1
|
Mochizuki K, Kobayashi S, Takahashi N, Sugimoto K, Sano H, Ohara Y, Mineishi S, Zhang Y, Kikuta A. Alloantigen-activated (AAA) CD4 + T cells reinvigorate host endogenous T cell immunity to eliminate pre-established tumors in mice. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2021; 40:314. [PMID: 34625113 PMCID: PMC8499505 DOI: 10.1186/s13046-021-02102-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 09/12/2021] [Indexed: 11/10/2022]
Abstract
BACKGROUND Cancer vaccines that induce endogenous antitumor immunity represent an ideal strategy to overcome intractable cancers. However, doing this against a pre-established cancer using autologous immune cells has proven to be challenging. "Allogeneic effects" refers to the induction of an endogenous immune response upon adoptive transfer of allogeneic lymphocytes without utilizing hematopoietic stem cell transplantation. While allogeneic lymphocytes have a potent ability to activate host immunity as a cell adjuvant, novel strategies that can activate endogenous antitumor activity in cancer patients remain an unmet need. In this study, we established a new method to destroy pre-developed tumors and confer potent antitumor immunity in mice using alloantigen-activated CD4+ (named AAA-CD4+) T cells. METHODS AAA-CD4+ T cells were generated from CD4+ T cells isolated from BALB/c mice in cultures with dendritic cells (DCs) induced from C57BL/6 (B6) mice. In this culture, allogeneic CD4+ T cells that recognize and react to B6 mouse-derived alloantigens are preferentially activated. These AAA-CD4+ T cells were directly injected into the pre-established melanoma in B6 mice to assess their ability to elicit antitumor immunity in vivo. RESULTS Upon intratumoral injection, these AAA-CD4+ T cells underwent a dramatic expansion in the tumor and secreted high levels of IFN-γ and IL-2. This was accompanied by markedly increased infiltration of host-derived CD8+ T cells, CD4+ T cells, natural killer (NK) cells, DCs, and type-1 like macrophages. Selective depletion of host CD8+ T cells, rather than NK cells, abrogated this therapeutic effect. Thus, intratumoral administration of AAA-CD4+ T cells results in a robust endogenous CD8+ T cell response that destroys pre-established melanoma. This locally induced antitumor immunity elicited systemic protection to eliminate tumors at distal sites, persisted over 6 months in vivo, and protected the animals from tumor re-challenge. Notably, the injected AAA-CD4+ T cells disappeared within 7 days and caused no adverse reactions. CONCLUSIONS Our findings indicate that AAA-CD4+ T cells reinvigorate endogenous cytotoxic T cells to eradicate pre-established melanoma and induce long-term protective antitumor immunity. This approach can be immediately applied to patients with advanced melanoma and may have broad implications in the treatment of other types of solid tumors.
Collapse
Affiliation(s)
- Kazuhiro Mochizuki
- Department of Pediatric Oncology, Fukushima Medical University Hospital, 1 Hikarigaoka, 960-1295, Fukushima City, Japan.
| | - Shogo Kobayashi
- Department of Pediatric Oncology, Fukushima Medical University Hospital, 1 Hikarigaoka, 960-1295, Fukushima City, Japan
| | - Nobuhisa Takahashi
- Department of Pediatric Oncology, Fukushima Medical University Hospital, 1 Hikarigaoka, 960-1295, Fukushima City, Japan
| | - Kotaro Sugimoto
- Department of Basic Pathology, Fukushima Medical University, Fukushima, Japan
| | - Hideki Sano
- Department of Pediatric Oncology, Fukushima Medical University Hospital, 1 Hikarigaoka, 960-1295, Fukushima City, Japan
| | - Yoshihiro Ohara
- Department of Pediatric Oncology, Fukushima Medical University Hospital, 1 Hikarigaoka, 960-1295, Fukushima City, Japan
| | - Shin Mineishi
- Department of Medicine, Penn State Hershey Medical Center, Hershey, Pennsylvania, USA
| | - Yi Zhang
- Fels Institute for Cancer Research and Molecular Biology, Temple University, Philadelphia, USA.,Department of Cancer and Cellular Biology, Temple University, Philadelphia, USA
| | - Atsushi Kikuta
- Department of Pediatric Oncology, Fukushima Medical University Hospital, 1 Hikarigaoka, 960-1295, Fukushima City, Japan
| |
Collapse
|
2
|
Ayaz F, Ersan RH, Kuzu B, Algul O. New-Generation Benzimidazole-Based Plasmid Delivery Reagents with High Transfection Efficiencies on the Mammalian Cells. In Vitro Cell Dev Biol Anim 2020; 56:34-41. [DOI: 10.1007/s11626-019-00418-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 10/31/2019] [Indexed: 12/11/2022]
|
3
|
Abstract
Melanoma has a unique propensity for locoregional metastasis secondary to intralymphatic transit not seen in other cutaneous or soft tissue malignancies. Novel intralesional therapies using oncolytic immunotherapy exhibit increasing response rates with observed bystander effect. Intralesional modalities in combination with systemic immunotherapy are the subject of ongoing clinical trials. Regional therapy is used in isolated limb locoregional metastasis whereby chemotherapy is delivered to an isolated limb avoiding systemic side effects. Multimodal treatment strategy is imperative in the treatment of locoregionally advanced melanoma. One must be versed on these quickly evolving therapeutic options.
Collapse
Affiliation(s)
- David T Pointer
- Department of Cutaneous Oncology, Moffitt Cancer Center, 10920 McKinley Drive, Tampa, FL, 33612; Department of Surgery, University of South Florida Morsani College of Medicine, 13220 USF Laurel Dr., Tampa, FL 33612
| | - Jonathan S Zager
- Department of Cutaneous Oncology, Moffitt Cancer Center, 10920 McKinley Drive, Tampa, FL, 33612; Department of Surgery, University of South Florida Morsani College of Medicine, 13220 USF Laurel Dr., Tampa, FL 33612.
| |
Collapse
|
4
|
Miura JT, Zager JS. Intralesional therapy as a treatment for locoregionally metastatic melanoma. Expert Rev Anticancer Ther 2018; 18:399-408. [PMID: 29466885 DOI: 10.1080/14737140.2018.1444482] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION The emergence of novel intralesional therapies have dramatically changed the treatment landscape for melanoma. The heterogeneous presentation of melanoma continues to pose challenges for clinicians, especially when dealing with advanced locoregional disease. Intralesional therapies have the benefit of causing local tumor destruction, while minimizing systemic toxicity. Moreover, the integration of immunotherapeutic agents into intralesional compounds has resulted in the additional benefit of a bystander effect, whereby untreated distant lesions also derive a benefit from treatment. Intralesional therapy has assumed an important role in the management of unresectable, locoregional disease for melanoma. Areas covered: Multiple intralesional agents have been studied over the years, with only a few demonstrating promising results. This review will provide an overview of the different intralesional agents for melanoma. Mechanisms of action, clinical efficacy, and side effects will be the primary focus. Expert commentary: Treatment options for advanced melanoma continue to evolve. Attractive new therapies delivered by an intralesional route has demonstrated promising results, with minimal side effects. The ideal treatment strategy for melanoma will remain a multimodal approach; intralesional therapy provides an additional tool in the treatment armamentarium for melanoma.
Collapse
Affiliation(s)
- John T Miura
- a Departments of Cutaneous Oncology and Sarcoma, Moffitt Cancer Center , University of South Florida School of Medicine , Tampa , FL , USA
| | - Jonathan S Zager
- a Departments of Cutaneous Oncology and Sarcoma, Moffitt Cancer Center , University of South Florida School of Medicine , Tampa , FL , USA
| |
Collapse
|
5
|
Sloot S, Rashid OM, Sarnaik AA, Zager JS. Developments in Intralesional Therapy for Metastatic Melanoma. Cancer Control 2016; 23:12-20. [PMID: 27009452 DOI: 10.1177/107327481602300104] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND Locoregional advanced melanoma poses a complex clinical challenge that requires a multidisciplinary, patient-centered approach. Numerous agents have been studied for their suitability as intralesional therapy in the past decades, but few have successfully completed phase 3 clinical trial testing. METHODS The relevant medical literature was searched for articles regarding use of intralesional therapies in metastatic melanoma. Therapies with data from phase 2 or higher studies were selected for review. This review also summarizes the mechanisms of action, adverse-event profiles, and clinical data for these agents. RESULTS Intralesional therapies demonstrate promising effects in select patients with advanced melanoma. The optimal approach should be individually tailored and consist of a combination of intralesional therapies, regional perfusions, systemic immunotherapies, targeted therapies, and surgery, if necessary. CONCLUSIONS Due to its relatively good local response rates and tolerable adverse-event profile, intralesional therapy may be a treatment option for select patients with unresectable, locally advanced or metastatic melanoma.
Collapse
Affiliation(s)
| | | | | | - Jonathan S Zager
- Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa, FL 33612, USA.
| |
Collapse
|
6
|
Abstract
Background: Gene therapy is an innovative and exciting new branch of medicine. Despite the fact that a human disease has yet to be cured using this therapeutic approach, numerous clinical trials are taking place around the world based on encouraging preclinical data. Objective: The aim of this review is to bring the reader up to date with this rapidly advancing field and to highlight the technical advances that must occur before gene therapy will become common practice in dermatology. Methods: The current level of gene delivery technology restricts the applications. The advantages and disadvantages of viral and nonviral gene delivery systems are discussed. Results: Considerable advances are being made in the areas of cancer immunotherapy and vaccines. Of particular importance to the treatment of skin diseases will be the isolation and ex vivo manipulation of epidermal stem cells, the development of skin-specific regulatory sequences for gene expression, and the formulation of gene delivery systems suitable for systemic administration. Conclusions: In general, skin and keratinocytes are considered to be good targets for gene transfer applications, and several diseases have been identified as potential candidates for treatment in the near future.
Collapse
Affiliation(s)
- Michael J. Hope
- Skin Barrier Research Laboratory, Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
7
|
Abstract
INTRODUCTION Intralesional therapy for metastatic melanoma has some advantages over systemic therapy. Local drug administration allows for delivery of an increased concentration of the agent and reduced systemic exposure, thereby increasing local efficacy and limiting toxicity. Moreover, since in vivo tumor nodules contain the tumor antigens, this tumor tissue may serve as an autologous vaccine to induce systemic immunity. This so-called 'bystander effect', where uninjected distant lesions exhibit a response, has been reported in select intralesional therapy trials. AREAS COVERED This review will give an overview of the working mechanisms, clinical evidence and side effects for available intralesional and topical therapies and summarize the most recent developments in this field. EXPERT OPINION The ideal treatment approach for locoregionally advanced melanoma should be multidisciplinary and tailored to the patient, taking into consideration patient-related, tumor-related factors (such as location, tumor burden, mutation status) and previous treatments received. It will likely not be a single therapy, but rather a combination of injectable treatments, regional perfusions and systemic therapies.
Collapse
Affiliation(s)
- Sarah Sloot
- University Medical Center Groningen , Groningen , Netherlands
| | | | | |
Collapse
|
8
|
Liu SX, Xia ZS, Zhong YQ. Gene therapy in pancreatic cancer. World J Gastroenterol 2014; 20:13343-68. [PMID: 25309069 PMCID: PMC4188890 DOI: 10.3748/wjg.v20.i37.13343] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Revised: 12/29/2013] [Accepted: 06/12/2014] [Indexed: 02/06/2023] Open
Abstract
Pancreatic cancer (PC) is a highly lethal disease and notoriously difficult to treat. Only a small proportion of PC patients are eligible for surgical resection, whilst conventional chemoradiotherapy only has a modest effect with substantial toxicity. Gene therapy has become a new widely investigated therapeutic approach for PC. This article reviews the basic rationale, gene delivery methods, therapeutic targets and developments of laboratory research and clinical trials in gene therapy of PC by searching the literature published in English using the PubMed database and analyzing clinical trials registered on the Gene Therapy Clinical Trials Worldwide website (http://www. wiley.co.uk/genmed/ clinical). Viral vectors are main gene delivery tools in gene therapy of cancer, and especially, oncolytic virus shows brighter prospect due to its tumor-targeting property. Efficient therapeutic targets for gene therapy include tumor suppressor gene p53, mutant oncogene K-ras, anti-angiogenesis gene VEGFR, suicide gene HSK-TK, cytosine deaminase and cytochrome p450, multiple cytokine genes and so on. Combining different targets or combination strategies with traditional chemoradiotherapy may be a more effective approach to improve the efficacy of cancer gene therapy. Cancer gene therapy is not yet applied in clinical practice, but basic and clinical studies have demonstrated its safety and clinical benefits. Gene therapy will be a new and promising field for the treatment of PC.
Collapse
|
9
|
Xu R, Guo LJ, Xin J, Li WM, Gao Y, Zheng YX, Guo YH, Lin YJ, Xie YH, Wu YQ, Xu RA. Luciferase Assay to Screen Tumour-specific Promoters in Lung Cancer. Asian Pac J Cancer Prev 2013; 14:6557-62. [DOI: 10.7314/apjcp.2013.14.11.6557] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
10
|
The development of gene-based vectors for immunization. Vaccines (Basel) 2013. [PMCID: PMC7151937 DOI: 10.1016/b978-1-4557-0090-5.00064-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
11
|
Mechanisms of action underlying the immunotherapeutic activity of Allovectin in advanced melanoma. Cancer Gene Ther 2012; 19:811-7. [PMID: 23037806 PMCID: PMC3499708 DOI: 10.1038/cgt.2012.69] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Allovectin (velimogene aliplasmid) is a cancer immunotherapeutic currently completing a pivotal phase 3 study for metastatic melanoma. Consisting of a bicistronic plasmid encoding both major histocompatibility complex (MHC) class I heavy and light chains (HLA-B7 and β2-microglobulin, respectively) formulated with a cationic lipid-based system, it is designed for direct intratumoral administration. Following injection into a single lesion, the product is intended to induce anti-tumor immune responses against both treated and distal lesions. Both the plasmid and lipid components of Allovectin contribute to the biological activity of the drug product, and its therapeutic activity is hypothesized to derive from multiple mechanisms of actions (MOAs). These include the induction of both cytotoxic T-cell and innate immune responses directed against allogeneic as well as tumor-derived targets, consequences of both an increased MHC class I expression on tumor cells and the induction of a localized immune/inflammatory response. In this paper, we review Allovectin's proposed MOAs, placing their contributions in the context of anti-tumor immunity and highlighting both preclinical and clinical supporting data.
Collapse
|
12
|
Zhang N, Yang Y, Cheng L, Zhang XM, Zhang S, Wang W, Liu SY, Wang SY, Wang RB, Xu WJ, Dai L, Yan N, Fan P, Dai LX, Tian HW, Liu L, Deng HX. Combination of Caspy2 and IP-10 gene therapy significantly improves therapeutic efficacy against murine malignant neoplasm growth and metastasis. Hum Gene Ther 2012; 23:837-46. [PMID: 22548488 DOI: 10.1089/hum.2011.136] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
It has been shown that Caspy2, a zebrafish active caspase, can efficiently suppress the growth of malignant tumor. The present study was designed to test whether combined gene therapy with IP-10, a potent antitumor chemokine, and Caspy2 would improve therapy efficacy. Recombinant plasmid expressing both Caspy2 and IP-10 genes was mixed with DOTAP-cholesterol nanoparticles. Immunocompetent mice bearing CT26 colon carcinoma, B16-F10 melanoma, and 4T1 breast carcinoma were treated with the complex. We found that the combined gene therapy more efficiently inhibited tumor growth, while efficiently prolonging the survival of tumor-bearing animals, compared with monotherapy. Moreover, a significant reduction in spontaneous lung metastasis could be observed in the 4T1 breast carcinoma model. Infiltration of CD8(+) T lymphocytes was also observed. In addition, apoptotic cells were widely detected by TUNEL assay and caspase-3 immunostaining in coadministered tumor tissues. The combination treatment also successfully inhibited angiogenesis and tumor cell proliferation as assessed by CD31 and Ki-67 immunostaining, respectively. Furthermore, depletion of CD8(+) T lymphocytes could significantly abrogate the antitumor activity, whereas the depletion of CD4(+) cells or natural killer cells showed partial abrogation. Rechallenged CT26 tumors were rejected in all of the surviving mice treated by combination therapy. Our results suggest that combined therapy with Caspy2 and IP-10 can significantly enhance antitumor activity by acting as an immune response initiator, apoptosis inducer, and angiogenesis inhibitor, which may be important for further applications in clinical cancer therapy.
Collapse
Affiliation(s)
- Na Zhang
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Goletz TJ, Mackall CL, Berzofsky JA, Helman LJ. Molecular alterations in pediatric sarcomas: potential targets for immunotherapy. Sarcoma 2011; 2:77-87. [PMID: 18521238 PMCID: PMC2395387 DOI: 10.1080/13577149878037] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022] Open
Abstract
Purpose/results/discussion. Recurrent chromosomal translocations are common features of many human malignancies. While such translocations often serve as diagnostic markers, molecular analysis of these breakpoint regions and the characterization of the affected genes is leading to a greater understanding of the causal role such translocations play in
malignant transformation. A common theme that is emerging from the study of tumor-associated translocations is the generation of chimeric genes that, when expressed, frequently retain many of the functional properties of the wild-type genes from which they originated. Sarcomas, in particular, harbor chimeric genes that are often derived from transcription factors, suggesting that the resulting chimeric transcription factors contribute to tumorigenesis. The tumor-specific expression of the fusion proteins make them likely candidates for tumor-associated antigens (TAA) and are thus of interest in the development of new therapies. The focus of this review will be on the translocation events associated with Ewing's sarcomas/PNETs (ES), alveolar rhabdomyosarcoma (ARMS), malignant melanoma of soft parts (MMSP) (clear cell sarcoma), desmoplastic small round cell tumor (DSRCT), synovial sarcoma (SS), and liposarcoma (LS), and the potential for targeting the resulting chimeric proteins in novel immunotherapies.
Collapse
Affiliation(s)
- T J Goletz
- Molecular Immunogenetics and Vaccine Research Section Metabolism Branch National Cancer Institute National Institutes of Health Bethesda MD 20892 USA
| | | | | | | |
Collapse
|
14
|
Abstract
Allovectin-7, a bicistronic plasmid encoding human leukocyte antigen-B7 and beta-2 microglobulin formulated with a cationic lipid system, is an immunotherapeutic agent designed to express allogeneic major histocompatibility complex class I antigen upon intralesional administration. A phase 2 dose-escalation study (VCL-1005-208) was conducted to evaluate the safety and efficacy of Allovectin-7 in patients with metastatic melanoma. Eligible patients had stage III or IV metastatic melanoma recurrent or unresponsive to prior therapy, an Eastern Cooperative Oncology Group performance status 0 or 1, and adequate organ function. Patients with brain or visceral (except lung) metastases, abnormal lactate dehydrogenase, or any lesion greater than 100 cm were excluded. Patients received six weekly intralesional injections followed by 3 weeks of observation and evaluation. Overall response was assessed using Response Evaluation Criteria in Solid Tumors guidelines. Patients with stable or responding disease were eligible to receive additional cycles of Allovectin-7. All 133 patients were evaluated for safety and 127 patients (2 mg, high dose) were evaluated for efficacy. Fifteen patients (11.8%, 95% confidence interval: 6.2-17.4) achieved an objective response with median duration of response of 13.8 months (95% confidence interval: 8.5, not estimable). A histological examination of tissue from two responding patients who had their lesions resected has shown no evidence of melanoma. Median time-to-progression in this study was 1.6 months. In conclusion, these results indicate that high-dose Allovectin-7 seems to be an active, well-tolerated treatment for selected stage III/IV metastatic melanoma patients with injectable cutaneous, subcutaneous, or nodal lesions.
Collapse
|
15
|
Abstract
IMPORTANCE OF THE FIELD Immunotherapy for cancer has been investigated for several decades, achieving limited success. The development of effective new immunotherapeutic agents has reignited interest in the filed. Intralesional injection of plasmids in order to transfect genes capable of stimulating or augmenting immune recognition and destruction of tumors is a relatively new approach. AREAS COVERED IN THIS REVIEW Our objective is to discuss the role velimogene aliplasmid (Allovectin-7, Vical Incorporated), a plasmid-lipid complex containing the DNA sequences encoding HLA-B7 and beta2 microglobulin, as an immunotherapeutic agent. WHAT THE READER WILL GAIN Intralesional velimogene aliplasmid induces anti-tumor responses in a proportion of melanoma patients with locoregional and limited distant metastases. Preclinical data and the results of Phase I, II and III clinical trials with this drug are reviewed. The limited data in other malignancies is also reviewed. Velimogene aliplasmid in humans appears safe, with minimal drug-related adverse events. TAKE HOME MESSAGE Velimogene aliplasmid has activity in melanoma with local and limited distant disease associated with an excellent safety profile. The activity of this approach is also being investigated in other malignancies.
Collapse
Affiliation(s)
- Heloisa P Soares
- Department of Medicine, Mount Sinai Medical Center, 4300 Alton Road, Miami Beach, FL 33140, USA
| | | |
Collapse
|
16
|
Luo S, Chen P, Luo ZC, Zhang P, Sun P, Shi W, Li ZY, Zhang XL, Wang LQ, Chen X, Wei YQ, Wen YJ. Combination of vesicular stomatitis virus matrix protein gene therapy with low-dose cisplatin improves therapeutic efficacy against murine melonoma. Cancer Sci 2010; 101:1219-25. [PMID: 20331624 PMCID: PMC11159948 DOI: 10.1111/j.1349-7006.2010.01507.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
Vesicular stomatitis virus (VSV) matrix protein (MP) can directly induce apoptosis via the mitochondrial pathway due to the inhibition of host gene expression. Our previous studies have demonstrated that MP gene therapy efficiently suppressed the growth of malignant tumor in vitro and in vivo. The present study was designed to determine the possibility that the combination of MP gene therapy with low-dose cisplatin would improve therapeutic efficacy against murine melanoma. Immunocompetent C57BL/6 mice bearing B16-F10 melanoma were established. Mice were treated once every 5 days with i.v. administration of 10 microg pVAX-MP/30 microg liposome complex per mouse for 16 days and i.p. delivery of cisplatin at 4 mg/kg/mouse on days 6 and 12 after the initiation of MP treatment. We found that MP + cisplatin treatment resulted in significant inhibition of tumor growth and improved the survival time of melanoma-bearing mice. MP successfully inhibited angiogenesis as assessed by CD31. Histological examination revealed that the combination therapy led to significant increased induction of apoptosis, tumor necrosis, and elevated CD8(+) lymphocyte infiltration. Furthermore, the induction efficacy of the CTL response was dramatically enhanced by the combination therapy. Our findings may prove useful in further explorations of the application of these combinational approaches to the treatment of malignant melanoma.
Collapse
Affiliation(s)
- Shan Luo
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Thomas SM, Grandis JR. The current state of head and neck cancer gene therapy. Hum Gene Ther 2010; 20:1565-75. [PMID: 19747066 DOI: 10.1089/hum.2009.163] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The incidence of head and neck cancer continues to increase worldwide, with tobacco exposure and human papillomavirus type 16 infections being the major etiological factors. Current therapeutic options are ineffective in approximately half of the individuals afflicted with this malignancy. Developments in the identification of molecules that sustain head and neck squamous cell carcinoma (HNSCC) growth and survival have made molecular targeting by gene therapy approaches a feasible therapeutic strategy. Although gene therapy was originally designed to correct single gene defects, it has now evolved to encompass all forms of therapeutic interventions involving engineered cells and nucleic acids that modify the overall pattern of gene expression within target tissues. Several preclinical studies and clinical trials have tested the efficacy of targeting specific molecules in patients with HNSCC, using genetic therapy approaches. This review discusses promising preclinical and clinical approaches and new directions for HNSCC gene therapy.
Collapse
Affiliation(s)
- Sufi Mary Thomas
- Department of Otolaryngology, University of Pittsburgh and University of Pittsburgh Cancer Institute, Pittsburgh, PA 15213, USA
| | | |
Collapse
|
18
|
Bozeman EN, Shashidharamurthy R, Paulos SA, Palaniappan R, D’Souza M, Selvaraj P. Cancer vaccine development: designing tumor cells for greater immunogenicity. FRONT BIOSCI-LANDMRK 2010; 15:309-20. [PMID: 20036822 PMCID: PMC4513666 DOI: 10.2741/3622] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Cancer vaccine development is one of the most hopeful and exhilarating areas in cancer research. For this reason, there has been a growing interest in the development and application of novel immunotherapies for the treatment of cancer with the focus being on stimulating the immune system to target tumor cells specifically while leaving normal cells unharmed. From such research has emerged a host of promising immunotherapies such as dendritic cell-based vaccines, cytokine therapies and gene transfer technology. These therapies seek to counteract the poor immunogenicity of tumors by augmenting the host's immune system with a variety of immunostimulatory proteins such as cytokines and costimulatory molecules. While such therapies have proven effective in the induction of anti-tumor immunity in animal models, they are less than optimal and pose a high risk of clinical infeasibility. Herein, we further discuss these immunotherapies as well as a feasible and efficient alternative that, in pre-clinical animal models, allows for the expression of specific immunostimulatory molecules on the surface of tumor cells by a novel protein transfer technology.
Collapse
Affiliation(s)
- Erica N. Bozeman
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA 30322, USA
| | | | - Simon A. Paulos
- Department of Pharmaceutical Sciences, Mercer University College of Pharmacy and Health Sciences, 3001 Mercer University Dr., Atlanta, GA 30341, USA
| | - Ravi Palaniappan
- Department of Pharmaceutical Sciences, Mercer University College of Pharmacy and Health Sciences, 3001 Mercer University Dr., Atlanta, GA 30341, USA
| | - Martin D’Souza
- Department of Pharmaceutical Sciences, Mercer University College of Pharmacy and Health Sciences, 3001 Mercer University Dr., Atlanta, GA 30341, USA
| | - Periasamy Selvaraj
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA 30322, USA
| |
Collapse
|
19
|
Van Gool S, Maes W, Ardon H, Verschuere T, Van Cauter S, De Vleeschouwer S. Dendritic cell therapy of high-grade gliomas. Brain Pathol 2009; 19:694-712. [PMID: 19744041 DOI: 10.1111/j.1750-3639.2009.00316.x] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The prognosis of patients with malignant glioma is poor in spite of multimodal treatment approaches consisting of neurosurgery, radiochemotherapy and maintenance chemotherapy. Among innovative treatment strategies like targeted therapy, antiangiogenesis and gene therapy approaches, immunotherapy emerges as a meaningful and feasible treatment approach for inducing long-term survival in at least a subpopulation of these patients. Setting up immunotherapy for an inherent immunosuppressive tumor located in an immune-privileged environment requires integration of a lot of scientific input and knowledge of both tumor immunology and neuro-oncology. The field of immunotherapy is moving into the direction of active specific immunotherapy using autologous dendritic cells (DCs) as vehicle for immunization. In the translational research program of the authors, the whole cascade from bench to bed to bench of active specific immunotherapy for malignant glioma is covered, including proof of principle experiments to demonstrate immunogenicity of patient-derived mature DCs loaded with autologous tumor lysate, preclinical in vivo experiments in a murine orthotopic glioma model, early phase I/II clinical trials for relapsing patients, a phase II trial for patients with newly diagnosed glioblastoma (GBM) for whom immunotherapy is integrated in the current multimodal treatment, and laboratory analyses of patient samples. The strategies and results of this program are discussed in the light of the internationally available scientific literature in this fast-moving field of basic science and translational clinical research.
Collapse
Affiliation(s)
- Stefaan Van Gool
- Laboratory of Experimental Immunology, and Department of Child & Woman, Catholic University of Leuven, Leuven, Belgium.
| | | | | | | | | | | |
Collapse
|
20
|
Induction of Antitumor Response by In Vivo Allogeneic Major Histocompatibility Complex Gene Transfer Using Electroporation. J Surg Res 2009; 154:60-7. [DOI: 10.1016/j.jss.2008.05.035] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2008] [Revised: 05/12/2008] [Accepted: 05/27/2008] [Indexed: 11/21/2022]
|
21
|
Liu L, Deng H, Wang Y, Chen P, Yang Y, Yang H, Chen X, Chen L, Zhu W, Liang S, Yang J, Qian Z, Li J, Wen Y, Kan B, Mao Y, Zhao X, Wei Y. A novel strategy for tumour therapy combining cell apoptosis and active immunity induced by caspy2, a zebrafish caspase. J Cell Mol Med 2009; 13:2271-2281. [PMID: 19220579 DOI: 10.1111/j.1582-4934.2009.00693.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Caspy2, a zebrafish protease, is an active caspase for inducing apoptosis in mammalian cells. To investigate whether caspy2-mediated apoptosis could be used in cancer therapy, its cDNA was amplified and cloned into eukaryotic expression vector pcDNA3.1(+). The recombinant plasmid was mixed with cationic liposome and introduced into various tumour cell lines in vitro. Our data showed that caspy2 induced remarkable apoptosis of cancer cells in vitro. Treatment of mice-bearing CT26 colon carcinoma or MethA fibrosarcoma with intratumoral injection of liposome-caspy2 plasmid complex resulted in substantial killing of neoplastic cells and long-term survivors. Apoptotic cells were widely distributed in caspy2-treated tumour tissue. Infiltration of CD8(+) T lymphocyte was also observed apparently in the tumour tissue after the treatment with caspy2. Tumour-specific major histocompatibility complex (MHC) class I-dependent CD8(+) cytotoxic T lymphocyte activity was found by means of (51)Cr release assay. In MethA model, the mice acquired a long-time protective immunity against the parental tumour cell re-challenge. These results indicated that caspy2 can act as both apoptosis inducer and immune response initiator, which may account for its extraordinary antitumour effect. Furthermore, in vivo depletion of CD8(+) T lymphocytes could completely abrogate the antitumour immune activity, whereas the depletion of CD4(+) cells showed partial abrogation. In this study, we developed a novel anticancer strategy that combines both induction of apoptosis and immune response in mice-bearing tumours with a single caspy2 gene. This approach may provide an important way for treatment of cancer.
Collapse
Affiliation(s)
- Lei Liu
- State Key Laboratory of Biotherapy, West China Hospital and College of Life Science, Sichuan University, Chengdu, Sichuan, The People's Republic of China
| | - Hongxin Deng
- State Key Laboratory of Biotherapy, West China Hospital and College of Life Science, Sichuan University, Chengdu, Sichuan, The People's Republic of China
| | - Yongsheng Wang
- State Key Laboratory of Biotherapy, West China Hospital and College of Life Science, Sichuan University, Chengdu, Sichuan, The People's Republic of China
| | - Ping Chen
- State Key Laboratory of Biotherapy, West China Hospital and College of Life Science, Sichuan University, Chengdu, Sichuan, The People's Republic of China
| | - Yang Yang
- State Key Laboratory of Biotherapy, West China Hospital and College of Life Science, Sichuan University, Chengdu, Sichuan, The People's Republic of China
| | - Hanshuo Yang
- State Key Laboratory of Biotherapy, West China Hospital and College of Life Science, Sichuan University, Chengdu, Sichuan, The People's Republic of China
| | - Xiancheng Chen
- State Key Laboratory of Biotherapy, West China Hospital and College of Life Science, Sichuan University, Chengdu, Sichuan, The People's Republic of China
| | - Lijuan Chen
- State Key Laboratory of Biotherapy, West China Hospital and College of Life Science, Sichuan University, Chengdu, Sichuan, The People's Republic of China
| | - Wen Zhu
- State Key Laboratory of Biotherapy, West China Hospital and College of Life Science, Sichuan University, Chengdu, Sichuan, The People's Republic of China
| | - Shufang Liang
- State Key Laboratory of Biotherapy, West China Hospital and College of Life Science, Sichuan University, Chengdu, Sichuan, The People's Republic of China
| | - Jinliang Yang
- State Key Laboratory of Biotherapy, West China Hospital and College of Life Science, Sichuan University, Chengdu, Sichuan, The People's Republic of China
| | - Zhiyong Qian
- State Key Laboratory of Biotherapy, West China Hospital and College of Life Science, Sichuan University, Chengdu, Sichuan, The People's Republic of China
| | - Jiong Li
- State Key Laboratory of Biotherapy, West China Hospital and College of Life Science, Sichuan University, Chengdu, Sichuan, The People's Republic of China
| | - Yanjun Wen
- State Key Laboratory of Biotherapy, West China Hospital and College of Life Science, Sichuan University, Chengdu, Sichuan, The People's Republic of China
| | - Bin Kan
- State Key Laboratory of Biotherapy, West China Hospital and College of Life Science, Sichuan University, Chengdu, Sichuan, The People's Republic of China
| | - Yongqiu Mao
- State Key Laboratory of Biotherapy, West China Hospital and College of Life Science, Sichuan University, Chengdu, Sichuan, The People's Republic of China
| | - Xia Zhao
- State Key Laboratory of Biotherapy, West China Hospital and College of Life Science, Sichuan University, Chengdu, Sichuan, The People's Republic of China
| | - Yuquan Wei
- State Key Laboratory of Biotherapy, West China Hospital and College of Life Science, Sichuan University, Chengdu, Sichuan, The People's Republic of China
| |
Collapse
|
22
|
Navarro J, Risco R, Toschi M, Schattman G. Gene therapy and intracytoplasmatic sperm injection (ICSI) - a review. Placenta 2008; 29 Suppl B:193-9. [PMID: 18790332 DOI: 10.1016/j.placenta.2008.08.014] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2008] [Revised: 08/14/2008] [Accepted: 08/14/2008] [Indexed: 01/31/2023]
Abstract
Human gene therapy (HGT), the treatment or prevention of disease by gene transfer is, regarded by many, as a potential revolution in medicine, because gene therapies target the causes of disease, whereas most current drugs treat the symptoms. Micro-assisted fertilization in the form of intracytoplasmatic sperm injection (ICSI) has truly revolutionized the treatment options for couples with impaired semen quality, and those with both obstructive and non-obstructive azoospermia. ICSI involves the injection of a single sperm directly into the cytoplasm of a mature egg (oocyte) using a glass needle (pipette). Fertilization with this technique occurs in 50%-80% of injected oocytes, but may damage a small percentage of them. With gene therapy, there are new and varied strategies for gene transfer and genome sequence manipulation with improved methodologies that use the technique of microinjection such as the intracytoplasmatic sperm injection-mediated transgenesis (ICSI-Tr), active transgenesis or the pronuclear microinjection technique. This review will look at these methods as well as their potential applications and limitations.
Collapse
Affiliation(s)
- J Navarro
- Instituto de Biotecnología Aplicada (BIONAC), Seville, Spain.
| | | | | | | |
Collapse
|
23
|
Bedikian AY, Del Vecchio M. Allovectin-7 therapy in metastatic melanoma. Expert Opin Biol Ther 2008; 8:839-44. [PMID: 18476795 DOI: 10.1517/14712598.8.6.839] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND Patients with metastatic melanoma are immunosuppressed by the growing tumor. Allovectin-7 therapy is a form of active immunotherapy that aims at immunization of the host with substances designed to elicit an immune reaction that will eliminate or slow down the growth and spread of the cancer. OBJECTIVE to describe the rationale for immunotherapy with Allovectin-7 and assess its safety profile and efficacy based on the results of completed melanoma clinical trials. METHODS we reviewed both the published medical literature and the results of trials pending publication. RESULTS/CONCLUSION Allovectin-7 is a safe and active immunotherapeutic agent. It induces local and systemic durable responses in patients with metastatic melanoma.
Collapse
Affiliation(s)
- Agop Y Bedikian
- The University of Texas MD Anderson Cancer Center, Department of Melanoma Medical Oncology, 1515 Holcombe Blvd, Box 430, Houston, TX 77030, USA.
| | | |
Collapse
|
24
|
Son KK. Chemical Toxicants Activate Murine Ovarian Ascitic Tumor Cells for In Situ Lipofection. Drug Deliv 2008. [DOI: 10.1080/107175499266986] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022] Open
|
25
|
Sternberg B. Morphology of Cationic Liposome/DNA Complexes in Relation to Their Chemical Composition. J Liposome Res 2008. [DOI: 10.3109/08982109609031134] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
26
|
|
27
|
|
28
|
|
29
|
Hui KM, Sabapathy TK, Oei AA, Singhal A, Huang L. Induction of alloreactive cytotoxic T lymphocytes by intra-splenic immunization with allogeneic class I Major Histocompatibility Complex DNA and DC-chol cationic liposomes. J Liposome Res 2008; 4:1075-1090. [DOI: 10.3109/08982109409018623] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Affiliation(s)
- Kam M. Hui
- Molecular Immunology Laboratory, Institute of Molecular and Cell Biology, National University of Singapore, Kent Ridge, Singapore, 0511
| | - Tr. Kanaga Sabapathy
- Molecular Immunology Laboratory, Institute of Molecular and Cell Biology, National University of Singapore, Kent Ridge, Singapore, 0511
| | - Audrey A. Oei
- Molecular Immunology Laboratory, Institute of Molecular and Cell Biology, National University of Singapore, Kent Ridge, Singapore, 0511
| | - Arun Singhal
- Molecular Immunology Laboratory, Institute of Molecular and Cell Biology, National University of Singapore, Kent Ridge, Singapore, 0511
| | - Leaf Huang
- department of Pharmacology, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| |
Collapse
|
30
|
Litzinger DC. Limitations of Cationic Liposomes for Antisense Oligonucleotide Delivery in Vivo. J Liposome Res 2008. [DOI: 10.3109/08982109709035481] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
31
|
Zhao J, Wen Y, Li Q, Wang Y, Wu H, Xu J, Chen X, Wu Y, Fan L, Yang H, Liu T, Ding Z, Du X, Diao P, Li J, Wu H, Kan B, Lei S, Deng H, Mao Y, Zhao X, Wei Y. A promising cancer gene therapy agent based on the matrix protein of vesicular stomatitis virus. FASEB J 2008; 22:4272-80. [PMID: 18716030 DOI: 10.1096/fj.08-110049] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Ju‐mei Zhao
- State Key Laboratory of BiotherapyWest China Hospital, and Sichuan UniversityChengduSichuanChina
- Pharmacological Department of Medical CollegeYan'an UniversityYan'anshanxiChina
| | - Yan‐jun Wen
- State Key Laboratory of BiotherapyWest China Hospital, and Sichuan UniversityChengduSichuanChina
| | - Qiu Li
- State Key Laboratory of BiotherapyWest China Hospital, and Sichuan UniversityChengduSichuanChina
| | - Yong‐sheng Wang
- State Key Laboratory of BiotherapyWest China Hospital, and Sichuan UniversityChengduSichuanChina
| | - Hong‐bo Wu
- State Key Laboratory of BiotherapyWest China Hospital, and Sichuan UniversityChengduSichuanChina
| | - Jian‐rong Xu
- College of Life ScienceSichuan UniversityChengduSichuanChina
| | - Xian‐cheng Chen
- State Key Laboratory of BiotherapyWest China Hospital, and Sichuan UniversityChengduSichuanChina
| | - Yang Wu
- State Key Laboratory of BiotherapyWest China Hospital, and Sichuan UniversityChengduSichuanChina
| | - Ling‐yu Fan
- State Key Laboratory of BiotherapyWest China Hospital, and Sichuan UniversityChengduSichuanChina
| | - Han‐shuo Yang
- State Key Laboratory of BiotherapyWest China Hospital, and Sichuan UniversityChengduSichuanChina
| | - Tao Liu
- State Key Laboratory of BiotherapyWest China Hospital, and Sichuan UniversityChengduSichuanChina
| | - Zhen‐yu Ding
- State Key Laboratory of BiotherapyWest China Hospital, and Sichuan UniversityChengduSichuanChina
| | - Xiao‐bo Du
- State Key Laboratory of BiotherapyWest China Hospital, and Sichuan UniversityChengduSichuanChina
| | - Peng Diao
- State Key Laboratory of BiotherapyWest China Hospital, and Sichuan UniversityChengduSichuanChina
| | - Jiong Li
- State Key Laboratory of BiotherapyWest China Hospital, and Sichuan UniversityChengduSichuanChina
| | - Hong‐bing Wu
- State Key Laboratory of BiotherapyWest China Hospital, and Sichuan UniversityChengduSichuanChina
| | - Bing Kan
- State Key Laboratory of BiotherapyWest China Hospital, and Sichuan UniversityChengduSichuanChina
| | - Song Lei
- State Key Laboratory of BiotherapyWest China Hospital, and Sichuan UniversityChengduSichuanChina
| | - Hong‐xin Deng
- State Key Laboratory of BiotherapyWest China Hospital, and Sichuan UniversityChengduSichuanChina
| | - Yong‐qiu Mao
- State Key Laboratory of BiotherapyWest China Hospital, and Sichuan UniversityChengduSichuanChina
| | - Xia Zhao
- State Key Laboratory of BiotherapyWest China Hospital, and Sichuan UniversityChengduSichuanChina
| | - Yu‐quan Wei
- State Key Laboratory of BiotherapyWest China Hospital, and Sichuan UniversityChengduSichuanChina
| |
Collapse
|
32
|
Henshaw JW, Yuan F. Field distribution and DNA transport in solid tumors during electric field-mediated gene delivery. J Pharm Sci 2008; 97:691-711. [PMID: 17624918 DOI: 10.1002/jps.21000] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Gene therapy has a great potential in cancer treatment. However, the efficacy of cancer gene therapy is currently limited by the lack of a safe and efficient means to deliver therapeutic genes into the nucleus of tumor cells. One method under investigation for improving local gene delivery is based on the use of pulsed electric field. Despite repeated demonstration of its effectiveness in vivo, the underlying mechanisms behind electric field-mediated gene delivery remain largely unknown. Without a thorough understanding of these mechanisms, it will be difficult to further advance the gene delivery. In this review, the electric field-mediated gene delivery in solid tumors will be examined by following individual transport processes that must occur in vivo for a successful gene transfer. The topics of examination include: (i) major barriers for gene delivery in the body, (ii) distribution of electric fields at both cell and tissue levels during the application of external fields, and (iii) electric field-induced transport of genes across each of the barriers. Through this approach, the review summarizes what is known about the mechanisms behind electric field-mediated gene delivery and what require further investigations in future studies.
Collapse
Affiliation(s)
- Joshua W Henshaw
- Department of Biomedical Engineering, Duke University, 136 Hudson Hall, Durham, North Carolina 27708, USA
| | | |
Collapse
|
33
|
Kobayashi A, Hara H, Ohashi M, Nishimoto T, Yoshida K, Ohkohchi N, Yoshida T, Aoki K. Allogeneic MHC gene transfer enhances an effective antitumor immunity in the early period of autologous hematopoietic stem cell transplantation. Clin Cancer Res 2008; 13:7469-79. [PMID: 18094431 DOI: 10.1158/1078-0432.ccr-07-1163] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE In autologous hematopoietic stem cell transplantation (HSCT), lymphopenia-induced homeostatic proliferation of T cells is driven by the recognition of self-antigens, and there is an opportunity to skew the T-cell repertoire during the T-cell recovery by engaging tumor-associated antigens, leading to a break of tolerance against tumors. However, the homeostatic proliferation-driven antitumor responses seem to decline rapidly in association with tumor growth. We hypothesized that a tumor-specific immune response induced by an immune gene therapy could enhance and sustain homeostatic proliferation-induced antitumor immunity. EXPERIMENTAL DESIGN The antitumor effect of allogeneic MHC (alloMHC) gene transfer was examined at the early phase of the immune reconstitution after syngeneic HSCT. RESULTS Syngeneic HSCT showed significant tumor growth inhibition of syngeneic colon cancer cells within a period of 30 days; however, the tumor then resumed rapid growth and the survival of the mice was not prolonged. In contrast, when the alloMHC plasmid was intratumorally injected at the early phase after syngeneic HSCT, the established tumors were markedly regressed and the survival of recipient mice was prolonged without significant toxicities, whereas no survival advantage was recognized in recipient mice injected with a control plasmid. This tumor suppression was evident even in the other tumors that were not injected with the alloMHC plasmid. The antitumor response was characterized by the development of tumor-specific T cell- and natural killer cell-mediated cytotoxicities. CONCLUSION The results suggest the efficacy and safety of integrating intratumoral alloMHC gene transfer with an autologous HSCT for the treatment of solid cancers.
Collapse
Affiliation(s)
- Akihiko Kobayashi
- Authors' Affiliations: Section for Studies on Host-Immune Response and Genetics Division, National Cancer Center Research Institute, Tokyo, Japan
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Sokolova V, Epple M. Inorganic nanoparticles as carriers of nucleic acids into cells. Angew Chem Int Ed Engl 2008; 47:1382-95. [PMID: 18098258 DOI: 10.1002/anie.200703039] [Citation(s) in RCA: 392] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The transfer of nucleic acids (DNA or RNA) into living cells, that is, transfection, is a major technique in current biochemistry and molecular biology. This process permits the selective introduction of genetic material for protein synthesis as well as the selective inhibition of protein synthesis (antisense or gene silencing). As nucleic acids alone are not able to penetrate the cell wall, efficient carriers are needed. Besides viral, polymeric, and liposomal agents, inorganic nanoparticles are especially suitable for this purpose because they can be prepared and surface-functionalized in many different ways. Herein, the current state of the art is discussed from a chemical viewpoint. Advantages and disadvantages of the available methods are compared.
Collapse
Affiliation(s)
- Viktoriya Sokolova
- Institut für Anorganische Chemie, Universität Duisburg-Essen, Universitätsstrasse 5-7, 45117 Essen, Germany
| | | |
Collapse
|
35
|
Scott-Ram NR. Review Biologicals & Immunologicals: Gene therapy: review of recent patenting activity. Expert Opin Ther Pat 2008. [DOI: 10.1517/13543776.5.3.219] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
36
|
Sokolova V, Epple M. Anorganische Nanopartikel zum Transport von Nucleinsäuren in Zellen. Angew Chem Int Ed Engl 2008. [DOI: 10.1002/ange.200703039] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
37
|
Merrick A, Diaz RM, O'Donnell D, Selby P, Vile R, Melcher A. Autologous versus allogeneic peptide-pulsed dendritic cells for anti-tumour vaccination: expression of allogeneic MHC supports activation of antigen specific T cells, but impairs early naïve cytotoxic priming and anti-tumour therapy. Cancer Immunol Immunother 2007; 57:897-906. [PMID: 18057935 DOI: 10.1007/s00262-007-0426-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2007] [Accepted: 11/10/2007] [Indexed: 01/01/2023]
Abstract
BACKGROUND Dendritic cells (DC) pulsed with MHC class I-restricted tumour associated antigen (TAA) peptides have been widely tested in pre-clinical models and early clinical studies for their ability to prime cytotoxic T cell (CTL) responses. The effect of co-expression of allogeneic MHC antigens on DC immunogenicity has not been addressed, and has implications for the feasibility of clinical applications. OBJECTIVE This study compared DC from autologous H-2(b) or semi-allogeneic F1 H-2(bxk) mice pulsed with the H-2(b)-restricted model ovalbumin (OVA) peptide SIINFEKL, and compared in vitro and in vivo their ability to (i) activate specific OT1 cells, (ii) prime naïve CTL, and (iii) protect against B16.OVA challenge. Peptide-pulsed autologous and allogeneic DC were also tested in naïve human CTL priming assays. RESULTS Semi-allogeneic DC expressed higher levels of co-stimulatory molecules. On pulsing with SIINFEKL they triggered greater proliferation of OT1 cells in vitro and in vivo, but were less effective at naïve CTL priming and tumour protection. Autologous human DC were similarly more potent at naïve CTL priming against the melanoma-associated TAA MART-1 in vitro. CONCLUSION The expression of allogeneic MHC antigens on peptide-pulsed DC impairs naïve CTL priming and anti-tumour effects, despite effective TAA presentation both in vitro and in vivo.
Collapse
Affiliation(s)
- Alison Merrick
- Cancer Research UK Clinical Centre, St James's University Hospital, Beckett St, Leeds LS9 7TF, UK.
| | | | | | | | | | | |
Collapse
|
38
|
Recillas-Targa F. Multiple strategies for gene transfer, expression, knockdown, and chromatin influence in mammalian cell lines and transgenic animals. Mol Biotechnol 2007; 34:337-54. [PMID: 17284781 DOI: 10.1385/mb:34:3:337] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/1999] [Revised: 11/30/1999] [Accepted: 11/30/1999] [Indexed: 12/31/2022]
Abstract
Manipulation of the eukaryotic genome has contributed to the progress in our knowledge of multicellular organisms but has also ameliorated our experimental strategies. Biological questions can now be addressed with more efficiency and reproducibility. There are new and varied strategies for gene transfer and sequence manipulation with improved methodologies that facilitate the acquisition of results. Cellular systems and transgenic animals have demonstrated their invaluable benefits. In this review, I present an overview of the methods of gene transfer with particular attention to cultured cell lines and large-scale sequence vectors, like artificial chromosomes, with the possibility of their manipulation based on homologous recombination strategies. Alternative strategies of gene transfer, including retroviral vectors, are also described and the applications of such methods are discussed. Finally, several comments are made about the influence of chromatin structure on gene expression. Recent experimental data have shown that for convenient stable transgene expression, the influence of chromatin structure should be seriously taken into account. Novel chromatin regulatory and structural elements are proposed as an alternative for proper and sustained gene expression. These chromatin elements are facing a new era in transgenesis and we are probably beginning a new generation of gene and cancer therapy vectors.
Collapse
Affiliation(s)
- Félix Recillas-Targa
- Instituto de Fisiología Celular, Departamento de Genética Molecular, Universidad Nacional Autónoma de México Apartado Postal 70-242, México D.F. 04510.
| |
Collapse
|
39
|
Ohashi M, Kobayashi A, Hara H, Miura Y, Yoshida K, Kushida M, Ikarashi Y, Mandai M, Kitajima M, Yoshida T, Aoki K. Allogeneic MHC gene transfer enhances antitumor activity of allogeneic hematopoietic stem cell transplantation without exacerbating graft-versus-host disease. Clin Cancer Res 2006; 12:2208-15. [PMID: 16609036 DOI: 10.1158/1078-0432.ccr-05-2657] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Enhancement of the specific antitumor activity of allogeneic hematopoietic stem cell transplantation (alloHSCT) against solid cancers is a major issue in the clinical oncology. In this study, we examined whether intratumoral allogeneic MHC (alloMHC) gene transfer can enhance the recognition of tumor-associated antigens by donor T cells and augment the antitumor activity of alloHSCT. In minor histocompatibility antigen-mismatched alloHSCT (DBA/2-->BALB/c: H-2(d)) recipients, alloMHC gene (H-2K(b)) was transduced directly into a s.c. tumor of CT26 colon cancer cells. Because CT26 cells have an aggressive tumorigenicity in syngeneic BALB/c mice, an H-2K(b) gene transfer provides only a limited antitumor effect after syngeneic (BALB/c-->BALB/c) HSCT. By contrast, the H-2K(b) gene transfer caused significant tumor suppression in the alloHSCT recipients, and this suppression was evident not only in the gene-transduced tumors but also in simultaneously inoculated distant tumors without gene transduction. In vitro cytotoxicity assay showed specific tumor cell lysis by donor T cells responding to the H-2K(b) gene transfer. Graft-versus-host disease was not exacerbated serologically or clinically in the treated mice, demonstrating that alloMHC gene transfer enhances the antitumor effects of alloHSCT without exacerbating graft-versus-host disease. This combination strategy has important implications for the development of therapies for human solid cancers.
Collapse
Affiliation(s)
- Masaki Ohashi
- Genetics Division, National Cancer Center Research Institute, Department of Surgery, Keio University School of Medicine Tokyo, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Kobayashi N, Nishikawa M, Takakura Y. The hydrodynamics-based procedure for controlling the pharmacokinetics of gene medicines at whole body, organ and cellular levels. Adv Drug Deliv Rev 2005; 57:713-31. [PMID: 15757757 DOI: 10.1016/j.addr.2004.12.006] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2004] [Accepted: 12/18/2004] [Indexed: 10/25/2022]
Abstract
Hydrodynamics-based gene delivery, involving a large-volume and high-speed intravenous injection of naked plasmid DNA (pDNA), gives a significantly high level of transgene expression in vivo. This has attracted a lot of attention and has been used very frequently as an efficient, simple and convenient transfection method for laboratory animals. Until recently, however, little information has been published on the pharmacokinetics of the injected DNA molecules and of the detailed mechanisms underlying the efficient gene transfer. We and other groups have very recently demonstrated that the mechanism for the hydrodynamics-based gene transfer would involve, in part, the direct cytosolic delivery of pDNA through the cell membrane due to transiently enhanced permeability. Along with the findings in our series of studies, this article reviews the cumulative reports and other intriguing information on the controlled pharmacokinetics of naked pDNA in the hydrodynamics-based gene delivery. In addition, we describe various applications reported so far, as well as the current attempts and proposals to develop novel gene medicines for future gene therapy using the concept of the hydrodynamics-based procedure. Furthermore, the issues associated with the clinical feasibility of its seemingly invasive nature, which is probably the most common concern about this hydrodynamics-based procedure, are discussed along with its future prospects and challenges.
Collapse
Affiliation(s)
- Naoki Kobayashi
- Department of Biopharmaceutics and Drug Metabolism, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | | | | |
Collapse
|
41
|
Gückel B, Stumm S, Rentzsch C, Marmé A, Mannhardt G, Wallwiener D. A CD80-transfected human breast cancer cell variant induces HER-2/neu-specific T cells in HLA-A*02-matched situations in vitro as well as in vivo. Cancer Immunol Immunother 2005; 54:129-40. [PMID: 15365776 PMCID: PMC11034349 DOI: 10.1007/s00262-004-0583-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2004] [Accepted: 06/14/2004] [Indexed: 10/26/2022]
Abstract
Adjuvant treatment is still only working in a small percentage of breast cancer patients. Therefore, new strategies need to be developed. Immunotherapies are a very promising approach because they could successfully attack tumor cells in the stage of dormancy. To assess the feasibility of using an allogeneic approach for vaccination of breast cancer patients, we selected a CD80-transfected breast cancer cell line based on its immunogenic properties. Using CD80+ KS breast cancer cells and human leukocyte antigen (HLA)-A*02-matched peripheral blood mononuclear cells (PBMCs) of breast cancer patients in allogeneic mixed lymphocyte-tumor cell cultures (MLTCs), it was possible to isolate HLA-A*02-restricted cytotoxic T cells (CTLs). Furthermore, a genetically modified KS variant expressing influenza A matrix protein serving as a surrogate tumor-associated antigen (TAA) was able to stimulate flu peptide-specific T cells alongside the induction of alloresponses in MLTCs. KS breast cancer cells were demonstrated to express already known TAAs such as CEA, MUC-1, MAGE-1, MAGE-2, and MAGE-3. To further improve antigenicity, HER-2/neu was added to this panel as a marker antigen known to elicit HLA-A*02-restricted CTLs in patients with breast cancer. Thus, the antigen-processing and antigen-presentation capacity of KS cells was further demonstrated by the stimulation of HER-2/neu-specific CD8+ T cells in PBMCs of breast cancer patients in vitro. These results gave a good rationale for a phase I/II trial, where the CD80+ HER-2/neu-overexpressing KS variant is actually used as a cellular vaccine in patients with metastatic breast cancer. As a proof of principle, we present data from two patients where a significant increase of interferon-gamma (IFN-gamma) release was detected when postvaccination PBMCs were stimulated by allogeneic vaccine cells as well as by HLA-A*02-restricted HER-2/neu epitopes. In whole cell vaccine trials, monitoring is particularly challenging because of strong alloresponses and limited knowledge of TAAs. In this study, a panel of HER-2/neu epitopes, together with the quantitative real time (qRT)-PCR method to analyze vaccine-induced cytokines secreted by T cells, proved to be highly sensitive and feasible to perform an "immunological staging" following vaccination.
Collapse
Affiliation(s)
- Brigitte Gückel
- Department of Gynecology and Obstetrics, University of Tübingen, Calwerstrasse 7, 72076, Tübingen, Germany.
| | | | | | | | | | | |
Collapse
|
42
|
Nishikawa M, Takakura Y, Hashida M. Pharmacokinetics of Plasmid DNA-Based Non-viral Gene Medicine. ADVANCES IN GENETICS 2005; 53PA:47-68. [PMID: 16243060 DOI: 10.1016/s0065-2660(05)53003-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Non-viral gene therapy can be realized by optimization of the pharmacokinetic properties of both the vector and the encoded therapeutic protein. A major obstacle to its successful clinical application is the limited ability of plasmid DNA, the most convenient gene-coding compound, to distribute within the body after in vivo administration. Under normal conditions, plasmid DNA and its non-viral vector complexes have difficulty in passing through various anatomical and biological barriers. These characteristics greatly limit the number and distribution of cells transduced with the vector, because transgene expression only occurs in cells that are reached by the vector. New approaches to the design of vectors as well as the methods of administration, such as electroporation and a hydrodynamic delivery, have increased the transgene expression in vivo, suggesting that improved distribution of plasmid DNA is possible by these approaches. In this chapter, the basic pharmacokinetic properties of naked plasmid DNA under normal conditions are first reviewed, then the properties of both naked and complexed plasmid DNA are discussed under conditions where significant transgene expression takes place.
Collapse
Affiliation(s)
- Makiya Nishikawa
- Department of Biopharmaceutics and Drug Metabolism Graduate School of Pharmaceutical Sciences Kyoto University, Sakyo‐ku, Kyoto 606–8501, Japan
| | | | | |
Collapse
|
43
|
De Vleeschouwer S, Van Gool SW, Van Calenbergh F. Immunotherapy for malignant gliomas: emphasis on strategies of active specific immunotherapy using autologous dendritic cells. Childs Nerv Syst 2005; 21:7-18. [PMID: 15452731 DOI: 10.1007/s00381-004-0994-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2004] [Indexed: 12/25/2022]
Abstract
REVIEW In this review, we discuss immunotherapy for malignant gliomas. EMPHASIS The emphasis is on the novel strategy of active specific immunotherapy using dendritic cells as antigen-presenting cells, especially its theoretical concepts and advantages, specific requirements, critical issues, pre-clinical and early clinical experience. Dendritic cell vaccination is situated in the diversity of other immunotherapeutical approaches. FURTHER DISCUSSION Future directions, challenges, and drawbacks will be discussed.
Collapse
Affiliation(s)
- Steven De Vleeschouwer
- Department of Neurosurgery, University Hospital Gasthuisberg, Herestraat 49, 3000 Leuven, Belgium
| | | | | |
Collapse
|
44
|
Brus C, Kleemann E, Aigner A, Czubayko F, Kissel T. Stabilization of oligonucleotide-polyethylenimine complexes by freeze-drying: physicochemical and biological characterization. J Control Release 2004; 95:119-31. [PMID: 15013239 DOI: 10.1016/j.jconrel.2003.10.021] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2003] [Accepted: 10/18/2003] [Indexed: 10/26/2022]
Abstract
In the present study the lyophilization of oligodeoxynucleotide-polyethylenimine (ODN-PEI) complexes was investigated regarding the maintenance of physicochemical properties and influence on biological activity. To achieve this, we used PEI of different molecular weights, in the range of 800-0.8 kDa, as complexing agents for unmodified ODN and ribozymes. The hydrodynamic diameter was measured by photon correlation spectroscopy (PCS) and the zeta potential was determined using laser Doppler anemometry (LDA) of ODN complexes with PEI derivatives of different molecular weights both before and after lyophilization. Atomic force microscopy (AFM) was used to visualize freshly prepared, stored and lyophilized complexes in solution. The biological activity of the ODN, as well as of plasmid DNA, in lyophilized PEI complexes was examined and compared to freshly prepared complexes using standard transfection assays. All PEI derivatives formed very small complexes with ODN displaying hydrodynamic diameters ranging from 15 to 30 nm. Marginal changes in size after lyophilization were observed for ODN-PEI complexes. In contrast, plasmid complexed with PEI was found to aggregate. In either cases minimal or no influence of the added amount of lyoprotectant was observed. The shape of the very small and highly condensed ODN complexes was not altered by lyophilization as seen in the AFM images. The transfection efficiency of lyophilized ribozyme-PEI complexes relative to freshly prepared complexes was approximately 100%, whereas a decrease was seen for lyophilized plasmid-PEI complexes. An additive of the lyoprotectants trehalose, mannitol or sucrose preserved biological activity. This study demonstrates the particular suitability of ODN-PEI complexes to be formulated as lyophilized systems with no loss in physical stability or biological activity.
Collapse
Affiliation(s)
- Carola Brus
- Department of Pharmaceutics and Biopharmacy, Philipps-University Marburg, Ketzerbach 63, 35037 Marburg, Germany
| | | | | | | | | |
Collapse
|
45
|
Salatino M, Schillaci R, Proietti CJ, Carnevale R, Frahm I, Molinolo AA, Iribarren A, Charreau EH, Elizalde PV. Inhibition of in vivo breast cancer growth by antisense oligodeoxynucleotides to type I insulin-like growth factor receptor mRNA involves inactivation of ErbBs, PI-3K/Akt and p42/p44 MAPK signaling pathways but not modulation of progesterone receptor activity. Oncogene 2004; 23:5161-74. [PMID: 15122317 DOI: 10.1038/sj.onc.1207659] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The present study addresses the effect of targeting type I insulin-like growth factor receptor (IGF-IR) with antisense strategies in in vivo growth of breast cancer cells. Our research was carried out on C4HD tumors from an experimental model of hormonal carcinogenesis in which the synthetic progestin medroxyprogesterone acetate (MPA) induced mammary adenocarcinomas in Balb/c mice. We employed two different experimental strategies. With the first one we demonstrated that direct intratumor injection of phosphorothioate antisense oligodeoxynucleotides (AS[S]ODNs) to IGF-IR mRNA resulted in a significant inhibition of C4HD tumor growth. In the second experimental strategy, we assessed the effect of intravenous (i.v.) injection of AS [S]ODN on C4HD tumor growth. This systemic treatment also resulted in significant reduction in tumor growth. The antitumor effect of IGF-IR AS[S]ODNs in both experimental protocols was due to a specific antisense mechanism, since growth inhibition was dose-dependent and no abrogation of tumor proliferation was observed in mice treated with phosphorothioate sense ODNs (S[S]ODNs). In addition, IGF-IR expression was inhibited in tumors from mice receiving AS[S]ODNs, as compared to tumors from control groups. We then investigated signal transduction pathways modulated in vivo by AS[S]ODNs treatment. Tumors from AS[S]ODN-treated mice of both intratumoral and intravenous protocols showed a significant decrease in the degree of insulin receptor substrate-1 (IRS-1) tyrosine phosphorylation. Activation of two of the main IGF-IR signaling pathways, phosphatidylinositol 3-kinase (PI-3K)/Akt and p42/p44 mitogen-activated protein kinases (MAPK) was abolished in tumors growing in AS[S]ODN-treated animals. Moreover, ErbB-2 tyrosine phosphorylation was blocked by in vivo administration of AS[S]ODNs. On the other hand, we found no regulation of either progesterone receptor expression or activity by in vivo AS[S]ODNs administration. Our results for the first time demonstrated that breast cancer growth can be inhibited by direct in vivo administration of IGF-IR AS[S]ODNs.
Collapse
MESH Headings
- Animals
- Cell Division/drug effects
- Dose-Response Relationship, Drug
- Enzyme Activation
- Epithelial Cells/drug effects
- Epithelial Cells/metabolism
- Epithelial Cells/pathology
- Female
- Genes, erbB-1/drug effects
- Mammary Neoplasms, Experimental/therapy
- Mice
- Mice, Inbred BALB C
- Mitogen-Activated Protein Kinase 1/antagonists & inhibitors
- Mitogen-Activated Protein Kinase 1/metabolism
- Neoplasm Transplantation
- Oligodeoxyribonucleotides, Antisense/pharmacology
- Phosphatidylinositol 3-Kinases/metabolism
- Phosphoinositide-3 Kinase Inhibitors
- RNA, Messenger/drug effects
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Receptor, IGF Type 1/antagonists & inhibitors
- Receptor, IGF Type 1/drug effects
- Receptor, IGF Type 1/metabolism
- Receptors, Progesterone/metabolism
- Signal Transduction/drug effects
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- Mariana Salatino
- Laboratory of Molecular Mechanisms of Carcinogenesis, Instituto de Biología y Medicina Experimental (IBYME), CONICET, Obligado 2490, Buenos Aires 1428, Argentina
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Salamone FN, Gleich LL, Li YQ, Stambrook PJ. Major histocompatibility gene therapy: the importance of haplotype and beta 2-microglobulin. Laryngoscope 2004; 114:612-5. [PMID: 15064612 DOI: 10.1097/00005537-200404000-00004] [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: 12/21/2022]
Abstract
OBJECTIVES/HYPOTHESIS Alloantigen gene therapy with the genes for the Class I major histocompatibility complex (MHC) HLA-B7 and beta 2-microglobulin in HLA-B7-negative patients has potential efficacy in the treatment of head and neck cancer, although the mechanism of response is unclear. Whether tumor regression is due to a response to HLA-B7 in HLA-B7-negative patients (i.e., due to "foreign" antigen) or simply to MHC overexpression is unknown. Therefore, a mouse model was used to compare tumor growth following syngeneic MHC transfection to alloantigenic MHC transfection. The importance of the beta 2-microglobulin gene was also evaluated. STUDY DESIGN Prospective animal study. METHODS The head and neck cancer cell line SCC-VII that grows in immunocompetent C3H mice, which are MHC haplotype H2-K, was used. Stable transfections were made with H2-K, H2-K, and beta 2-microglobulin in the SCC-VII cells. To test the importance of MHC "foreignness," mice were injected with SCC-VII cells, SCC-VII plus H2-K plus beta 2-microglobulin transfected cells, and SCC-VII plus H2-K plus beta 2-microglobulin transfected cells. To evaluate beta 2-microglobulin, mice were injected with SCC-VII cells, SCC-VII plus H2-K plus beta 2-microglobulin transfected cells, SCC-VII plusH2-K transfected cells, and SCC-VII plus beta 2-microglobulin transfected cells. Tumor growth in all groups was compared statistically. RESULTS Major histocompatibility complex foreignness was a part of the antitumor response. Foreign MHC routinely abrogated tumor growth, whereas syngeneic MHC only slowed tumor growth. beta 2-microglobulin aided the MHC tumor inhibition but did not inhibit tumor without the MHC. CONCLUSION The antitumor response was greater when the MHC gene used was foreign. beta 2-microglobulin increased the efficacy of MHC gene therapy. Both of these findings are important when designing clinical trials of immunologically based gene therapies for head and neck cancer.
Collapse
Affiliation(s)
- Frank N Salamone
- Departments of Otolaryngology-Head and Neck Surgery, The University of Cincinnati Medical Center, Cincinnati, Ohio, U.S.A.
| | | | | | | |
Collapse
|
47
|
Kirby TO, Curiel DT, Alvarez RD. Gene therapy for ovarian cancer: progress and potential. Hematol Oncol Clin North Am 2003; 17:1021-50. [PMID: 12959190 DOI: 10.1016/s0889-8588(03)00055-8] [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: 02/04/2023]
Abstract
Gene therapy remains a promising therapeutic modality for ovarian cancer. Yet much work remains to be done to see gene therapy realize its full potential in elucidating the complex genetic interactions of delivered genes within target cancer cells and in the development of improved vector systems. Because most neoplasms involve multiple mutations, the targeting of a single mutation is unlikely to achieve total tumor control: gene therapy strategies that target multiple cellular processes or invoke various antitumor approaches need to be investigated. Additionally, current vector systems do not transduce ovarian cancer cells efficiently and are hampered by immune responses that further limit their efficacy. Additionally, limitations in vector specificity lead to transduction of normal cells and subsequent toxicity. Investigators are developing refinements to current gene therapy approaches that would address these limitations and that are soon to be incorporated into clinical trials. It is hoped that these advances will lead to improvements in the therapeutic index for ovarian cancer gene therapy and provide another effective therapeutic tool for this deadly disease.
Collapse
Affiliation(s)
- Tyler O Kirby
- Department of Obstetrics and Gynecology, University of Alabama at Birmingham, Birmingham, AL 35233-7333, USA
| | | | | |
Collapse
|
48
|
Kawase A, Nomura T, Yasuda K, Kobayashi N, Hashida M, Takakura Y. Disposition and gene expression characteristics in solid tumors and skeletal muscle after direct injection of naked plasmid DNA in mice. J Pharm Sci 2003; 92:1295-304. [PMID: 12761818 DOI: 10.1002/jps.10392] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Previous studies have suggested that direct injection of naked plasmid DNA (pDNA) into solid tumors can be a useful method for in vivo gene transfer into tumor cells. To gain more insight into this approach, we studied the disposition and gene expression characteristics of naked pDNA after intratumoral injection by direct comparison with those after intramuscular injection in mice. pDNA encoding reporter genes were directly injected into subcutaneous solid tumor models and skeletal muscles. Biodistribution studies using radiolabeled pDNA showed that the elimination of pDNA from the injection site was relatively fast and a part of the pDNA was absorbed from the lymphatic system after both local injections. Confocal microscopic studies using fluorescein-labeled pDNA demonstrated that pDNA distributed efficiently throughout the muscle tissue whereas pDNA localization in the tumor tissue was restricted. Characterization of gene expression clarified the variation in expression level between tumor preparations and some factors affecting the expression level in the tumor. Reporter gene expression was significantly inhibited by simultaneous administration of some polyanions in both cases, suggesting that a specific mechanism may be involved in the naked pDNA uptake by muscle and tumor cells. These findings provide useful information for direct naked pDNA delivery into solid tumors.
Collapse
Affiliation(s)
- Atsushi Kawase
- Department of Biopharmaceutics and Drug Metabolism, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | | | | | | | | | | |
Collapse
|
49
|
Guo SY, Gu QL, Zhu ZG, Hong HQ, Lin YZ. TK gene combined with mIL-2 and mGM-CSF genes in treatment of gastric cancer. World J Gastroenterol 2003; 9:233-7. [PMID: 12532437 PMCID: PMC4611317 DOI: 10.3748/wjg.v9.i2.233] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: Cancer gene therapy has received more and more attentions in the recent decade. Various systems of gene therapy for cancer have been developed. One of the most promising choices is the suicide gene. The product of thymidine kinase (TK) gene can convert ganciclovir (GCV) to phosphorylated GCV, which inhibits the synthesis of cell DNA, and then induces the cells to death. Cytokines play an important role in anti-tumor immunity. This experiment was designed to combine the TK gene and mIL-2/mGM-CSF genes to treat gastric cancer, and was expected to produce a marked anti-tumor effect.
METHODS: TK gene was constructed into the retroviral vector pLxSN, and the mIL-2 and mGM-CSF genes were inserted into the eukaryotic expressing vector pIRES. The gastric cancer cells were transfected by retroviral serum that was harvested from the package cells. In vitro study, the transfected gastric cancer cells were maintained in the GCV- contained medium, to assay the cell killing effect and bystander effect. In vivo experiment, retroviral serum and cytokines plasmid were transfected into tumor-bearing mice, to observe the changes of tumor volumes and survival of the mice.
RESULTS: In vitro experiment, 20% TK gene transduced cells could cause 70%-80% of total cells to death. In vivo results showed that there was no treatment effect in control group and TK/GCV could inhibit the tumor growth. The strongest anti-tumor effect was shown in TK+mIL-2+mGM-CSF group. The pathologic examination showed necrosis of the cancer in the treated groups.
CONCLUSION: TK/GCV can kill tumor cells and inhibit the tumor growth in vivo. IL-2 and GM-CSF strongly enhance the anti-tumor effect. Through the retrovirus and liposome methods, the suicide gene and cytokine genes are all expressed in the tissues.
Collapse
Affiliation(s)
- Shan-Yu Guo
- Department of Surgery, the Affiliated Shanghai Ninth People's Hospital, Shanghai Second Medical University, Shanghai 200011, China.
| | | | | | | | | |
Collapse
|
50
|
Dols A, Meijer SL, Smith JW, Fox BA, Urba WJ. Allogeneic breast cancer cell vaccines. Clin Breast Cancer 2003; 3 Suppl 4:S173-80. [PMID: 12620156 DOI: 10.3816/cbc.2003.s.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Cancer vaccines are currently a major focus of immunotherapy research. The combination of specific targeting and low levels of toxicity makes vaccination an attractive approach. There are a variety of immunogens that can be employed to vaccinate patients in order to induce or enhance an antitumor response. The observation that most T-cell priming occurs via presentation of tumor antigens from tumor cells engulfed by host antigen-presenting cells, rather than by direct presentation by vaccine tumor cells themselves, provides the immunological rationale for an allogeneic tumor cell vaccine approach. Furthermore, there are practical advantages over an autologous tumor cell vaccine approach. We summarize herein the limited experience using allogeneic whole cell vaccines in patients with breast cancer. We also describe in vitro immunological results using peripheral blood mononuclear cells from women with stage IV breast cancer who were enrolled in a phase I trial employing a human leukocyte antigen-A2-matched, CD80-modified, allogeneic, whole cell vaccine. Clinical trials employing allogeneic tumor cell vaccines have achieved encouraging immunological and clinical effects in stage IV patients. Allogeneic tumor cell vaccines are safe, feasible, and associated with low toxicity, and the early clinical results suggest that they are worthy of further study
Collapse
Affiliation(s)
- Annemieke Dols
- Robert W. Franz Cancer Research Center, Earle A. Chiles Research Institute, Providence Portland Medical Center, OR 97213, USA
| | | | | | | | | |
Collapse
|