1
|
Dong L, Zhang X, Yu C, Yu T, Liu S, Hou L, Fu L, Yi S, Chen W. Monitoring luciferase-labeled human prostate stem cell antigen-expressing tumor growth in a mouse model. Exp Ther Med 2013; 6:1208-1212. [PMID: 24223645 PMCID: PMC3820663 DOI: 10.3892/etm.2013.1293] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2013] [Accepted: 09/03/2013] [Indexed: 11/11/2022] Open
Abstract
The aim of this study was to establish a tumor model in mice with the expression of luciferase (Luc) and human prostate stem cell antigen (PSCA), in order to evaluate the activities of anticancer drugs or vaccines for prostate cancer. RM-1 cells were stably transfected with pcDNA-Luc and pcDNA-PSCA plasmids. The Luc-expressing cells were examined using a luminometer and the PSCA-expressing cells were examined using a reverse transcription-polymerase chain reaction (RT-PCR) and flow cytometric analysis. Male C57BL/6 mice were inoculated subcutaneously with the RM-PSCA/Luc cells, prior to the tumor growth and survival time of the mice being measured, respectively. In vivo bioluminescence imaging was used to detect Luc expression and immunohistochemical analysis was used to detect PSCA expression. Inoculation of the tumor cells into the C57BL/6 mice closely mimicked the tumor growth of prostate cancer. All of the inoculated mice exhibited a detectable tumor within two weeks. Tumor progression was able to be quantitatively monitored following the inoculation of 1×106 RM-PSCA/Luc cells. There was an excellent correlation (R2=0.9849) between the photon counts and tumor volume. The expression of PSCA in tumor tissues was confirmed using immunohistochemical analysis. The Luc and PSCA co-expression tumor model was successfully established in mice, which is likely to accelerate the understanding of the pathogenesis of prostate cancer and facilitate the development of novel antitumor drugs or vaccines for the disease.
Collapse
Affiliation(s)
- Lei Dong
- Clinical Laboratory Center, PLA Air Force General Hospital, Haidian, Beijing 100142
| | | | | | | | | | | | | | | | | |
Collapse
|
2
|
Dong L, Zhang X, Ren J, Wu S, Yu T, Hou L, Fu L, Yi S, Yu C. Human Prostate Stem Cell Antigen and HSP70 Fusion Protein Vaccine Inhibits Prostate Stem Cell Antigen-Expressing Tumor Growth in Mice. Cancer Biother Radiopharm 2013; 28:391-7. [DOI: 10.1089/cbr.2012.1357] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Lei Dong
- Clinical Laboratory Center, PLA Air Force General Hospital, Beijing, China
| | - Xiaopeng Zhang
- Beijing Institute of Biotechnology, Academy of Military Medical Sciences, Beijing, China
| | - Jun Ren
- Beijing Institute of Biotechnology, Academy of Military Medical Sciences, Beijing, China
| | - Shipo Wu
- Beijing Institute of Biotechnology, Academy of Military Medical Sciences, Beijing, China
| | - Ting Yu
- Beijing Institute of Biotechnology, Academy of Military Medical Sciences, Beijing, China
| | - Lihua Hou
- Beijing Institute of Biotechnology, Academy of Military Medical Sciences, Beijing, China
| | - Ling Fu
- Beijing Institute of Biotechnology, Academy of Military Medical Sciences, Beijing, China
| | - Shaoqiong Yi
- Beijing Institute of Biotechnology, Academy of Military Medical Sciences, Beijing, China
| | - Changming Yu
- Beijing Institute of Biotechnology, Academy of Military Medical Sciences, Beijing, China
| |
Collapse
|
3
|
Prostate-specific membrane antigen-based therapeutics. Adv Urol 2011; 2012:973820. [PMID: 21811498 PMCID: PMC3145341 DOI: 10.1155/2012/973820] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2011] [Accepted: 05/09/2011] [Indexed: 12/21/2022] Open
Abstract
Prostate cancer (PC) is the most common noncutaneous malignancy affecting men in the US, leading to significant morbidity and mortality. While significant therapeutic advances have been made, available systemic therapeutic options are lacking. Prostate-specific membrane antigen (PSMA) is a highly-restricted prostate cell-surface antigen that may be targeted. While initial anti-PSMA monoclonal antibodies were suboptimal, the development of monoclonal antibodies such as J591 which are highly specific for the external domain of PSMA has allowed targeting of viable, intact prostate cancer cells. Radiolabeled J591 has demonstrated accurate and selective tumor targeting, safety, and efficacy. Ongoing studies using anti-PSMA radioimmunotherapy with 177Lu-J591 seek to improve the therapeutic profile, select optimal candidates with biomarkers, combine with chemotherapy, and prevent or delay the onset of metastatic disease for men with biochemical relapse. Anti-PSMA monoclonal antibody-drug conjugates have also been developed with completed and ongoing early-phase clinical trials. As PSMA is a selective antigen that is highly overexpressed in prostate cancer, anti-PSMA-based immunotherapy has also been studied and utilized in clinical trials.
Collapse
|
4
|
Shanmugam A, Suriano R, Chaudhuri D, Rajoria S, George A, Mittelman A, Tiwari RK. Identification of PSA peptide mimotopes using phage display peptide library. Peptides 2011; 32:1097-102. [PMID: 21539876 DOI: 10.1016/j.peptides.2011.04.018] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2010] [Revised: 04/15/2011] [Accepted: 04/16/2011] [Indexed: 11/21/2022]
Abstract
Prostate cancer (PCa) is one of the most common types of cancer in men in the United States and is the second leading cause of cancer related death in men. Clinically, secreted prostate specific antigen (PSA) has gained recognition because of its proteolytic activity being directly linked to PCa cell proliferation leading to disease initiation and progression. Using phage display technology, we identified four distinct cyclical peptides. These peptides apart from differences in their amino acid sequence, elicited minimal cross reactive antibody responses against each other. One of the four peptides analyzed produced an antibody response that recognizes the PSA protein. We demonstrate that the synthetic PSA peptide mimics identified in our study are immunologically active and produce neutralizing activity and this has relevance and utility for prostate cancer disease progression.
Collapse
Affiliation(s)
- Arulkumaran Shanmugam
- Department of Microbiology and Immunology, New York Medical College, Valhalla, New York 10595, USA
| | | | | | | | | | | | | |
Collapse
|
5
|
DNA vaccination: using the patient's immune system to overcome cancer. Clin Dev Immunol 2010; 2010:169484. [PMID: 21197271 PMCID: PMC3010826 DOI: 10.1155/2010/169484] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2010] [Revised: 10/08/2010] [Accepted: 10/21/2010] [Indexed: 12/15/2022]
Abstract
Cancer is one of the most challenging diseases of today. Optimization of standard treatment protocols consisting of the main columns of chemo- and radiotherapy followed or preceded by surgical intervention is often limited by toxic side effects and induction of concomitant malignancies and/or development of resistant mechanisms. This requires the development of therapeutic strategies which are as effective as standard therapies but permit the patients a life without severe negative side effects. Along this line, the development of immunotherapy in general and the innovative concept of DNA vaccination in particular may provide a venue to achieve this goal. Using the patient's own immune system by activation of humoral and cellular immune responses to target the cancer cells has shown first promising results in clinical trials and may allow reduced toxicity standard therapy regimen in the future. The main challenge of this concept is to transfer the plethora of convincing preclinical and early clinical results to an effective treatment of patients.
Collapse
|
6
|
Vaccination with a chaperone complex based on PSCA and GRP170 adjuvant enhances the CTL response and inhibits the tumor growth in mice. Vaccine 2010; 28:6333-7. [PMID: 20637304 DOI: 10.1016/j.vaccine.2010.06.093] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2009] [Revised: 05/13/2010] [Accepted: 06/30/2010] [Indexed: 01/03/2023]
Abstract
Increasing knowledge demonstrate that prostate stem cell antigen (PSCA) is a promising candidate for immunotherapy of advanced prostate cancer. However, tumor escape with down-regulation of target antigens may limit the susceptibility of tumor cells to the immune attack. Concomitant generation of T-cell responses against several immunodominant antigens may circumvent this potential drawback. In this study, we prepared the chaperone complex vaccine based on PSCA and GRP170, and utilized it to immunize the C57BL/6 mice. In addition, the T-cell response was monitored with ELISPOT and (51)Cr-release assays, and the tumor growth and the life span of tumor-bearing mice were assessed. The results demonstrated the chaperone complex based on PSCA and GRP170 could enhance the T-cell mediate immune responses, which significantly inhibited the tumor growth and prolonged the life span of tumor-bearing mice. In conclusion, our findings supported the strategy of chaperone complex, based on PSCA and GRP170, could be an effective treatment for prostate cancer therapy.
Collapse
|
7
|
Prostate cancer immunotherapy yields superior long-term survival in TRAMP mice when administered at an early stage of carcinogenesis prior to the establishment of tumor-associated immunosuppression at later stages. Vaccine 2010; 27 Suppl 6:G52-9. [PMID: 20006141 DOI: 10.1016/j.vaccine.2009.09.106] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2009] [Accepted: 09/24/2009] [Indexed: 12/15/2022]
Abstract
Prostate cancer immunotherapy clinical trials have been performed, but often in immunocompromised patients with limited clinical success. The study aim was to determine whether the stage of prostate cancer development at which immunization occurs affects vaccine efficacy, and if so which tumor-associated immunosuppressive mechanisms may be involved at later stages. Therapeutic vaccination of TRAMP mice with only precancerous PIN lesions confered superior protection to immunization after development of invasive carcinoma. The presence of Treg, upregulation of tumor indoleamine-2,3-dioxygenase and TGFbeta and an immunosuppressive intratumoral cytokine milieu were identified in more advanced prostate cancer. These results indicate that prostate cancer immunotherapy trials will be more successful if conducted in patients with less advanced disease.
Collapse
|
8
|
Garcia-Hernandez MDLL, Gray A, Hubby B, Klinger OJ, Kast WM. Prostate stem cell antigen vaccination induces a long-term protective immune response against prostate cancer in the absence of autoimmunity. Cancer Res 2008; 68:861-9. [PMID: 18245488 DOI: 10.1158/0008-5472.can-07-0445] [Citation(s) in RCA: 136] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Prostate stem cell antigen (PSCA) is an attractive antigen to target using therapeutic vaccines because of its overexpression in prostate cancer, especially in metastatic tissues, and its limited expression in other organs. Our studies offer the first evidence that a PSCA-based vaccine can induce long-term protection against prostate cancer development in prostate cancer-prone transgenic adenocarcinoma mouse prostate (TRAMP) mice. Eight-week-old TRAMP mice displaying prostate intraepithelial neoplasia were vaccinated with a heterologous prime/boost strategy consisting of gene gun-delivered PSCA-cDNA followed by Venezuelan equine encephalitis virus replicons encoding PSCA. Our results show the induction of an immune response against a newly defined PSCA epitope that is mediated primarily by CD8 T cells. The prostates of PSCA-vaccinated mice were infiltrated by CD4-positive, CD8-positive, CD11b-positive, and CD11c-positive cells. Vaccination induced MHC class I expression and cytokine production [IFN-gamma, tumor necrosis factor-alpha, interleukin 2 (IL-2), IL-4, and IL-5] within prostate tumors. This tumor microenvironment correlated with low Gleason scores and weak PSCA staining on tumor cells present in hyperplastic zones and in areas that contained focal and well-differentiated adenocarcinomas. PSCA-vaccinated TRAMP mice had a 90% survival rate at 12 months of age. In contrast, all control mice had succumbed to prostate cancer or had heavy tumor loads. Crucially, this long-term protective immune response was not associated with any measurable induction of autoimmunity. The possibility of inducing long-term protection against prostate cancer by vaccination at the earliest signs of its development has the potential to cause a dramatic paradigm shift in the treatment of this disease.
Collapse
Affiliation(s)
- Maria de la Luz Garcia-Hernandez
- Department of Molecular Microbiology and Immunology and Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California 90033, USA
| | | | | | | | | |
Collapse
|
9
|
Li N, Qin H, Li X, Zhou C, Wang D, Ma W, Lin C, Zhang Y, Wang S, Zhang S. Synergistic antitumor effect of chemotactic-prostate tumor-associated antigen gene-modified tumor cell vaccine and anti-CTLA-4 mAb in murine tumor model. Immunol Lett 2007; 113:90-8. [PMID: 17913245 DOI: 10.1016/j.imlet.2007.07.019] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2007] [Revised: 07/22/2007] [Accepted: 07/27/2007] [Indexed: 11/23/2022]
Abstract
In this study, we demonstrate that an effective immune response against prostate tumors in mouse tumor model can be elicited using a strategy that combines CTLA-4 blockade and pSLC-3P-Fc-modified tumor cell vaccine (named B16F10-SLC-3P-Fc). Treatment of B16F10-3P-bearing mice resulted in a significant reduction in tumor incidence as assessed 2 months after treatment. In vivo Ab depletion confirmed that the antitumor effect was primarily CD8+ T cells and CD4+ T lymphocytes were required for the induction of CD8+ CTL response in B16F10-SLC-3P-Fc+anti-CTLA-4 mAb-immunized mice. Moreover, mice that were cured of an established tumor were protected against a rechallenge with the same tumor for at least 4 months, suggesting the generation of memory responses. Adoptive transfer experiments further indicate that antitumor reactivity can be transferred to naïve mice by splenocytes. These findings demonstrate that this combinatorial treatment can elicit a potent anti-tumor immune response and suggest potential of this approach for treatment of prostate cancer.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/immunology
- Antibodies, Monoclonal/therapeutic use
- Antibodies, Neoplasm/immunology
- Antigens, CD/immunology
- Antigens, Differentiation/immunology
- Antigens, Neoplasm/immunology
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/metabolism
- CTLA-4 Antigen
- Cancer Vaccines/immunology
- Cancer Vaccines/therapeutic use
- Immunotherapy
- Immunotherapy, Adoptive
- Male
- Melanoma, Experimental/immunology
- Mice
- Mice, Inbred C57BL
- Prostate/immunology
- Prostate-Specific Antigen/immunology
- Prostatic Neoplasms/immunology
- Prostatic Neoplasms/therapy
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Cytotoxic/metabolism
Collapse
Affiliation(s)
- Ning Li
- Department of Immunology, Cancer Institute, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Garcia-Hernandez MDLL, Gray A, Hubby B, Kast WM. In vivo effects of vaccination with six-transmembrane epithelial antigen of the prostate: a candidate antigen for treating prostate cancer. Cancer Res 2007; 67:1344-51. [PMID: 17283172 DOI: 10.1158/0008-5472.can-06-2996] [Citation(s) in RCA: 85] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Immunotherapy may provide an alternative treatment for cancer patients, especially when tumors overexpress antigens that can be recognized by immune cells. The identification of markers and therapeutic targets that are up-regulated in prostate cancer has been important to design new potential treatments for prostate cancer. Among them, the recently identified six-transmembrane epithelial antigen of the prostate (STEAP) is considered attractive due to its overexpression in human prostate cancer tissues. Our study constitutes the first assessment of the in vivo effectiveness of STEAP-based vaccination in prophylactic and therapeutic mouse models. Two delivery systems, cDNA delivered by gene gun and Venezuelan equine encephalitis virus-like replicon particles (VRP), both encoding mouse STEAP (mSTEAP) and three vaccination strategies were used. Our results show that mSTEAP-based vaccination was able to induce a specific CD8 T-cell response against a newly defined mSTEAP epitope that prolonged the overall survival rate in tumor-challenged mice very significantly. This was achieved without any development of autoimmunity. Surprisingly, CD4 T cells that produced IFNgamma, tumor necrosis factor-alpha (TNF-alpha), and interleukin-2 (IL-2) played the main role in tumor rejection in our model as shown by using CD4- and CD8-deficient mice. In addition, the presence of high IL-12 levels in the tumor environment was associated with a favorable antitumor response. Finally, the therapeutic effect of STEAP vaccination was also assessed and induced a modest but significant delay in growth of established, 31 day old tumors. Taken together, our data suggest that vaccination against mSTEAP is a viable option to delay tumor growth.
Collapse
|
11
|
Wang Y, Xu M, Che M, Von Hofe E, Abbas A, Kallinteris NL, Lu X, Liss ZJ, Forman JD, Hillman GG. Curative Antitumor Immune Response Is Optimal with Tumor Irradiation Followed by Genetic Induction of Major Histocompatibility Complex Class I and Class II Molecules and Suppression of Ii Protein. Hum Gene Ther 2005; 16:187-99. [PMID: 15761259 DOI: 10.1089/hum.2005.16.187] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Transfecting genes into tumors, to upregulate major histocompatibility complex (MHC) class I and class II molecules and inhibit MHC class II associated invariant chain (Ii), induces a potent anti-tumor immune response when preceded by tumor irradiation, in murine RM-9 prostate carcinoma. The transfected genes are cDNA plasmids for interferon-gamma (pIFN-gamma), MHC class II transactivator (pCIITA), an Ii reverse gene construct (pIi-RGC), and a subtherapeutic dose of adjuvant IL-2 (pIL-2). Responding mice rejected challenge with parental tumor and demonstrated tumor-specific cytotoxic T lymphocytes (CTLs). We have extended our investigation to determine the relative roles of each one of the four plasmids pIFN-gamma, pCIITA, pIi-RGC, and pIL-2 in conjunction with radiation for the induction of a curative immune response. Upregulation of MHC class I with pIFN-gamma or class II with pCIITA, separately, does not lead to a complete response even if supplemented with pIL-2 or pIi-RGC. An optimal and specific antitumor response is achieved in more than 50% of the mice when, after tumor irradiation, tumor cells are converted in situ to a MHC class I+/class II+/Ii- phenotype with pIFN-gamma, pCIITA, pIi-RGC, and pIL-2. We demonstrate further that both CD4+ helper T cells and CD8+ cytotoxic T cells are essential for induction of an antitumor response because in vivo depletion of either subset abrogates the response. The radiation contributes to the gene therapy by causing tumor debulking and increasing the permeability of tumors to infiltration of inflammatory cells.
Collapse
MESH Headings
- Animals
- Antigens, Differentiation, B-Lymphocyte/chemistry
- Antigens, Differentiation, B-Lymphocyte/genetics
- Antigens, Differentiation, B-Lymphocyte/metabolism
- CD4-Positive T-Lymphocytes/metabolism
- CD8-Positive T-Lymphocytes/metabolism
- Cell Survival/physiology
- Cell Survival/radiation effects
- Colony-Forming Units Assay
- Combined Modality Therapy
- Gene Expression
- Genetic Therapy
- Histocompatibility Antigens Class I/genetics
- Histocompatibility Antigens Class II/chemistry
- Histocompatibility Antigens Class II/genetics
- Histocompatibility Antigens Class II/metabolism
- Interferon-gamma/genetics
- Interferon-gamma/metabolism
- Interleukin-2/genetics
- Interleukin-2/metabolism
- Lymphocyte Depletion
- Male
- Mice
- Mice, Inbred C57BL
- Neoplasm Proteins/genetics
- Nuclear Proteins/genetics
- Nuclear Proteins/metabolism
- Prostatic Neoplasms/genetics
- Prostatic Neoplasms/immunology
- Prostatic Neoplasms/radiotherapy
- T-Lymphocytes, Cytotoxic/immunology
- Trans-Activators/genetics
- Trans-Activators/metabolism
- Transduction, Genetic
- Tumor Cells, Cultured
- X-Rays
Collapse
Affiliation(s)
- Yu Wang
- Department of Radiation Oncology, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine and Harper Hospital, Detroit, MI 48201, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Trudel S, Trachtenberg J, Toi A, Sweet J, Li ZH, Jewett M, Tshilias J, Zhuang LH, Hitt M, Wan Y, Gauldie J, Graham FL, Dancey J, Stewart AK. A phase I trial of adenovector-mediated delivery of interleukin-2 (AdIL-2) in high-risk localized prostate cancer. Cancer Gene Ther 2004; 10:755-63. [PMID: 14502228 DOI: 10.1038/sj.cgt.7700626] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Preclinical studies demonstrate that intratumoral delivery of adenovirus expressing IL-2 eradicates pre-established tumors in mice and confers immune protection from rechallenge. To explore the activity of AdCAIL-2 in prostate cancer, a Phase I clinical trial was conducted in patients with localized disease and Gleason score >7 or prostate-specific antigen (PSA) >10 plus Gleason score 7. A total of 12 patients were injected 4 weeks prior to prostatectomy in a dose-escalation study at doses of 10(9), 5 x 10(9) and 10(10) PFU of virus. No dose-limiting toxicity was observed. Side effects included perineal discomfort, hematuria, flu-like symptoms in two patients and urinary hesitancy in one patient. Pathology demonstrated an inflammatory response consisting predominantly of CD3+CD8+ T lymphocytes with areas of tumor necrosis. Intracellular cytokine staining of tumor-infiltrating lymphocytes demonstrated increases in both gamma-interferon and IL-4 secreting T cells after vaccination. PSA levels fell in five of five evaluable patients treated at the lowest dose (mean decline of 33.3%, range 17-69%). At higher doses, PSA values initially increased after injection, then fell to baseline prior to surgery. This trial demonstrates the feasibility and safety of intraprostatic adenovector-mediated IL-2 gene delivery.
Collapse
Affiliation(s)
- Suzane Trudel
- 1Division of Hematology-Oncology and Urology, The Princess Margaret Hospital, 610 University Avenue, Toronto, Ontario, Canada M5G 2M9
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Hillman GG, Maughan RL, Grignon DJ, Yudelev M, Che M, Abrams J, Wang Y, Layer A, Wright JL, Rubio J, Forman JD. Responsiveness of experimental prostate carcinoma bone tumors to neutron or photon radiation combined with cytokine therapy. Int J Radiat Oncol Biol Phys 2003; 56:1426-37. [PMID: 12873689 DOI: 10.1016/s0360-3016(03)00438-3] [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: 11/19/2022]
Abstract
PURPOSE To improve the outcome of radiotherapy for prostate carcinoma bone tumors, we investigated bone tumor irradiation with photons or neutrons followed by interleukin 2 (IL-2) therapy in a tumor model. METHODS AND MATERIALS Implantation of PC-3 cells in nude mouse femur cavity induced a bone tumor that progressed to the formation of a palpable tumor, at the hip joint, by Day 20. Established bone tumors were irradiated with photons or neutrons, and a day later, mice were treated with IL-2 therapy for 3 weekly cycles. RESULTS PC-3 bone tumors responded to radiation with photons or neutrons in a dose-dependent manner. Combination of photon or neutron radiation with IL-2 therapy increased tumor growth delay, compared to that with photons or neutrons alone. Radiation alone or combined with IL-2 significantly increased mouse survival compared to that with IL-2 or no treatment. After combined therapy, a complete inhibition of bone tumor growth was observed in 45% to 50% of the mice. Histologically, the combined therapy resulted in greater tumor destruction associated with fibrosis, new bone formation, and inflammatory infiltrates than that observed with each modality alone. CONCLUSIONS The efficacy of tumor irradiation with neutrons or photons was enhanced by IL-2 therapy for the treatment of prostate carcinoma bone tumors.
Collapse
Affiliation(s)
- Gilda G Hillman
- Department of Radiation Oncology, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine and Harper Hospital, Detroit, MI 48201, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Hillman GG, Xu M, Wang Y, Wright JL, Lu X, Kallinteris NL, Tekyi-Mensah S, Thompson TC, Mitchell MS, Forman JD. Radiation improves intratumoral gene therapy for induction of cancer vaccine in murine prostate carcinoma. Hum Gene Ther 2003; 14:763-75. [PMID: 12804139 DOI: 10.1089/104303403765255156] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Our goal was to convert murine RM-9 prostate carcinoma cells in vivo into antigen-presenting cells capable of presenting endogenous tumor antigens and triggering a potent T-helper cell-mediated immune response essential for the generation of a specific antitumor response. We showed that generating the major histocompatibility complex (MHC) class I+/class II+/Ii- phenotype, within an established subcutaneous RM-9 tumor nodule, led to an effective immune response limiting tumor growth. This phenotype was created by intratumoral injection of plasmid cDNAs coding for interferon gamma, MHC class II transactivator, and an antisense reverse gene construct (RGC) for a segment of the gene for Ii protein (-92,97). While this protocol led to significant suppression of tumor growth, there were no disease-free survivors. Nevertheless, irradiation of the tumor nodule on the day preceding initiation of gene therapy yielded 7 of 16 mice that were disease-free in a long-term follow up of 57 days compared to 1 of 7 mice receiving radiotherapy alone. Mice receiving radiotherapy and gene therapy rejected challenge with parental RM-9 cells and demonstrated specific cytotoxic T-cell activity in their splenocytes but not the mouse cured by radiation alone. These data were reproduced in additional experiments and confirmed that tumor irradiation prior to gene therapy resulted in complete tumor regression and specific tumor immunity in more than 50% of the mice. Increasing the number of plasmid injections after tumor irradiation induced tumor regression in 70% of the mice. Administering radiation before this novel gene therapy approach, that creates an in situ tumor vaccine, holds promise for the treatment of human prostate carcinoma.
Collapse
Affiliation(s)
- Gilda G Hillman
- Department of Radiation Oncology, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine and Harper Hospital, Detroit, MI 48201, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Looking to the Future: Advances in the Management of Hormone-Refractory Prostate Cancer. ACTA ACUST UNITED AC 2002. [DOI: 10.1016/s1569-9056(02)00087-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
16
|
Reader S, Ménard S, Filion B, Filion MC, Phillips NC. Pro-apoptotic and immunomodulatory activity of a mycobacterial cell wall-DNA complex towards LNCaP prostate cancer cells. Prostate 2001; 49:155-65. [PMID: 11746260 DOI: 10.1002/pros.1130] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
BACKGROUND We have isolated a mycobacterial cell wall-DNA complex (MCC) possessing anti-cancer activity against bladder cancer cells. The anti-cancer activity of MCC appears to be due to two effects: a direct interaction with bladder cancer cells resulting in the induction of apoptosis and an indirect effect via the stimulation of monocytes and macrophages cytokine synthesis. In this study, the direct effect of MCC towards LNCaP cancer cells was evaluated. METHODS Inhibition of proliferation, cell cycle arrest and induction of apoptosis were evaluated in vitro using LNCaP cells treated with MCC. The synthesis of IL-12, GM-CSF, and TNF-alpha by LNCaP cells in response to MCC was also determined. Experiments were performed to gain insight into the mechanism of action of MCC towards LNCaP cells. RESULTS MCC caused a dose-dependent inhibition of the proliferation of LNCaP cells that was associated with cell cycle arrest at the G0/G1 phase. MCC-induced apoptosis of LNCaP cells was consistent with a mitochondrial pathway involving mitochondrial disruption, release of cytochrome c, and an increase in Bax protein levels leading to caspase-3 and -7 activation and cleavage of poly (ADP-ribose) polymerase and nuclear mitotic apparatus protein. Surprisingly, MCC also directly induced the synthesis of IL-12 and GM-CSF, but not TNF-alpha, by LNCaP cells. CONCLUSIONS MCC possesses the ability to directly induce apoptosis of LNCaP cells and to trigger the synthesis of IL-12 and GM-CSF by these cells, suggesting a potential role of MCC for the treatment of prostate cancer.
Collapse
Affiliation(s)
- S Reader
- Bioniche Therapeutics Research Centre, Montréal, Québec, Canada
| | | | | | | | | |
Collapse
|
17
|
Heiser A, Dahm P, Yancey DR, Maurice MA, Boczkowski D, Nair SK, Gilboa E, Vieweg J. Human dendritic cells transfected with RNA encoding prostate-specific antigen stimulate prostate-specific CTL responses in vitro. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2000; 164:5508-14. [PMID: 10799919 DOI: 10.4049/jimmunol.164.10.5508] [Citation(s) in RCA: 151] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Although immunological tolerance to self Ags represents an important mechanism to prevent normal tissue injury, there is growing evidence that tolerance to tumor Ags, which often represent normal peripherally expressed proteins, is not absolute and can be effectively reverted. Prostate-specific Ag (PSA) is a self Ag expressed by both normal and malignant prostatic epithelium, and therefore offers a unique opportunity to examine the ability of self Ags to serve as specific CTL targets. In this study, we investigated the efficacy of autologous dendritic cells (DC) transfected with mRNA encoding PSA to stimulate CTL against PSA Ags in vitro. Ag in form of RNA carries the advantage to encode multiple epitopes for many HLA alleles, thus permitting induction of CTL responses among many cancer patients independent of their HLA repertoire. In this study, we show that PSA mRNA-transfected DC were capable of stimulating primary CTL responses against PSA Ags in vitro. The PSA-specific CTL did not cross-react with kallikrein Ags, a protein, which shares significant homology with PSA, suggesting that harmful autoimmune toxicity may not represent a significant problem with this approach. PSA RNA-transfected DC generated from male or female healthy volunteers or from cancer patients were equally effective in stimulating PSA-specific CTL in vitro, implying that neither natural tolerance to PSA Ags nor tumor-mediated T cell anergy may represent major barriers for CTL generation against the self Ag PSA. This study provides a preclinical rationale for using PSA RNA-transfected DC in active or adoptive immunization protocols.
Collapse
Affiliation(s)
- A Heiser
- Division of Urology and Department of Surgery, Duke University Medical Center, Durham, NC 27710, USA
| | | | | | | | | | | | | | | |
Collapse
|