine use of sargramostim as an anticancer agent. Preclinical work with GM-CSF gene-transfected vaccines, however, did demonstrate significant activity, thereby justifying clinical investigation. Patients with metastatic NSCLC who had previously failed chemotherapy demonstrated response to GVAX (3 of 33 complete responses) and dose-related improvement in survival (471 days vs. 174 days).
Collapse
|
23
|
Abstract
The field of cancer gene therapy is in continuous expansion, and technology is quickly moving ahead as far as gene targeting and regulation of gene expression are concerned. This review focuses on the endocrine aspects of gene therapy, including the possibility to exploit hormone and hormone receptor functions for regulating therapeutic gene expression, the use of endocrine-specific genes as new therapeutic tools, the effects of viral vector delivery and transgene expression on the endocrine system, and the endocrine response to viral vector delivery. Present ethical concerns of gene therapy and the risk of germ cell transduction are also discussed, along with potential lines of innovation to improve cell and gene targeting.
Collapse
Affiliation(s)
- Luisa Barzon
- Department of Histology, Microbiology, and Medical Biotechnologies, University of Padova, I-35121 Padua, Italy
| | | | | |
Collapse
|
24
|
Kortylewski M, Komyod W, Kauffmann ME, Bosserhoff A, Heinrich PC, Behrmann I. Interferon-γ-Mediated Growth Regulation of Melanoma Cells: Involvement of STAT1-Dependent and STAT1-Independent Signals. J Invest Dermatol 2004; 122:414-22. [PMID: 15009724 DOI: 10.1046/j.0022-202x.2004.22237.x] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Interferon-gamma, a known inhibitor of tumor cell growth, has been used in several protocols for the treatment of melanoma. We have studied the molecular events underlying interferon-gamma-induced G0/G1 arrest in four metastatic melanoma cell lines with different responsiveness to interferon-gamma. The growth arrest did not result from enhanced expression of cyclin-dependent kinase inhibitors p21 and p27. Instead, it correlated with downregulation of cyclin E and cyclin A and inhibition of their associated kinase activities. We show that interferon-gamma-induced growth inhibition could be abrogated by overexpression of dominant negative STAT1 (signal transducer and activator of transcription 1) in the melanoma cell line A375, suggesting that STAT1 plays a crucial part for the anti-proliferative effect. Erythropoietin stimulation of a chimeric receptor led to a concentration-dependent STAT1 activation and concomitant growth arrest when it contained the STAT recruitment motif Y440 of the interferon-gamma receptor 1. In contrast, dose-response studies for interferon-gamma revealed a discrepancy between levels of STAT1 activation and the extent of growth inhibition; whereas STAT1 was activated by low doses of interferon-gamma (10 U per mL), growth inhibitory effects were only visible with 100-fold higher concentrations. Our results suggest the presence of additional signals emanating from the interferon-gamma receptor, which may counteract the anti-proliferative function of STAT1.
Collapse
|
25
|
Abstract
The potential for gene therapy to cure a wide range of diseases has lead to high expectations and a great increase in research efforts in this area. At present, viral vectors are the most efficient means of delivering a corrective gene into human cells. While a number of different viral vectors are under development, retroviral vectors are currently the most common type used in clinical trials today. However, the production of retroviral vectors for gene therapy applications faces a number of challenges. Of primary concern is the low titre of vector stocks produced by packaging cells in culture and the inherent instability of retroviral vector activity. The problems facing large-scale retroviral vector production are outlined in this review and the research efforts by a number of groups who have attempted to optimise production methods are presented.
Collapse
Affiliation(s)
- Sally McTaggart
- Animal Cell Technology Group, School of Chemical Engineering, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | | |
Collapse
|
26
|
Sakurai F, Terada T, Maruyama M, Watanabe Y, Yamashita F, Takakura Y, Hashida M. Therapeutic effect of intravenous delivery of lipoplexes containing the interferon-beta gene and poly I: poly C in a murine lung metastasis model. Cancer Gene Ther 2003; 10:661-8. [PMID: 12944985 DOI: 10.1038/sj.cgt.7700617] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
We have evaluated and compared the efficacy of systemic administration of lipoplex formulations containing plasmids encoding IFN-beta or IFN-gamma, and a synthetic double-strand RNA poly I:poly C (pI:pC), a type I IFN inducer, in a lung metastasis model in which colon carcinoma CT-26 cells were inoculated intravenously into immunocompatible mice. Injection of lipoplexes containing plasmid DNA, regardless of IFN gene insertion, stimulated a transient increase in the serum concentration of proinflammatory cytokines such as tumor necrosis factor (TNF)-alpha and IFN-gamma, while injection of lipoplexes containing pI:pC led to a low level of TNF-alpha and undetectable IFN-gamma production. Furthermore, injection of these lipoplexes containing plasmids resulted in the production of a mixture of type I and type II IFNs, partly derived from the inserted IFN genes, in lung tissue cultures. In tumor-prophylactic experiments, intravenous injection of lipoplexes containing plasmid, regardless of IFN gene insertion, showed a significant reduction in lung metastatic nodules probably due to proinflammatory cytokines such as TNF-alpha and IFN-gamma nonspecifically induced by the CpG motifs in the plasmid and the type I IFNs produced. On the other hand, the antimetastatic effect of pI:pC-lipoplex seemed to be due mainly to IFN-beta induced by pI:pC. In established lung metastasis experiments, a single intravenous administration of lipoplexes containing IFN-beta gene or pI:pC, but not other lipoplexes, showed a significant therapeutic effect on the tumor metastasis: reduction in tumor nodules and prolongation of survival time of tumor-burden mice. The therapeutic effects were specifically impaired by anti-IFN-beta antibody treatment, indicating that IFN-beta produced by the lipoplexes played an important role in the suppression of established metastatic lung tumors. Thus, the local IFN-beta in the lung delivered by intravenous administration of lipoplex containing IFN-beta gene or pI:pC may be a convenient and useful method of inhibiting established metastatic lung tumors.
Collapse
Affiliation(s)
- Fuminori Sakurai
- Department of Drug Delivery Research, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
| | | | | | | | | | | | | |
Collapse
|
27
|
Harrington KJ, Melcher AA, Bateman AR, Ahmed A, Vile RG. Cancer gene therapy: Part 2. Candidate transgenes and their clinical development. Clin Oncol (R Coll Radiol) 2002; 14:148-69. [PMID: 12069125 DOI: 10.1053/clon.2001.0004] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Kevin J Harrington
- CRC Centre for Cell and Molecular Biology, Institute for Cancer Research, London, UK.
| | | | | | | | | |
Collapse
|
28
|
Nemunaitis J, O'Brien J. Head and neck cancer: gene therapy approaches. Part II: genes delivered. Expert Opin Biol Ther 2002; 2:311-24. [PMID: 11890870 DOI: 10.1517/14712598.2.3.311] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
In Part I, the review summarised the safety of adenoviral vectors and provided insight into approaches being undertaken to improve the specificity, durability and potency of adenoviral delivery vehicles. In Part II, brief discussions are held regarding results of preclinical and clinical trials with a variety of different genes, which have demonstrated antitumour activity in squamous cell carcinoma of the head and neck region (HNSCC). Studies have been performed with a variety of immune modulatory genes. Preliminary results demonstrate activity with several cytokine genes, tumour antigen genes and co-stimulatory molecule genes. Despite only preliminary results, thus far, a theoretical attractive feature for the use of gene therapy for the enhancement of immune modulation is that local injection of the gene product appears to be well tolerated. It is also successful in inducing systemic immune response, potentially providing effect to metastatic sites distal from the injected site. Animal studies have confirmed efficacy in the use of specific targeting of molecules regulating cancer growth (EGF receptor [EGFR], super oxide dismutase [SOD], cyclin D1, E1A and Bcl-2). These approaches are discussed. However, the most significant clinical advances for the use of gene therapy in advanced HNSCC involves two agents: Adp53 and ONYX-015. Preliminary Phase I and II results suggest evidence of efficacy and justify accrual Phase III trials, which are currently ongoing.
Collapse
Affiliation(s)
- John Nemunaitis
- 3535 Worth Street, Collins Building, 5th Floor, Dallas, Texas 75246, USA.
| | | |
Collapse
|
29
|
Iwai M, Harada Y, Tanaka S, Muramatsu A, Mori T, Kashima K, Imanishi J, Mazda O. Polyethylenimine-mediated suicide gene transfer induces a therapeutic effect for hepatocellular carcinoma in vivo by using an Epstein-Barr virus-based plasmid vector. Biochem Biophys Res Commun 2002; 291:48-54. [PMID: 11829460 DOI: 10.1006/bbrc.2002.6383] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
The present study aimed to establish a novel efficient nonviral strategy for suicide gene transfer in hepatocellular carcinoma (HCC) in vivo. We employed branched polyethylenimine (PEI) and combined it with Epstein-Barr virus (EBV)-based plasmid vectors. The HCC cells transfected with an EBV-based plasmid carrying the herpes simplex virus-1 thymidine kinase (HSV-1 Tk) gene (pSES.Tk) showed up to 30-fold higher susceptibilities to ganciclovir (GCV) than those transfected with a conventional plasmid vector carrying the HSV-1 Tk gene (pS.Tk). The therapeutic effect in vivo was tested by intratumoral injection of the plasmids into HuH-7 hepatomas transplanted into C.B-17 scid/scid mutant (SCID) mice and subsequent GCV administrations. Treatment with pSES.Tk, but not pS.Tk, markedly suppressed growth of hepatomas in vivo, resulting in a significantly prolonged survival period of the mice. These findings suggest that PEI-mediated gene transfer system can confer efficient expression of the suicide gene in HCC cells in vivo by using EBV-based plasmid vectors.
Collapse
Affiliation(s)
- Masaki Iwai
- Third Department of Internal Medicine, Department of Microbiology, Kyoto Prefectural University of Medicine, Kamikyo-ku, Kyoto, 602-0841, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Younes HM, Amsden BG. Interferon-gamma therapy: evaluation of routes of administration and delivery systems. J Pharm Sci 2002; 91:2-17. [PMID: 11782893 DOI: 10.1002/jps.10007] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Although different routes and delivery systems have been used to deliver interferon-gamma (IFN-gamma) for the treatment of a variety of viral and neoplastic diseases, little has been reported regarding the most efficient and least toxic routes and drug delivery modes required to achieve these goals. To have a greater understanding of the best strategies to use to administer this cytokine in an efficient, stable, and safe manner, this review details aspects of IFN-gamma concerning its mechanism of action, physical properties, and pharmacokinetics. One important conclusion that is drawn from this analysis is that a consistent, local concentration of IFN-gamma is necessary to achieve an optimal therapeutic response. A critical discussion covering the advantages and limitations of the currently used methodologies to deliver IFN-gamma in such a fashion is presented.
Collapse
Affiliation(s)
- Husam M Younes
- Faculty of Pharmacy and Pharmaceutical Science, University of Alberta, Edmonton, Alberta T6G 2N8, Canada
| | | |
Collapse
|
31
|
Yu D, Thomas-Tikhonenko A. Intratumoral delivery of an interferon gamma retrovirus-producing cells inhibits growth of a murine melanoma by a non-immune mechanism. Cancer Lett 2001; 173:145-54. [PMID: 11597789 DOI: 10.1016/s0304-3835(01)00612-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Interferon gamma (IFNgamma) is a potent inhibitor of cell growth effective against a wide range of tumor-derived cell lines. We cloned murine IFNgamma cDNA into a retroviral vector and created a packaging cell line (Am-gamma) producing this IFNgamma-encoding retrovirus. In a pilot experiment, admixing and co-injection of equal numbers of retrovirus-producing and target B16 melanoma cells led to high rates of infection and strong suppression of neoplastic growth. This effect was observed in the absence of measurable systemic production of IFNgamma and could be reproduced in animals lacking cytotoxic immune responses. Tumor angiogenesis was unaffected and no increase in apoptosis was apparent; however, mitotic indices were greatly reduced in Am-gamma-containing abortive tumors. We thus concluded that IFNgamma directly affects proliferation of B16 cells. Indeed, exposure of B16 cells to IFNgamma in vitro inhibits cell division, as measured by a thymidine incorporation assay. Most importantly, repeated intratumoral injections of Am-gamma stunted growth of established B16 melanomas in 75% of treated animals. Thus, this approach can serve as a prototype for new anti-cancer modalities.
Collapse
Affiliation(s)
- D Yu
- Department of Pathobiology, University of Pennsylvania, 3800 Spruce Street, 368E, Philadelphia, PA 19104-6051, USA
| | | |
Collapse
|
32
|
Abstract
Modification of the tumor microenvironment with gene transfer techniques stimulates two immune mechanisms that effectuate tumor destruction. One involves improved tumor-antigen presentation for the development of specific cellular and humoral immunity. The second involves compromise of the tumor vasculature by soluble factors and leukocytes.
Collapse
Affiliation(s)
- N Mach
- Department of Adult Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | | |
Collapse
|
33
|
Okamoto M, Ohe G, Oshikawa T, Nishikawa H, Furuichi S, Bando T, Yoshida H, Sakai T, Himeno K, Sato M, Ohkubo S. Purification and characterization of cytokine-inducing protein of seed extract from Aeginetia indica L., a parasitic plant. IMMUNOPHARMACOLOGY 2000; 49:377-89. [PMID: 10996035 DOI: 10.1016/s0162-3109(00)00253-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
We have isolated 55 kDa protein from the seed extract of Aeginetia indica L. (AIL), a parasitic plant, by an affinity chromatography on N-hydroxysuccinimide (NHS)-activated Sepharose High Performance column bound F3 monoclonal antibody which neutralizes cytokine-inducing and antitumor effect of AIL. In in vitro model using human peripheral blood mononuclear cells (PBMC), the 55 kDa protein (AILb-A) induced multiple cytokines, such as IFN-gamma, tumor necrosis factor (TNF)-alpha, granulocyte macrophage-colony stimulating factor (GM-CSF), IL-2, IL-6, IL-10, IL-12 and IL-18, and also accelerated killer cell activities of PBMC. When compared with a commonly used immunotherapeutic agent OK-432, AILb-A induced Th1 cytokines are greater than OK-432. Of the Th2 cytokines, the amounts of IL-6 and IL-10 induced by AILb-A were lower than those by OK-432. No significant induction of IL-4 and IL-13 was observed in AILb-A-stimulated PBMC. TNF family including TNF-alpha, TNF-beta, Fas ligand (FasL) and TNF-related apoptosis-inducing ligand (TRAIL) were suggested to be important for AILb-A-induced killing activity of PBMC by reverse transcription-polymerase chain reaction (RT-PCR) analysis. Furthermore, the neutralizing test using cytokine-specific antibodies demonstrated that IL-18 plays a most significant role for IFN-gamma- and killer cell-inducing ability of AILb-A among the cytokines tested. These findings clearly indicated that AILb-A, a 55 kDa protein of AIL, is a potent Th1 cytokine inducer and may be a useful immunotherapeutic agent for the patients with malignancies.
Collapse
MESH Headings
- Adjuvants, Immunologic/isolation & purification
- Adjuvants, Immunologic/metabolism
- Adjuvants, Immunologic/pharmacology
- Antibodies, Monoclonal/metabolism
- Caspase 1/metabolism
- Cells, Cultured
- Cytokines/biosynthesis
- Cytokines/immunology
- Cytokines/metabolism
- Enzyme Activation/drug effects
- Humans
- Immune Sera/pharmacology
- Interleukin-18/metabolism
- Killer Cells, Natural/immunology
- Killer Cells, Natural/metabolism
- Leukocytes, Mononuclear/drug effects
- Leukocytes, Mononuclear/immunology
- Leukocytes, Mononuclear/metabolism
- Lymphocyte Activation/drug effects
- Picibanil/pharmacology
- Plant Proteins/isolation & purification
- Plant Proteins/metabolism
- Plant Proteins/pharmacology
- Seeds/chemistry
- Th1 Cells/immunology
- Th1 Cells/metabolism
Collapse
Affiliation(s)
- M Okamoto
- Second Department of Oral and Maxillofacial Surgery, Tokushima University School of Dentistry, 3-18-15 Kuramoto-cho, 7708504, Tokushima, Japan.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
|
35
|
Nemunaitis J, Kuhn JA. Immune Modulation as Cancer Treatment Using Gene Therapy. Proc (Bayl Univ Med Cent) 1999. [DOI: 10.1080/08998280.1999.11930181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
Affiliation(s)
- John Nemunaitis
- Mary Crowley Medical Research Center, Dallas, Texas
- US Oncology, Dallas, Texas
- Baylor University Medical Center, Dallas, Texas
| | - Joseph A. Kuhn
- Mary Crowley Medical Research Center, Dallas, Texas
- Baylor University Medical Center, Dallas, Texas
| |
Collapse
|