1
|
Knockdown of LncRNA MALAT1 contributes to cell apoptosis via regulating NF-κB/CD80 axis in neonatal respiratory distress syndrome. Int J Biochem Cell Biol 2018; 104:138-148. [PMID: 30243953 DOI: 10.1016/j.biocel.2018.09.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 09/16/2018] [Accepted: 09/18/2018] [Indexed: 01/28/2023]
Abstract
Neonatal respiratory distress syndrome (NRDS) is a leading cause of morbidity in premature newborns and is a common reason for admission to the neonatal intensive care unit (NICU). Recent studies found that the pathogenesis of NRDS is not simply lung immaturity. Apoptosis is an essential process for the development and maturation of the lungs. In this study, we report a critical role of lncRNA MALAT1 in regulating CD80 transcription in the NRDS-associated apoptosis via binding with NF-κB. We first showed MALAT1 and CD80 were highly expressed in the peripheral blood mononuclear cells of NRDS with infection exposure. Then we found MALAT1 expressions were significantly increased by the treatment of LPS. We confirmed knockdown of MALAT1 promoted cell viability by CCK-8 assays, cell apoptosis by flow cytometric assays and cytoskeleton destruction by immunocytochemistry. We confirmed CD80 expression level was associated with cell apoptosis by affecting PARP and caspase-3. Then we demonstrated knockdown of MALAT1 promoted CD80 transcription in A549 cells. Furthermore, we confirmed that MALAT1 downregulated transcriptional expression of CD80 by interfering with NF-κB activation and disrupting its binding efficiency with the CD80 promoter in the cell nucleus. In conclusion, we first identified lncRNA MALAT1 as an important prognosis maker for NRDS patients. Most significantly, this study then demonstrated a novel regulatory function of knocked-down MALAT1 on the transcriptional level of CD80 by enhancing the binding of NF-κB to CD80 promoter. Since the interaction between MALAT1 and CD80 plays an essential role in the cell apoptosis of NRDS, our findings demonstrate the possibility of using MALAT1 as therapeutic target for treatment of NRDS, and extend existing knowledge about the molecular mechanism that underlies NRDS pathogensis.
Collapse
|
2
|
Du X, Tang F, Liu M, Su J, Zhang Y, Wu W, Devenport M, Lazarski CA, Zhang P, Wang X, Ye P, Wang C, Hwang E, Zhu T, Xu T, Zheng P, Liu Y. A reappraisal of CTLA-4 checkpoint blockade in cancer immunotherapy. Cell Res 2018; 28:416-432. [PMID: 29472691 PMCID: PMC5939050 DOI: 10.1038/s41422-018-0011-0] [Citation(s) in RCA: 173] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Revised: 12/11/2017] [Accepted: 12/20/2017] [Indexed: 12/13/2022] Open
Abstract
It is assumed that anti-CTLA-4 antibodies cause tumor rejection by blocking negative signaling from B7-CTLA-4 interactions. Surprisingly, at concentrations considerably higher than plasma levels achieved by clinically effective dosing, the anti-CTLA-4 antibody Ipilimumab blocks neither B7 trans-endocytosis by CTLA-4 nor CTLA-4 binding to immobilized or cell-associated B7. Consequently, Ipilimumab does not increase B7 on dendritic cells (DCs) from either CTLA4 gene humanized (Ctla4h/h) or human CD34+ stem cell-reconstituted NSG™ mice. In Ctla4h/m mice expressing both human and mouse CTLA4 genes, anti-CTLA-4 antibodies that bind to human but not mouse CTLA-4 efficiently induce Treg depletion and Fc receptor-dependent tumor rejection. The blocking antibody L3D10 is comparable to the non-blocking Ipilimumab in causing tumor rejection. Remarkably, L3D10 progenies that lose blocking activity during humanization remain fully competent in inducing Treg depletion and tumor rejection. Anti-B7 antibodies that effectively block CD4 T cell activation and de novo CD8 T cell priming in lymphoid organs do not negatively affect the immunotherapeutic effect of Ipilimumab. Thus, clinically effective anti-CTLA-4 mAb causes tumor rejection by mechanisms that are independent of checkpoint blockade but dependent on the host Fc receptor. Our data call for a reappraisal of the CTLA-4 checkpoint blockade hypothesis and provide new insights for the next generation of safe and effective anti-CTLA-4 mAbs.
Collapse
Affiliation(s)
- Xuexiang Du
- Center for Cancer and Immunology Research, Children's Research Institute, Children's National Health System, Washington, DC, 20010, USA
| | - Fei Tang
- Center for Cancer and Immunology Research, Children's Research Institute, Children's National Health System, Washington, DC, 20010, USA
| | - Mingyue Liu
- Center for Cancer and Immunology Research, Children's Research Institute, Children's National Health System, Washington, DC, 20010, USA
| | - Juanjuan Su
- Center for Cancer and Immunology Research, Children's Research Institute, Children's National Health System, Washington, DC, 20010, USA
| | - Yan Zhang
- Center for Cancer and Immunology Research, Children's Research Institute, Children's National Health System, Washington, DC, 20010, USA
| | - Wei Wu
- Center for Cancer and Immunology Research, Children's Research Institute, Children's National Health System, Washington, DC, 20010, USA
| | | | - Christopher A Lazarski
- Center for Cancer and Immunology Research, Children's Research Institute, Children's National Health System, Washington, DC, 20010, USA
| | - Peng Zhang
- Center for Cancer and Immunology Research, Children's Research Institute, Children's National Health System, Washington, DC, 20010, USA
| | - Xu Wang
- Center for Cancer and Immunology Research, Children's Research Institute, Children's National Health System, Washington, DC, 20010, USA
| | - Peiying Ye
- Center for Cancer and Immunology Research, Children's Research Institute, Children's National Health System, Washington, DC, 20010, USA
| | | | - Eugene Hwang
- Center for Cancer and Immunology Research, Children's Research Institute, Children's National Health System, Washington, DC, 20010, USA
| | - Tinghui Zhu
- Alphamab, Inc., Suzhou, Jiangsu, 215125, China
| | - Ting Xu
- Alphamab, Inc., Suzhou, Jiangsu, 215125, China
| | - Pan Zheng
- Center for Cancer and Immunology Research, Children's Research Institute, Children's National Health System, Washington, DC, 20010, USA. .,OncoImmune, Inc., Rockville, MD, 20852, USA.
| | - Yang Liu
- Center for Cancer and Immunology Research, Children's Research Institute, Children's National Health System, Washington, DC, 20010, USA.,OncoImmune, Inc., Rockville, MD, 20852, USA
| |
Collapse
|
3
|
Hawley TS, Linsley PS, Hawley RG. Co-expression of B7–1 with Interleukin-12 Enhances Vaccine-induced Antitumour Immunity in Experimental Myeloma. Hematology 2016; 3:365-74. [DOI: 10.1080/10245332.1998.11746410] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Affiliation(s)
- Teresa S. Hawley
- Oncology Gene Therapy Program, The Toronto Hospital, Toronto, Ontario, Canada
| | - Peter S. Linsley
- Bristol-Meyers Squibb Pharmaceutical Research Institute, Seattle, Washington, USA
| | - Robert G. Hawley
- Oncology Gene Therapy Program, The Toronto Hospital, Toronto, Ontario, Canada
| |
Collapse
|
4
|
Mazzocco M, Martini M, Rosato A, Stefani E, Matucci A, Dalla Santa S, De Sanctis F, Ugel S, Sandri S, Ferrarini G, Cestari T, Ferrari S, Zanovello P, Bronte V, Sartoris S. Autologous cellular vaccine overcomes cancer immunoediting in a mouse model of myeloma. Immunology 2015; 146:33-49. [PMID: 25959091 PMCID: PMC4552499 DOI: 10.1111/imm.12477] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Revised: 04/08/2015] [Accepted: 04/27/2015] [Indexed: 12/16/2022] Open
Abstract
In the Sp6 mouse plasmacytoma model, a whole-cell vaccination with Sp6 cells expressing de novo B7-1 (Sp6/B7) induced anatomically localized and cytotoxic T cell (CTL)-mediated protection against wild-type (WT) Sp6. Both WT Sp6 and Sp6/B7 showed down-regulated expression of MHC H-2 L(d). Increase of H-2 L(d) expression by cDNA transfection (Sp6/B7/L(d)) raised tumour immune protection and shifted most CTL responses towards H-2 L(d)-restricted antigenic epitopes. The tumour-protective responses were not specific for the H-2 L(d)-restricted immunodominant AH1 epitope of the gp70 common mouse tumour antigen, although WT Sp6 and transfectants were able to present it to specific T cells in vitro. Gp70 transcripts, absent in secondary lymphoid organs of naive mice, were detected in immunized mice as well as in splenocytes from naive mice incubated in vitro with supernatants of CTL-lysed Sp6 cell cultures, containing damage-associated molecular patterns (DAMPs). It has been shown that Toll-like receptor triggering induces gp70 expression. Damage-associated molecular patterns are released by CTL-mediated killing of Sp6/B7-Sp6/B7/L(d) cells migrated to draining lymph nodes during immunization and may activate gp70 expression and presentation in most resident antigen-presenting cells. The same could also apply for Mus musculus endogenous ecotropic murine leukaemia virus 1 particles present in Sp6-cytosol, discharged by dying cells and superinfecting antigen-presenting cells. The outcome of such a massive gp70 cross-presentation would probably be tolerogenic for the high-affinity AH1-gp70-specific CTL clones. In this scenario, autologous whole-tumour-cell vaccines rescue tumour-specific immunoprotection by amplification of subdominant tumour antigen responses when those against the immune dominant antigens are lost.
Collapse
Affiliation(s)
- Marta Mazzocco
- Department of Pathology and Diagnostics, University of Verona, Verona, Italy
| | - Matteo Martini
- Department of Pathology and Diagnostics, University of Verona, Verona, Italy
| | - Antonio Rosato
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy.,Veneto Institute of Oncology IOV - IRCCS, Padua, Italy
| | - Elisabetta Stefani
- Department of Pathology and Diagnostics, University of Verona, Verona, Italy
| | - Andrea Matucci
- Department of Pathology and Diagnostics, University of Verona, Verona, Italy
| | | | | | - Stefano Ugel
- Department of Pathology and Diagnostics, University of Verona, Verona, Italy
| | - Sara Sandri
- Department of Pathology and Diagnostics, University of Verona, Verona, Italy
| | - Giovanna Ferrarini
- Department of Pathology and Diagnostics, University of Verona, Verona, Italy
| | - Tiziana Cestari
- Department of Pathology and Diagnostics, University of Verona, Verona, Italy
| | - Sergio Ferrari
- Department of Neurological and Movement Sciences, University of Verona, Verona, Italy
| | - Paola Zanovello
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy.,Veneto Institute of Oncology IOV - IRCCS, Padua, Italy
| | - Vincenzo Bronte
- Department of Pathology and Diagnostics, University of Verona, Verona, Italy
| | - Silvia Sartoris
- Department of Pathology and Diagnostics, University of Verona, Verona, Italy
| |
Collapse
|
5
|
Low dose decitabine treatment induces CD80 expression in cancer cells and stimulates tumor specific cytotoxic T lymphocyte responses. PLoS One 2013; 8:e62924. [PMID: 23671644 PMCID: PMC3650049 DOI: 10.1371/journal.pone.0062924] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2012] [Accepted: 03/26/2013] [Indexed: 12/14/2022] Open
Abstract
Lack of immunogenicity of cancer cells has been considered a major reason for their failure in induction of a tumor specific T cell response. In this paper, we present evidence that decitabine (DAC), a DNA methylation inhibitor that is currently used for the treatment of myelodysplastic syndrome (MDS), acute myeloid leukemia (AML) and other malignant neoplasms, is capable of eliciting an anti-tumor cytotoxic T lymphocyte (CTL) response in mouse EL4 tumor model. C57BL/6 mice with established EL4 tumors were treated with DAC (1.0 mg/kg body weight) once daily for 5 days. We found that DAC treatment resulted in infiltration of IFN-γ producing T lymphocytes into tumors and caused tumor rejection. Depletion of CD8+, but not CD4+ T cells resumed tumor growth. DAC-induced CTL response appeared to be elicited by the induction of CD80 expression on tumor cells. Epigenetic evidence suggests that DAC induces CD80 expression in EL4 cells via demethylation of CpG dinucleotide sites in the promoter of CD80 gene. In addition, we also showed that a transient, low-dose DAC treatment can induce CD80 gene expression in a variety of human cancer cells. This study provides the first evidence that epigenetic modulation can induce the expression of a major T cell co-stimulatory molecule on cancer cells, which can overcome immune tolerance, and induce an efficient anti-tumor CTL response. The results have important implications in designing DAC-based cancer immunotherapy.
Collapse
|
6
|
Collins SA, Buhles A, Scallan MF, Harrison PT, O'Hanlon DM, O'Sullivan GC, Tangney M. AAV2-mediated in vivo immune gene therapy of solid tumours. GENETIC VACCINES AND THERAPY 2010; 8:8. [PMID: 21172020 PMCID: PMC3016353 DOI: 10.1186/1479-0556-8-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/14/2010] [Accepted: 12/20/2010] [Indexed: 11/10/2022]
Abstract
Background Many strategies have been adopted to unleash the potential of gene therapy for cancer, involving a wide range of therapeutic genes delivered by various methods. Immune therapy has become one of the major strategies adopted for cancer gene therapy and seeks to stimulate the immune system to target tumour antigens. In this study, the feasibility of AAV2 mediated immunotherapy of growing tumours was examined, in isolation and combined with anti-angiogenic therapy. Methods Immune-competent Balb/C or C57 mice bearing subcutaneous JBS fibrosarcoma or Lewis Lung Carcinoma (LLC) tumour xenografts respectively were treated by intra-tumoural administration of AAV2 vector encoding the immune up-regulating cytokine granulocyte macrophage-colony stimulating factor (GM-CSF) and the co-stimulatory molecule B7-1 to subcutaneous tumours, either alone or in combination with intra-muscular (IM) delivery of AAV2 vector encoding Nk4 14 days prior to tumour induction. Tumour growth and survival was monitored for all animals. Cured animals were re-challenged with tumourigenic doses of the original tumour type. In vivo cytotoxicity assays were used to investigate establishment of cell-mediated responses in treated animals. Results AAV2-mediated GM-CSF, B7-1 treatment resulted in a significant reduction in tumour growth and an increase in survival in both tumour models. Cured animals were resistant to re-challenge, and induction of T cell mediated anti-tumour responses were demonstrated. Adoptive transfer of splenocytes to naïve animals prevented tumour establishment. Systemic production of Nk4 induced by intra-muscular (IM) delivery of Nk4 significantly reduced subcutaneous tumour growth. However, combination of Nk4 treatment with GM-CSF, B7-1 therapy reduced the efficacy of the immune therapy. Conclusions Overall, this study demonstrates the potential for in vivo AAV2 mediated immune gene therapy, and provides data on the inter-relationship between tumour vasculature and immune cell recruitment.
Collapse
Affiliation(s)
- Sara A Collins
- Cork Cancer Research Centre, Mercy University Hospital and Leslie C, Quick Jnr, Laboratory, University College Cork, Cork, Ireland.
| | | | | | | | | | | | | |
Collapse
|
7
|
Tumor cells engineered to codisplay on their surface 4-1BBL and LIGHT costimulatory proteins as a novel vaccine approach for cancer immunotherapy. Cancer Gene Ther 2010; 17:730-41. [PMID: 20559332 PMCID: PMC2941532 DOI: 10.1038/cgt.2010.29] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Primary tumor cells genetically modified to express on their surface a collection of immunological ligands may have utility as therapeutic autologous cancer vaccines. However, genetic modification of primary tumor cells is not only cost, labor, and time intensive, but also has safety repercussions. As an alternative, we developed the ProtEx™ technology that involves generation of immunological ligands with core streptavidin (SA) and their display on biotinylated cells in a rapid and efficient manner. We herein demonstrate that TC-1 tumor cells can be rapidly and efficiently engineered to codisplay on their surface two costimulatory proteins, SA-4-1BBL and SA-LIGHT, simultaneously. Vaccination with irradiated TC-1 cells codisplaying both chimeric proteins showed 100% efficacy in a prophylactic and > 55% efficacy in a therapeutic tumor setting. In contrast, vaccination with TC-1 cells engineered with either protein alone showed significantly reduced efficacy in the prophylactic setting. Vaccine efficacy was associated with the generation of primary and memory T cell and antibody responses against the tumor without detectable signs of autoimmunity. Engineering tumor cells in a rapid and effective manner to simultaneously display on their surface a collection of immunostimulatory proteins with additive/synergistic functions presents a novel alternative approach to gene therapy with considerable potential for cancer immunotherapy.
Collapse
|
8
|
Li XL, Zhang D, Knight D, Odaka Y, Glass J, Mathis JM, Zhang QJ. Priming of immune responses against transporter associated with antigen processing (TAP)-deficient tumours: tumour direct priming. Immunology 2010; 128:420-8. [PMID: 20067541 DOI: 10.1111/j.1365-2567.2009.03127.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
We previously showed that introduction of transporter associated with antigen processing (TAP) 1 into TAP-negative CMT.64, a major histocompatibility complex class I (MHC-I) down-regulated mouse lung carcinoma cell line, enhanced T-cell immunity against TAP-deficient tumour cells. Here, we have addressed two questions: (1) whether such immunity can be further augmented by co-expression of TAP1 with B7.1 or H-2K(b) genes, and (2) which T-cell priming mechanism (tumour direct priming or dendritic cell cross-priming) plays the major role in inducing an immune response against TAP-deficient tumours. We introduced the B7.1 or H-2K(b) gene into TAP1-expressing CMT.64 cells and determined which gene co-expressed with TAP1 was able to provide greater protective immunity against TAP-deficient tumour cells. Our results show that immunization of mice with B7.1 and TAP1 co-expressing but not H-2K(b) and TAP1 co-expressing CMT.64 cells dramatically augments T-cell-mediated immunity, as shown by an increase in survival of mice inoculated with live CMT.64 cells. In addition, our results suggest that induction of T-cell-mediated immunity against TAP-deficient tumour cells could be mainly through tumour direct priming rather than dendritic cell cross-priming as they show that T cells generated by tumour cell-lysate-loaded dendritic cells recognized TAP-deficient tumour cells much less than TAP-proficient tumour cells. These data suggest that direct priming by TAP1 and B7.1 co-expressing tumour cells is potentially a major mechanism to facilitate immune responses against TAP-deficient tumour cells.
Collapse
Affiliation(s)
- Xiao-Lin Li
- Department of Cellular Biology and Anatomy, Gene Therapy Program, Louisiana State University Health Sciences Center, Kings Hwy, Shreveport, LA 71130, USA
| | | | | | | | | | | | | |
Collapse
|
9
|
Li XL, Liu YY, Knight D, Odaka Y, Mathis JM, Shi R, Glass J, Zhang QJ. Effect of B7.1 costimulation on T-cell based immunity against TAP-negative cancer can be facilitated by TAP1 expression. PLoS One 2009; 4:e6385. [PMID: 19629186 PMCID: PMC2711302 DOI: 10.1371/journal.pone.0006385] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2009] [Accepted: 06/18/2009] [Indexed: 11/28/2022] Open
Abstract
Tumors deficient in expression of the transporter associated with antigen processing (TAP) usually fail to induce T-cell-mediated immunity and are resistant to T-cell lysis. However, we have found that introduction of the B7.1 gene into TAP-negative (TAP−) or TAP1-transfected (TAP1+) murine lung carcinoma CMT.64 cells can augment the capacity of the cells to induce a protective immune response against wild-type tumor cells. Differences in the strength of the protective immune responses were observed between TAP− and TAP1+ B7.1 expressing CMT.64 cells depending on the doses of γ-irradiated cell immunization. While mice immunized with either high or low dose of B7.1-expressing TAP1+ cells rejected TAP− tumors, only high dose immunization with B7.1-expressing TAP− cells resulted in tumor rejection. The induced protective immunity was T-cell dependent as demonstrated by dramatically reduced antitumor immunity in mice depleted of CD8 or CD4 cells. Augmentation of T-cell mediated immune response against TAP− tumor cells was also observed in a virally infected tumor cell system. When mice were immunized with a high dose of γ-irradiated CMT.64 cells infected with vaccinia viruses carrying B7.1 and/or TAP1 genes, we found that the cells co-expressing B7.1 and TAP1, but not those expressing B7.1 alone, induced protective immunity against CMT.64 cells. In addition, inoculation with live tumor cells transfected with several different gene(s) revealed that only B7.1- and TAP1-coexpressing tumor cells significantly decreased tumorigenicity. These results indicate that B7.1-provoked antitumor immunity against TAP− cancer is facilitated by TAP1-expression, and thus both genes should be considered for cancer therapy in the future.
Collapse
Affiliation(s)
- Xiao-Lin Li
- Department of Cellular Biology and Anatomy, Gene Therapy Program, Feist-Weiller Cancer Center, Louisiana State University Health Sciences Center, Shreveport, Louisiana, United States of America
| | - Yong-Yu Liu
- College of Pharmacy, Basic Pharmaceutical Sciences, University of Louisiana, Monroe, Louisiana, United States of America
| | - David Knight
- Department of Cellular Biology and Anatomy, Gene Therapy Program, Feist-Weiller Cancer Center, Louisiana State University Health Sciences Center, Shreveport, Louisiana, United States of America
| | - Yoshinobu Odaka
- Department of Cellular Biology and Anatomy, Gene Therapy Program, Feist-Weiller Cancer Center, Louisiana State University Health Sciences Center, Shreveport, Louisiana, United States of America
| | - J. Michael Mathis
- Department of Cellular Biology and Anatomy, Gene Therapy Program, Feist-Weiller Cancer Center, Louisiana State University Health Sciences Center, Shreveport, Louisiana, United States of America
| | - Runhua Shi
- Feist-Weiller Cancer Center, Louisiana State University Health Sciences Center, Shreveport, Louisiana, United States of America
| | - Jonathan Glass
- Feist-Weiller Cancer Center, Louisiana State University Health Sciences Center, Shreveport, Louisiana, United States of America
| | - Qian-Jin Zhang
- Department of Cellular Biology and Anatomy, Gene Therapy Program, Feist-Weiller Cancer Center, Louisiana State University Health Sciences Center, Shreveport, Louisiana, United States of America
- * E-mail:
| |
Collapse
|
10
|
Gao JQ, Tsuda Y, Han M, Xu DH, Kanagawa N, Hatanaka Y, Tani Y, Mizuguchi H, Tsutsumi Y, Mayumi T, Okada N, Nakagawa S. NK cells are migrated and indispensable in the anti-tumor activity induced by CCL27 gene therapy. Cancer Immunol Immunother 2009; 58:291-9. [PMID: 18629495 PMCID: PMC11030262 DOI: 10.1007/s00262-008-0554-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2008] [Accepted: 06/23/2008] [Indexed: 10/21/2022]
Abstract
Natural killer (NK) cells have been demonstrated could play an important role in the treatment of a number of tumors in mice. In the present study, chemokine CCL27, which be considered only selectively chemoattracts cutaneous lymphocyte antigen positive memory T cells and Langerhans cells, firstly demonstrated that it could induce the accumulation of NK cells into tumor by the intratumoral injection of CCL27-encoding fiber-mutant vector, AdRGD-CCL27. Experiments using spleen cell fractionation and RT-PCR showed CCL27 receptor, mCCR10, was strongly expressed in NK cells, suggesting the accumulation of NK cells in tumor was attributed to chemoattractant activity of CCL27 itself. Moreover, the combination of AdRGD-CCL27 and AdRGD-IL-12 induced the synergistic anti-tumor activity via NK-dependent manner and induced more NK cells infiltration into tumor nodule than that induced by AdRGD-CCL27 alone or AdRGD-IL-12 alone.
Collapse
Affiliation(s)
- Jian-Qing Gao
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, 388 Yuhangtang Road, Hangzhou, Zhejiang 310058 China
| | - Yasuhiro Tsuda
- Department of Biotechnology and Therapeutics, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871 Japan
| | - Min Han
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, 388 Yuhangtang Road, Hangzhou, Zhejiang 310058 China
| | - Dong-Hang Xu
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, 388 Yuhangtang Road, Hangzhou, Zhejiang 310058 China
| | - Naoko Kanagawa
- Department of Biotechnology and Therapeutics, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871 Japan
| | - Yutaka Hatanaka
- Department of Biomedical Science, Dako Japan Co., Ltd, Nishinotouin-higashiiru, Shijo-dori, Shimogyo-ku, Kyoto, Japan
| | - Yoichi Tani
- Department of Biomedical Science, Dako Japan Co., Ltd, Nishinotouin-higashiiru, Shijo-dori, Shimogyo-ku, Kyoto, Japan
| | - Hiroyuki Mizuguchi
- National Institute of Biomedical Innovation, Saito-Asagi, Ibaraki, Osaka, Japan
| | - Yasuo Tsutsumi
- National Institute of Biomedical Innovation, Saito-Asagi, Ibaraki, Osaka, Japan
| | | | - Naoki Okada
- Department of Biotechnology and Therapeutics, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871 Japan
| | - Shinsaku Nakagawa
- Department of Biotechnology and Therapeutics, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871 Japan
| |
Collapse
|
11
|
Modulation of NKT cell development by B7-CD28 interaction: an expanding horizon for costimulation. PLoS One 2008; 3:e2703. [PMID: 18628995 PMCID: PMC2442875 DOI: 10.1371/journal.pone.0002703] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2008] [Accepted: 06/19/2008] [Indexed: 11/19/2022] Open
Abstract
It has been demonstrated that the development of NKT cells requires CD1d. The contribution of costimulatory molecules in this process has not been studied. Here we show that in mice with targeted mutations of B7-1/2 and CD28, the TCRbeta(+)alpha-Galcer/CD1d(+) (iValpha14 NKT) subset is significantly reduced in the thymus, spleen and liver. This is mainly due to decreased cell proliferation; although increased cell death in the thymi of CD28-deficient mice was also observed. Moreover, in the B7-1/2- and CD28-deficient mice, we found a decreased percentage of the CD4(-)NK1.1(+) subset and a correspondingly increased portion of the CD4(+)NK1.1(-) subset. In addition, the mice with a targeted mutation of either B7 or CD28 had a reduced susceptibility to Con A induced hepatitis, which is known to be mediated by NKT cells. Our results demonstrate that the development, maturation and function of NKT cell are modulated by the costimulatory pathway and thus expand the horizon of costimulation into NKT, which is widely viewed as a bridge between innate and adaptive immunity. As such, costimulation may modulate all major branches of cell-mediated immunity, including T cells, NK cells and NKT cells.
Collapse
|
12
|
Mir MA, Agrewala JN. Signaling through CD80: an approach for treating lymphomas. Expert Opin Ther Targets 2008; 12:969-79. [DOI: 10.1517/14728222.12.8.969] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
13
|
A two-pronged mechanism for HIV-1 Nef-mediated endocytosis of immune costimulatory molecules CD80 and CD86. Cell Host Microbe 2008; 1:37-49. [PMID: 18005680 DOI: 10.1016/j.chom.2007.01.001] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2006] [Revised: 11/21/2006] [Accepted: 01/18/2007] [Indexed: 12/14/2022]
Abstract
The Nef protein of HIV-1 mediates immune evasion by relocating major histocompatibility complex (MHC) molecules and the immune costimulatory molecules CD80 and CD86 away from the monocytic cell surface. We describe a two-pronged mechanism by which Nef removes CD80 and CD86 from the cell surface. While MHCI, CD80, and CD86 are all internalized via a dynamin-independent pathway, the endocytic mechanism used for costimulatory molecules is distinct from MHCI relocation. Nef expression results in the activation and actin-dependent translocation of Src kinase to the cell periphery. At the cell surface, Src promotes Rac activation via TIAM, a guanine nucleotide exchange factor for Rac. Nef also binds to the cytosolic tails of CD80 and CD86, triggering their endocytosis via Rac-based actin polymerization. These data reveal the use of an unusual molecular mechanism triggered in the host cell by HIV to affect its viral immune evasion strategy and suggest approaches for its subversion.
Collapse
|
14
|
Cashman J, Larkin J, Casey G, Whelan M, Collins C, Aarons S, Tangney M, O’Sullivan G. Immune gene therapy as a neoadjuvant to surgical excision to control metastatic cancers. Cancer Lett 2008; 262:94-102. [DOI: 10.1016/j.canlet.2007.11.042] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2007] [Revised: 11/25/2007] [Accepted: 11/26/2007] [Indexed: 11/29/2022]
|
15
|
Ruybal P, Gravisaco MJ, Barcala V, Escalada A, Di Sciullo P, Waldner C, Mongini C. Complete rejection of a T-cell lymphoma due to synergism of T-cell receptor costimulatory molecules, CD80, CD40L, and CD40. Vaccine 2007; 26:697-705. [PMID: 18155328 DOI: 10.1016/j.vaccine.2007.11.041] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2007] [Revised: 10/20/2007] [Accepted: 11/16/2007] [Indexed: 11/17/2022]
Abstract
The equal importance of the qualitative and quantitative characteristics of antigen presentation as well as the set of costimulatory signals provided by antigen presenting cells to T-cells in determining the outcome of T-cell responses at the time of antigen recognition is now clear. Moreover, an important function in innate mechanisms has been recently attributed to costimulatory molecules demonstrating their relevant role in different stages of immune response. In this paper, we demonstrated the ability of CD40L (CD154) and CD80 costimulatory molecules expression in a T-cell lymphoma to induce both T-cell dependent and independent immune responses leading to an important anti-tumor effect. CD40 expression by LBC cells enhanced only T-cell dependent anti-tumor immune response resulting in tumor rejection. Furthermore, this work represents the first report to describe complete tumor rejection after co-inoculation of lymphoma cells transfected with CD40L and CD80 in either presence or absence of CD40 expressing lymphoma cells. In addition, this synergistic effect resulted in long lasting immunity to parental tumor cells. Co-inoculation of tumor cells each genetically modified to express a different costimulatory molecule circumvents the need to co-transfect genetically unstable tumor cells and represents an option for those weakly or non-immunogenic tumors where either treatment alone proved to be inefficient. This strategy represents a promising approach for inducing anti-tumor immunity and provides a new rational design of cancer therapies.
Collapse
Affiliation(s)
- Paula Ruybal
- Laboratorio de Inmunología Celular y Molecular, Centro de Estudios Farmacológicos y Botánicos, CEFyBO-CONICET, Universidad de Buenos Aires, Facultad de Medicina, Paraguay 2155, 1121 Buenos Aires, Argentina
| | | | | | | | | | | | | |
Collapse
|
16
|
Anderson RCE, Anderson DE, Elder JB, Brown MD, Mandigo CE, Parsa AT, Goodman RR, McKhann GM, Sisti MB, Bruce JN. Lack of B7 expression, not human leukocyte antigen expression, facilitates immune evasion by human malignant gliomas. Neurosurgery 2007; 60:1129-36; discussion 1136. [PMID: 17538388 DOI: 10.1227/01.neu.0000255460.91892.44] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVE Lack of human leukocyte antigens and costimulatory molecules have been suggested as mechanisms by which human malignant gliomas avoid immune recognition and elimination. METHODS Using quantitative multiparameter flow cytometric analysis, tumor cells from patients with glioblastoma multiforme (n = 18) were examined ex vivo for the expression of human leukocyte antigen Class I and II molecules and the costimulatory molecules B7-1 and B7-2. They were compared with reactive astrocytes from peritumoral brain metastases (n = 7), and astrocytes removed during nontumor surgery (n = 5). RESULTS In contrast to the vast majority of solid peripheral human tumors, these results demonstrate that glioblastoma multiforme frequently express both human leukocyte antigen Class I and II molecules. Like most solid peripheral tumors, glioblastoma multiforme tumor cells express few or no B7 costimulatory molecules. Functional assays using heterogeneous ex vivo tumor preparations or pure populations of ex vivo tumor cells and microglia obtained using fluorescence-activated cell sorting indicate that CD4+ T-cells are activated by tumor cells only in the presence of exogenous B7 costimulation (provided by addition of soluble agonist anti-CD28 monoclonal antibody). CONCLUSION Thus, in contrast to many solid peripheral tumors, failure to present tumor antigens is not a likely impediment to immunotherapeutic strategies against malignant gliomas. Rather, immunotherapeutic strategies need to overcome low levels of B7 costimulation.
Collapse
Affiliation(s)
- Richard C E Anderson
- Department of Neurological Surgery, The Neurological Institute, Columbia University College of Physicians and Surgeons, New York, New York 10032, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Hunter TB, Alsarraj M, Gladue RP, Bedian V, Antonia SJ. An Agonist Antibody Specific for CD40 Induces Dendritic Cell Maturation and Promotes Autologous Anti-tumour T-cell Responses in an In vitro Mixed Autologous Tumour Cell/Lymph Node Cell Model. Scand J Immunol 2007; 65:479-86. [PMID: 17444959 DOI: 10.1111/j.1365-3083.2007.01927.x] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
CD40-mediated interactions play an important role in the response to a variety of diseases, including cancer. Engagement of CD40 on antigen-presenting cells, namely dendritic cells (DC), by CD40L leads to maturation and up-regulation of co-stimulatory molecules B7.1 and B7.2 (CD80 and CD86). These molecules are requisite to subsequent antigen-specific activation of T cells. T-cell activation is a critical aspect of specific anti-tumour immune responses that have become the focus of a variety of cancer immunotherapy approaches. Clinical trials involving immunologic interventions have shown clinical responses confirming that the immune system can be harnessed for the treatment of cancer. However, the clinical response rate has been low, signifying the need for new immunotherapeutic strategies. To this end, an agonist antibody specific for CD40, CP-870,893, has been developed. A fully autologous mixed tumour cell/lymph node cell model was utilized to demonstrate that CP-870,893 promotes the responsiveness of lymph node-derived T cells to autologous tumour. Specifically, T cells from the tumour-draining lymph nodes are not responsive to autologous tumour cells; however, in the presence of CP-870,893, this unresponsiveness is reversed, as indicated by lymph node cell proliferation and cytokine secretion. Monocyte-derived DC treated with CP-870,893 consistently display a mature phenotype: up-regulation of CD80, CD83, CD86 and HLA-DR expression, increased Mip1alpha and IL-12 secretion, and the loss of exogenous antigen-presenting capability subsequent to treatment with the antibody. These data indicate that CP-870,893 binds to and activates DC, ultimately driving a specific anti-tumour T-cell response.
Collapse
Affiliation(s)
- T B Hunter
- Immunology and Immunotherapy Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | | | | | | | | |
Collapse
|
18
|
Chiang JY, Jang IK, Hodes R, Gu H. Ablation of Cbl-b provides protection against transplanted and spontaneous tumors. J Clin Invest 2007; 117:1029-36. [PMID: 17364027 PMCID: PMC1810570 DOI: 10.1172/jci29472] [Citation(s) in RCA: 92] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2006] [Accepted: 02/06/2007] [Indexed: 02/03/2023] Open
Abstract
A significant challenge to efforts aimed at inducing effective antitumor immune responses is that CD8(+) T cells, which play a prominent role in these responses, may be unable to respond to tumors that lack costimulatory signals and that are protected by an immune suppressive environment such as that mediated by TGF-beta produced by tumor cells themselves or by infiltrating Tregs, often resulting in tolerance or anergy of tumor-specific T cells. Here we show that the in vitro activation of Cblb(-/-) CD8(+) T cells does not depend on CD28 costimulation and is resistant to TGF-beta suppression. In vivo studies further demonstrated that Cblb(-/-) mice, but not WT controls, efficiently rejected inoculated E.G7 and EL4 lymphomas that did not express B7 ligands and that introduction of the Cblb(-/-) mutation into tumor-prone ataxia telangiectasia mutated-deficient mice markedly reduced the incidence of spontaneous thymic lymphomas. Immunohistological study showed that E.G7 tumors from Cblb(-/-) mice contained massively infiltrating CD8(+) T cells. Adoptive transfer of purified Cblb(-/-) CD8(+) T cells into E.G7 tumor-bearing mice led to efficient eradication of established tumors. Thus, our data indicate that ablation of Cbl-b can be an efficient strategy for eliciting immune responses against both inoculated and spontaneous tumors.
Collapse
Affiliation(s)
- Jeffrey Y. Chiang
- Experimental Immunology Branch, National Cancer Institute, NIH, Bethesda, Maryland, USA.
Department of Microbiology, Columbia University College of Physicians and Surgeons, New York, New York, USA.
National Institute on Aging, NIH, Bethesda, Maryland, USA
| | - Ihn Kyung Jang
- Experimental Immunology Branch, National Cancer Institute, NIH, Bethesda, Maryland, USA.
Department of Microbiology, Columbia University College of Physicians and Surgeons, New York, New York, USA.
National Institute on Aging, NIH, Bethesda, Maryland, USA
| | - Richard Hodes
- Experimental Immunology Branch, National Cancer Institute, NIH, Bethesda, Maryland, USA.
Department of Microbiology, Columbia University College of Physicians and Surgeons, New York, New York, USA.
National Institute on Aging, NIH, Bethesda, Maryland, USA
| | - Hua Gu
- Experimental Immunology Branch, National Cancer Institute, NIH, Bethesda, Maryland, USA.
Department of Microbiology, Columbia University College of Physicians and Surgeons, New York, New York, USA.
National Institute on Aging, NIH, Bethesda, Maryland, USA
| |
Collapse
|
19
|
Tangney M, Casey G, Larkin JO, Collins CG, Soden D, Cashman J, Whelan MC, O'Sullivan GC. Non-viral in vivo immune gene therapy of cancer: combined strategies for treatment of systemic disease. Cancer Immunol Immunother 2006; 55:1443-50. [PMID: 16612593 PMCID: PMC11031075 DOI: 10.1007/s00262-006-0169-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2006] [Accepted: 03/22/2006] [Indexed: 01/22/2023]
Abstract
Many patients with various types of cancers have already by the time of presentation, micrometastases in their tissues and are left after treatment in a minimal residual disease state [Am J Gastroenterol 95(12), 2000]. To prevent tumour recurrence these patients require a systemic based therapy, but current modalities are limited by toxicity or lack of efficacy. We have previously reported that immune reactivity to the primary tumour is an important regulator of micrometastases and determinant of prognosis. This suggests that recruitment of specific anti-tumour mechanisms within the primary tumour could be used advantageously for tumour control as either primary or neo-adjuvant treatments. Recently, we have focused on methods of stimulating immune eradication of solid tumours and minimal residual disease using gene therapy approaches. Gene therapy is now a realistic prospect and a number of delivery approaches have been explored, including the use of viral and non-viral vectors. Non-viral vectors have received significant attention since, in spite of their relative delivery inefficiency, they may be safer and have greater potential for delivery of larger genetic units. By in vivo electroporation of the primary tumour with plasmid expressing GM-CSF and B7-1, we aim to stimulate immune eradication of the treated tumour and associated metastases. In this symposium report, we describe an effective gene based approach for cancer immunotherapy by inducing cytokine and immune co-stimulatory molecule expression by the growing cells of the primary tumour using a plasmid electroporation gene delivery strategy. We discuss the potential for enhancement of this therapy by its application as a neoadjuvant to surgical excision and by its use in combination with suppressor T cell depletion.
Collapse
Affiliation(s)
- M Tangney
- Cork Cancer Research Centre, Mercy University Hospital and Leslie C. Quick Jnr. Laboratory, University College Cork, Cork, Ireland.
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Chen XY, Zhang W, Zhang W, Wu S, Bi F, Su YJ, Tan XY, Liu JN, Zhang J. Vaccination with Viable Human Umbilical Vein Endothelial Cells Prevents Metastatic Tumors by Attack on Tumor Vasculature with Both Cellular and Humoral Immunity. Clin Cancer Res 2006; 12:5834-40. [PMID: 17020991 DOI: 10.1158/1078-0432.ccr-06-1105] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Because tumor endothelium is rarely targeted by immunity but is critically important for tumor growth, the immunity against tumor endothelium is to be developed as a novel antitumor strategy. EXPERIMENTAL DESIGN First, viable human umbilical vein endothelial cells (HUVEC) were immunized to C57BL/6 and BALB/c mice to evoke specific CTLs as well as antibodies against tumor endothelium. Lewis lung carcinoma or myeloma cells were subsequently inoculated to evaluate the effect on tumor growth by vaccination. Second, the effect on tumor metastasis by vaccination was studied using tumor-resected mice receiving HUVEC immunization 3 days after excision. Third, the immune sera and T lymphocytes from HUVEC-immunized mice were transferred to tumor-bearing mice and added to cultured HUVECs to investigate their antiproliferative effect. RESULTS Viable HUVEC immunization showed potent antitumor effects in Lewis lung carcinoma and myeloma tumor models. Both immune sera and CTL inhibited tumor growth and specifically suppressed proliferation of HUVECs. Particularly, tumors entirely disappeared on day 90 after tumor inoculation in four of six tumor-bearing mice receiving CTL therapy. In a metastatic tumor model, we found that the HUVEC vaccination prolonged life span from 30.9 to 41.5 days after tumor resection compared with PBS-treated mice without apparent side effects. CONCLUSIONS Vaccination with viable HUVECs evoked both humoral and cellular immunity against tumor microvasculature, and therefore significantly inhibited tumor growth and prolonged life span of tumor-resected mice. This may provide with a novel treatment for metastatic tumors. Moreover, we have established a convenient method to evoke specific CTL against tumor angiogenesis.
Collapse
MESH Headings
- Animals
- Antibody Formation
- Antineoplastic Agents, Hormonal/pharmacology
- Carcinoma, Lewis Lung/blood supply
- Carcinoma, Lewis Lung/immunology
- Carcinoma, Lewis Lung/prevention & control
- Endothelium, Vascular/immunology
- Humans
- Immunity, Cellular
- Lung Neoplasms/blood supply
- Lung Neoplasms/immunology
- Lung Neoplasms/prevention & control
- Male
- Melanoma, Experimental/blood supply
- Melanoma, Experimental/immunology
- Melanoma, Experimental/prevention & control
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Nude
- Neoplasm Metastasis
- Neovascularization, Pathologic/immunology
- Survival Rate
- T-Lymphocytes/immunology
- Umbilical Veins/cytology
- Vaccination
- Vascular Endothelial Growth Factor A/metabolism
Collapse
Affiliation(s)
- Xin-Yuan Chen
- Institute of Molecular Medicine and State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, 22 Hankou Road, Nanjing 210093, China
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Collins CG, Tangney M, Larkin JO, Casey G, Whelan MC, Cashman J, Murphy J, Soden D, Vejda S, McKenna S, Kiely B, Collins JK, Barrett J, Aarons S, O'Sullivan GC. Local gene therapy of solid tumors with GM-CSF and B7-1 eradicates both treated and distal tumors. Cancer Gene Ther 2006; 13:1061-71. [PMID: 16874363 DOI: 10.1038/sj.cgt.7700976] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Gene therapy-induced expression of immunostimulatory molecules at tumor cell level may evoke antitumor immune mechanisms by recruiting and enhancing viability of antigen-processing cells and specific tumoricidal lymphocytes. The antitumor efficacy of a plasmid, coding for granulocyte-macrophage colony-stimulating factor (GM-CSF) and the B7-1 costimulatory immune molecule, delivered into growing solid tumors by electroporation was investigated. Murine fibrosarcomas (JBS) growing in Balb/C mice (<or=100 mm3) were transfected with GM-CSF/B7-1-expressing plasmid. Complete tumor regression occurred in greater than 60% of treated animals. This response was systemic, durable and tumor specific, with all responding animals resistant to repeat tumor challenge. Using a liver metastatic model, effective cure of distal metastases was achieved following treatment of the primary subcutaneous tumor. This treatment strategy could be applicable in the clinical setting for effective elimination of both primary tumors and associated metastatic disease.
Collapse
Affiliation(s)
- C G Collins
- Cork Cancer Research Centre, Mercy University Hospital and Leslie C Quick Jnr Laboratory, University College Cork, Cork, Ireland
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Blaser BW, Caligiuri MA. Autologous immune strategies to reduce the risk of leukemic relapse: Consideration for IL-15. Best Pract Res Clin Haematol 2006; 19:281-92. [PMID: 16516125 DOI: 10.1016/j.beha.2005.11.004] [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: 01/25/2023]
Abstract
The graft-versus-leukemia effect following allogeneic stem cell transplantation (SCT) reduces the incidence of leukemic relapse and establishes that effector cells can eliminate or at least contain resistant leukemic stem cells. Natural killer cells also appear to play a role in directly lowering the rate of relapse following allogeneic SCT in patients with acute myeloid leukemia. To date, however, effective prevention of leukemic relapse by autologous immune effector cells has not been demonstrated. This article examines some of the challenges that limit autologous antileukemia immunity as well as some possible immunotherapeutic approaches that may help control leukemic relapse following autologous SCT.
Collapse
Affiliation(s)
- Bradley W Blaser
- The Ohio State University Medical Center, The Ohio State University, 458 A Starling Loving Hall, 320 W 10th Avenue, Columbus, OH 43210-1214, USA
| | | |
Collapse
|
23
|
Hao S, Bi X, Su L, Dong W, Moyana T, Xiang J. Molecular and immunophenotypical characterization of progressive and regressive leukemia cell lines. Cancer Biother Radiopharm 2005; 20:290-9. [PMID: 15989474 DOI: 10.1089/cbr.2005.20.290] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The P815 and P198 cell lines are clonally related mouse mastocytoma cell lines. They differ in their biologic behavior in that P815 is a progressive tumor cell line, whereas P198 is a regressive one. These cell lines have been extensively used as models for the study of tumor-host relationships and tumor immunology. Although some of their biological properties have been well documented, the molecular mechanisms underlying tumor progression or regression have not been completely elucidated. In this study, we characterized the growth behavior and immunophenotype of these two cell lines, and analyzed their gene profiles using a complementary deoxynucleic acid (cDNA) microarray composed of 514 immunologically relevant genes. Our data showed that the two cell lines exhibited quite dissimilar and contrasting growth characteristics when inoculated into syngeneic mice. P815 tumors grew unremittingly, while P198 tumors gradually regressed. From a molecular viewpoint, P815 cells showed a higher expression of genes promoting tumor growth, such as IGF-1, IL-8R, FGFR1, VEGF-A, and VEGF-B. On the other hand, P198 tumor cells expressed CD11b and CD80, which favor the recruitment of lymphocytes and antigen-presenting cells (APCs), as well as the elicitation of antitumor immunity. P198 tumor cells also depicted a higher expression of genes inhibiting tumor growth, such as TNF-alpha, SOCS-1, CIS1, 4-1BB, and GDF-10. In conclusion, our results contribute further information in the understanding of the molecular mechanisms associated with the regression and progression of P815 and P198 tumor cells.
Collapse
Affiliation(s)
- Siguo Hao
- Research Unit, Saskatchewan Cancer Agency, Department of Oncology, University of Saskatchewan, Saskatoon, Saskatchewan, Canada.
| | | | | | | | | | | |
Collapse
|
24
|
Hatfield P, Merrick A, Harrington K, Vile R, Bateman A, Selby P, Melcher A. Radiation-induced cell death and dendritic cells: potential for cancer immunotherapy? Clin Oncol (R Coll Radiol) 2005; 17:1-11. [PMID: 15714922 DOI: 10.1016/j.clon.2004.06.014] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Dendritic cells are key orchestrators of the immune system. There is considerable interest in their use for treating cancer. Whether they initiate an effective cytotoxic response against antigen-bearing cells, or produce tolerance, depends on the context in which those antigens are presented. Ionising radiation, and the cell death it causes, has several properties that may facilitate such an effective response. A range of in-vitro and in-vivo data supports this, although potential problems exist that may require concurrent strategies.
Collapse
Affiliation(s)
- P Hatfield
- Cancer Research UK Clinical Centre, St James's University Hospital, Leeds, UK.
| | | | | | | | | | | | | |
Collapse
|
25
|
Engineering high avidity CTLA4Ig for therapy of rheumatic diseases. Expert Opin Ther Pat 2005. [DOI: 10.1517/13543776.12.9.1455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
26
|
Zheng P, Liu Y. Co-stimulatory molecules B7-1 and B7-2 as experimental therapeutic targets. ACTA ACUST UNITED AC 2005. [DOI: 10.1517/14728222.3.1.93] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
27
|
Zheng X, Gao JX, Chang X, Wang Y, Liu Y, Wen J, Zhang H, Zhang J, Liu Y, Zheng P. B7-CD28 Interaction Promotes Proliferation and Survival but Suppresses Differentiation of CD4−CD8− T Cells in the Thymus. THE JOURNAL OF IMMUNOLOGY 2004; 173:2253-61. [PMID: 15294937 DOI: 10.4049/jimmunol.173.4.2253] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Costimulatory molecules play critical roles in the induction and effector function of T cells. More recent studies reveal that costimulatory molecules enhance clonal deletion of autoreactive T cells as well as generation and homeostasis of the CD25(+)CD4(+) regulatory T cells. However, it is unclear whether the costimulatory molecules play any role in the proliferation and differentiation of T cells before they acquire MHC-restricted TCR. In this study, we report that targeted mutations of B7-1 and B7-2 substantially reduce the proliferation and survival of CD4(-)CD8(-) (double-negative (DN)) T cells in the thymus. Perhaps as a result of reduced proliferation, the accumulation of RAG-2 protein in the DN thymocytes is increased in B7-deficient mice, which may explain the increased expression of TCR gene and accelerated transition of CD25(+)CD44(-) (DN3) to CD25(-)CD44(-) (DN4) stage. Qualitatively similar, but quantitatively less striking effects were observed in mice with a targeted mutation of CD28, but not CTLA4. Taken together, our results demonstrate that the development of DN in the thymus is subject to modulation by the B7-CD28 costimulatory pathway.
Collapse
Affiliation(s)
- Xincheng Zheng
- Division of Cancer Immunology, Department of Pathology, Ohio State University Medical Center and Comprehensive Cancer Center, Columbus, OH 43210, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Khan ANH, Magner WJ, Tomasi TB. An epigenetically altered tumor cell vaccine. Cancer Immunol Immunother 2004; 53:748-54. [PMID: 14997346 PMCID: PMC11032794 DOI: 10.1007/s00262-004-0513-0] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2003] [Accepted: 01/09/2004] [Indexed: 11/27/2022]
Abstract
Functional inactivation of genes critical to immunity may occur by mutation and/or by repression, the latter being potentially reversible with agents that modify chromatin. This study was constructed to determine whether reversal of gene silencing, by altering the acetylation status of chromatin, might lead to an effective tumor vaccine. We show that the expression of selected genes important to tumor immunity, including MHC class II, CD40, and B7-1/2 are altered by treating tumor cells in vitro with a histone deacetylase inhibitor, trichostatin A (TSA). Tumor cells treated in vitro with TSA showed delayed onset and rate of tumor growth in 70% of the J558 plasmacytoma and 100% of the B16 melanoma injected animals. Long-term tumor specific immunity was elicited to rechallenge with wild-type cells in approximately 30% in both tumor models. Splenic T cells from immune mice lysed untreated tumor cells, and SCID mice did not manifest immunity, suggesting that T cells may be involved in immunity. We hypothesize that repression of immune genes is involved in the evasion of immunity by tumors and suggest that epigenetically altered cancer cells should be further explored as a strategy for the induction of tumor immunity.
Collapse
MESH Headings
- Animals
- Antigens, CD/genetics
- Antigens, CD/metabolism
- B7-1 Antigen/genetics
- B7-1 Antigen/metabolism
- B7-2 Antigen
- CD40 Antigens/genetics
- CD40 Antigens/metabolism
- Cancer Vaccines/therapeutic use
- Cell Division/immunology
- Enzyme Inhibitors/therapeutic use
- Genes, MHC Class I/drug effects
- Genes, MHC Class II/drug effects
- Histocompatibility Antigens Class II/genetics
- Histocompatibility Antigens Class II/metabolism
- Histone Deacetylase Inhibitors
- Humans
- Hydroxamic Acids/therapeutic use
- Mammary Neoplasms, Experimental/immunology
- Mammary Neoplasms, Experimental/prevention & control
- Melanoma, Experimental/immunology
- Melanoma, Experimental/prevention & control
- Membrane Glycoproteins/genetics
- Membrane Glycoproteins/metabolism
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, SCID
- Plasmacytoma/immunology
- Plasmacytoma/prevention & control
- Spleen/immunology
- Survival Rate
- T-Lymphocytes, Cytotoxic/immunology
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- A. Nazmul H. Khan
- Laboratory of Molecular Medicine, Department of Immunology, Roswell Park Cancer Institute, Buffalo, NY 14263 USA
| | - William J. Magner
- Laboratory of Molecular Medicine, Department of Immunology, Roswell Park Cancer Institute, Buffalo, NY 14263 USA
- Department of Microbiology, School of Medicine and Biomedical Sciences, State University of New York, Buffalo, NY 14214 USA
| | - Thomas B. Tomasi
- Laboratory of Molecular Medicine, Department of Immunology, Roswell Park Cancer Institute, Buffalo, NY 14263 USA
- Department of Medicine, School of Medicine and Biomedical Sciences, State University of New York, Buffalo, NY 14214 USA
- Department of Microbiology, School of Medicine and Biomedical Sciences, State University of New York, Buffalo, NY 14214 USA
| |
Collapse
|
29
|
Sartoris S, Testi MG, Stefani E, Chignola R, Guerriero C, Matucci A, Cestari T, Scarpa A, Riviera AP, Zanoni G, Tridente G, Andrighetto G. Induction of an antitumour adaptive immune response elicited by tumour cells expressing de novo B7-1 mainly depends on the anatomical site of their delivery: the dose applied regulates the expansion of the response. Immunology 2004; 110:474-81. [PMID: 14632645 PMCID: PMC1783082 DOI: 10.1111/j.1365-2567.2003.01760.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
De novo expression of costimulatory molecules in tumours generally increases their immunogenicity, but does not always induce a protective response against the parental tumour. This issue was addressed in the mouse Sp6 hybridoma model, comparing different immunization routes (subcutaneous, intraperitoneal and intravenous) and doses (0.5 x 10(6) and 5 x 10(6) cells) of Sp6 cells expressing de novo B7-1 (Sp6/B7). The results can be summarized as follows. First, de novo expression of B7-1 rendered Sp6 immunogenic, as it significantly reduced the tumour incidence to < or =15% with all delivery routes and doses tested, whereas wild-type Sp6 was invariably tumorigenic (100% tumour incidence). Second, long-lasting protection against wild-type Sp6 was mainly achieved when immunization with Sp6/B7 was subcutaneous: a dose of 0.5 x 10(6) Sp6/B7 cells elicited protection that was confined to sites in the same anatomical quarter as the immunizing injection. Repeated injections of the same dose extended protection against wild-type Sp6 to other anatomical districts, as well as a single injection of a 10-fold higher dose (5 x 10(6) cells). Finally, Sp6-specific cytotoxic T-lymphocyte activity was detected in draining lymph nodes, and the splenic expansion of Sp6-specific cytotoxic T-lymphocyte precursors quantitatively correlated with the dose of antigen. We conclude that activation of a protective immune response against Sp6 depends on the local environment where the immunogenic form of the 'whole tumour cell antigen' is delivered. The antigen dose regulates the anatomical extent of the protective response.
Collapse
Affiliation(s)
- Silvia Sartoris
- Dipartimento di Patologia, Sezione di Immunologia, Università di Verona, Verona, Italy.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Gao JX, Liu X, Wen J, Caligiuri MA, Stroynowski I, Zheng P, Liu Y. Two-signal requirement for activation and effector function of natural killer cell response to allogeneic tumor cells. Blood 2003; 102:4456-63. [PMID: 12933569 DOI: 10.1182/blood-2003-07-2480] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Optimal activation of T cells requires delivery of both antigenic and costimulatory signals. It is unclear, however, if the function of the natural killer (NK) cells is also modulated by these 2 signals. Here we report that efficient control of solid allogeneic tumors by NK cells depends on codelivery of both B7-1 and major histocompatibility complex (MHC) class I on the tumor cells. The codelivery is required for optimal expansion and effector function of NK cells in response to both melanoma and plasmocytoma that expressed allogeneic MHC class I. Our results demonstrate that the 2 signals required for T-cell function also can regulate NK immunity and reveal an important similarity between the innate NK response and the adaptive T-cell response.
Collapse
MESH Headings
- ATP Binding Cassette Transporter, Subfamily B, Member 3
- ATP-Binding Cassette Transporters/genetics
- ATP-Binding Cassette Transporters/immunology
- Animals
- Antigens, Ly/analysis
- B7-1 Antigen/genetics
- B7-1 Antigen/immunology
- Cell Cycle
- Cell Line, Tumor/immunology
- Cytotoxicity, Immunologic
- H-2 Antigens/genetics
- H-2 Antigens/immunology
- Histocompatibility Antigen H-2D
- Killer Cells, Natural/classification
- Killer Cells, Natural/immunology
- Lectins, C-Type
- Lymphocyte Activation
- Melanoma, Experimental/immunology
- Melanoma, Experimental/pathology
- Mice
- Mice, Inbred C57BL
- Plasmacytoma/immunology
- Plasmacytoma/pathology
- Receptors, NK Cell Lectin-Like
- Recombinant Fusion Proteins/immunology
- Signal Transduction/immunology
- Transfection
Collapse
Affiliation(s)
- Jian-Xin Gao
- Department of Pathology and Comprehensive Cancer Center, Ohio State University Medical Center, 129 Hamilton Hall, 1645 Neil Ave, Columbus, OH 43210, USA
| | | | | | | | | | | | | |
Collapse
|
31
|
Liu X, Gao JX, Wen J, Yin L, Li O, Zuo T, Gajewski TF, Fu YX, Zheng P, Liu Y. B7DC/PDL2 promotes tumor immunity by a PD-1-independent mechanism. J Exp Med 2003; 197:1721-30. [PMID: 12810690 PMCID: PMC2193953 DOI: 10.1084/jem.20022089] [Citation(s) in RCA: 115] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
B7H1 (PDL1) and B7DC (PDL2) are two new members of the B7 family that can interact with PD-1, a putative negative regulator for immune function. Recent studies have provided evidence for inhibitory functions of both members via PD-1. Meanwhile, compelling evidence exists for costimulatory function of both members. Here we demonstrate that expression of B7DC on the tumor cells promotes CD8 T cell-mediated rejection of tumor cells, at both the induction and effector phase of antitumor immunity. Moreover, B7DC binds to PD-1(-/-) cells and enhances T cell killing in a PD-1-independent mechanism. Our results demonstrate a novel pathway for B7DC to promote tumor immunity and may reconcile the apparently contradictory findings on the function of B7DC.
Collapse
MESH Headings
- Adoptive Transfer
- Animals
- Antigens, Neoplasm/genetics
- Antigens, Neoplasm/immunology
- Antigens, Surface
- Apoptosis Regulatory Proteins
- B7-1 Antigen/genetics
- B7-1 Antigen/immunology
- B7-1 Antigen/metabolism
- B7-H1 Antigen
- Blood Proteins
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- CHO Cells
- Cricetinae
- Cytotoxicity, Immunologic
- Membrane Glycoproteins
- Mice
- Mice, Inbred BALB C
- Mice, Transgenic
- Neoplasm Transplantation
- Neoplasms, Experimental/immunology
- Peptides
- Programmed Cell Death 1 Ligand 2 Protein
- Programmed Cell Death 1 Receptor
- Proteins/immunology
- Proteins/metabolism
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/metabolism
- Spleen/cytology
- Spleen/immunology
- T-Lymphocytes, Cytotoxic/immunology
- Transgenes
- Transplantation Chimera
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- Xingluo Liu
- Department of Pathology, Ohio State University Medical Center, Columbus, OH 43210, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Abstract
Prostate cancer is the second leading cause of cancer death in the US, largely because of the limitations of our current therapeutic options, especially once the cancer has metastasized. Investigators have long sought new therapeutic modalities such as angiogenesis inhibitors, vaccines, and gene therapy, among others. It appears that a combination approach will be required to cure the majority of malignancies. Immunotherapy for prostate cancer appears feasible and a likely therapeutic modality in the armamentarium. Unfortunately, further research in basic immunology and the interaction of the immune system with other forms of therapy is needed. Many obstacles exist in immunotherapy, including vector design, tumouricidal specificity, and tumor evasion, which will have to be overcome in order to realize the maximum therapeutic benefit from this treatment modality.
Collapse
Affiliation(s)
- Joseph M Kaminski
- Department of Radiology, Medical College of Georgia, Augusta, GA 30912, USA
| | | | | | | | | |
Collapse
|
33
|
Bai XF, Liu J, Li O, Zheng P, Liu Y. Antigenic drift as a mechanism for tumor evasion of destruction by cytolytic T lymphocytes. J Clin Invest 2003; 111:1487-96. [PMID: 12750398 PMCID: PMC155049 DOI: 10.1172/jci17656] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
It is established that mutations in viral antigenic epitopes, or antigenic drifts, allow viruses to escape recognition by both Ab's and T lymphocytes. It is unclear, however, whether tumor cells can escape immune recognition via antigenic drift. Here we show that adoptive therapy with both monoclonal and polyclonal transgenic CTLs, specific for a natural tumor antigen, P1A, selects for multiple mutations in the P1A antigenic epitope. These mutations severely diminish T cell recognition of the tumor antigen by a variety of mechanisms, including modulation of MHC:peptide interaction and TCR binding to MHC:peptide complex. These results provide the first evidence for tumor evasion of T cell recognition by antigenic drift, and thus have important implications for the strategy of tumor immunotherapy.
Collapse
MESH Headings
- Animals
- Antigens, Neoplasm/genetics
- Antigens, Neoplasm/immunology
- Clone Cells/immunology
- Clone Cells/pathology
- DNA Mutational Analysis
- DNA-Binding Proteins/deficiency
- DNA-Binding Proteins/genetics
- Disease Models, Animal
- Epitopes/genetics
- Epitopes/immunology
- Genetic Drift
- Immunotherapy, Adoptive/adverse effects
- Major Histocompatibility Complex/immunology
- Mice
- Mice, Inbred BALB C
- Mice, Transgenic
- Neoplasm Transplantation
- Plasmacytoma/immunology
- Plasmacytoma/pathology
- Plasmacytoma/therapy
- Receptors, Antigen, T-Cell, alpha-beta/genetics
- Receptors, Antigen, T-Cell, alpha-beta/immunology
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Cytotoxic/transplantation
- Tumor Escape/genetics
- Tumor Escape/immunology
Collapse
Affiliation(s)
- Xue-Feng Bai
- Division of Cancer Immunology, Department of Pathology, Ohio State University Medical Center, Columbus, Ohio 43210, USA
| | | | | | | | | |
Collapse
|
34
|
Ochsenbein AF. Principles of tumor immunosurveillance and implications for immunotherapy. Cancer Gene Ther 2002; 9:1043-55. [PMID: 12522443 DOI: 10.1038/sj.cgt.7700540] [Citation(s) in RCA: 73] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2002] [Indexed: 01/24/2023]
Abstract
Although antigen loss variants, major histocompatibility (MHC) class I down-regulation, or the expression of inhibitory molecules may explain the failure of immunosurveillance against some tumors, this seems not to apply for many other solid peripheral or lymphohematopoietic tumors. Why then is immunosurveillance so ineffective and can it be improved? This review focuses on one important aspect of tumor immunity, namely the relevance of antigen dose and localization. Immune responses in vivo are induced in organized lymphoid tissues, i.e., in lymph nodes and spleen. The antigen dose that reaches secondary lymphoid organs over time is a crucial parameter that drives antiviral and antitumoral immune responses. Tumors use various strategies to prevent efficient presentation of their antigens in lymphoid organs. A major obstacle to the induction of an endogenous tumor-specific cytotoxic T lymphocyte (CTL) response is the inefficient presentation of tumor antigen on MHC class I molecules of professional antigen-presenting cells. Peripheral solid tumors that develop outside lymphoid organs are, therefore, often ignored by the immune system. In other situations, tumors - especially of lymphohematopoietic origin - may tolerize specific CTLs. Understanding tumor immunosurveillance is key to the design of efficient antitumor vaccines. Attempts to improve immunity to tumors include vaccination strategies to (a) provide the tumor antigen to secondary lymphoid organs using recombinant viruses or dendritic cells as carriers, (b) express costimulatory signals on tumor cells, or (c) improve the efficiency of cross-priming.
Collapse
|
35
|
Ohata J, Sakurai J, Saito K, Tani K, Asano S, Azuma M. Differential graft-versus-leukaemia effect by CD28 and CD40 co-stimulatory blockade after graft-versus-host disease prophylaxis. Clin Exp Immunol 2002; 129:61-8. [PMID: 12100023 PMCID: PMC1906426 DOI: 10.1046/j.1365-2249.2002.01857.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Co-stimulatory blockade may be a promising strategy for tolerance induction in transplantation. In allogeneic bone marrow transplantation (BMT) for leukaemia treatment, however, preservation of the graft-versus-leukaemia (GVL) effect is another critical requirement for clinical application. In this study, we have compared the effect on GVL of using CD28 and CD40 co-stimulatory blockades as graft-versus-host disease (GVHD) prophylaxis in a murine allogeneic BMT model with simultaneous transfer of BCL1 leukaemia. Despite the relative improvement of GVHD as assessed by survival and body weight in both treatment regimes, treatment with anti-CD154 moAb clearly diminished the GVL effect, whereas treatment with anti-CD80 and CD86 MoAbs maintained this effect. Although T cell-mediated effector function at 14 days post-BMT assessed by IFNgamma expression and cytotoxicity against host alloantigen was comparable between both co-stimulatory blockades, IL-12 mRNA expression was preferentially reduced by CD40 blockade. Our results suggest the differential involvement of the CD28 and CD40 co-stimulatory pathways in the development of GVHD and GVL effects. CD28 blockade may be a favourable strategy for tolerance induction in leukaemia patients undergoing BMT.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/immunology
- Antibodies, Monoclonal/pharmacology
- Antibodies, Monoclonal/therapeutic use
- Antibodies, Monoclonal/toxicity
- Antigen-Presenting Cells/immunology
- Antigens, CD/immunology
- B7-1 Antigen/immunology
- B7-2 Antigen
- Bone Marrow Transplantation/immunology
- CD28 Antigens/immunology
- CD40 Antigens/immunology
- CD40 Ligand/immunology
- Female
- Gene Expression Regulation/drug effects
- Graft vs Host Disease/prevention & control
- Graft vs Leukemia Effect/drug effects
- Graft vs Leukemia Effect/immunology
- Immune Tolerance
- Immunosuppressive Agents/immunology
- Immunosuppressive Agents/pharmacology
- Immunosuppressive Agents/therapeutic use
- Interferon-gamma/biosynthesis
- Interleukin-12/biosynthesis
- Killer Cells, Natural/immunology
- Leukemia, Experimental/immunology
- Leukemia, Experimental/therapy
- Lymphocyte Activation/drug effects
- Lymphocyte Activation/immunology
- Membrane Glycoproteins/immunology
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Nude
- RNA, Messenger/biosynthesis
- Radiation Chimera
- T-Lymphocyte Subsets/immunology
- Transplantation, Homologous/immunology
Collapse
Affiliation(s)
- J Ohata
- Department of Immunology, National Children's Medical Research Center, Tokyo, Japan
| | | | | | | | | | | |
Collapse
|
36
|
Bai XF, Liu J, May KF, Guo Y, Zheng P, Liu Y. B7-CTLA4 interaction promotes cognate destruction of tumor cells by cytotoxic T lymphocytes in vivo. Blood 2002; 99:2880-9. [PMID: 11929778 DOI: 10.1182/blood.v99.8.2880] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Costimulatory molecules B7-1 and B7-2 (hereby collectively called B7) interact with CD28 and CTLA4 on T cells and promote antitumor immunity. The function of B7-CTLA4 interaction in antitumor CTL response remains controversial. Here we used CD28(-/-) and CD28(+/-) or CD28(+/+) transgenic mice that express the T-cell receptor specific for an unmutated tumor antigen, P1A, and for tumor cells expressing a CTLA4-specific B7 mutant to evaluate the function of CD28-B7 and CTLA4-B7 interactions in induction and effector phases of antitumor immunity. We report that B7-CD28 and B7-CTLA4 interactions promote tumor rejection. However, this is achieved by distinct mechanisms. B7-CD28 interaction enhances T-cell clonal expansion, though a role for this interaction in the effector phase cannot be ruled out. In contrast, B7-CTLA4 interaction enhances the CTL-mediated destruction of tumors, but not T-cell clonal expansion.
Collapse
MESH Headings
- Abatacept
- Adoptive Transfer
- Animals
- Antigens, CD/genetics
- Antigens, CD/immunology
- Antigens, CD/metabolism
- Antigens, Differentiation/immunology
- Antigens, Differentiation/metabolism
- Antigens, Neoplasm/immunology
- B7-1 Antigen/genetics
- B7-1 Antigen/immunology
- B7-1 Antigen/metabolism
- B7-2 Antigen
- CD28 Antigens/genetics
- CD28 Antigens/metabolism
- CTLA-4 Antigen
- Cytotoxicity, Immunologic/drug effects
- Immunity
- Immunoconjugates
- Lymphocyte Activation/drug effects
- Membrane Glycoproteins/genetics
- Membrane Glycoproteins/immunology
- Membrane Glycoproteins/metabolism
- Mice
- Mice, Transgenic
- Mutation
- Neoplasms, Experimental/therapy
- Protein Binding/immunology
- Receptors, Antigen, T-Cell/immunology
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Cytotoxic/transplantation
Collapse
Affiliation(s)
- Xue-Feng Bai
- Department of Pathology and the Comprehensive Cancer Center, Ohio State University Medical Center, Columbus 43210, USA
| | | | | | | | | | | |
Collapse
|
37
|
Mittrücker HW, Kursar M, Köhler A, Hurwitz R, Kaufmann SH. Role of CD28 for the generation and expansion of antigen-specific CD8(+) T lymphocytes during infection with Listeria monocytogenes. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2001; 167:5620-7. [PMID: 11698433 DOI: 10.4049/jimmunol.167.10.5620] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Infection of mice with the intracellular bacterium Listeria monocytogenes results in a strong CD8(+) T cell response that is critical for efficient control of infection. We used CD28-deficient mice to characterize the function of CD28 during Listeria infection, with a main emphasis on Listeria-specific CD8(+) T cells. Frequencies and effector functions of these T cells were determined using MHC class I tetramers, single cell IFN-gamma production and Listeria-specific cytotoxicity. During primary Listeria infection of CD28(-/-) mice we observed significantly reduced numbers of Listeria-specific CD8(+) T cells and only marginal levels of specific IFN-gamma production and cytotoxicity. Although frequencies were also reduced in CD28(-/-) mice during secondary response, we detected a considerable population of Listeria-specific CD8(+) T cells in these mice. In parallel, IFN-gamma production and cytotoxicity were observed, revealing that Listeria-specific CD8(+) T cells in CD28(-/-) mice expressed normal effector functions. Consistent with their impaired CD8(+) T cell activation, CD28(-/-) mice suffered from exacerbated listeriosis both after primary and secondary infection. These results demonstrate participation of CD28 signaling in the generation and expansion of Ag-specific CD8(+) T cells in listeriosis. However, Ag-specific CD8(+) T cells generated in the absence of CD28 differentiated into normal effector and memory T cells.
Collapse
Affiliation(s)
- H W Mittrücker
- Department of Immunology, Max Planck Institute for Infection Biology, Berlin, Germany.
| | | | | | | | | |
Collapse
|
38
|
Liu X, Bai XF, Wen J, Gao JX, Liu J, Lu P, Wang Y, Zheng P, Liu Y. B7H costimulates clonal expansion of, and cognate destruction of tumor cells by, CD8(+) T lymphocytes in vivo. J Exp Med 2001; 194:1339-48. [PMID: 11696598 PMCID: PMC2195972 DOI: 10.1084/jem.194.9.1339] [Citation(s) in RCA: 99] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2000] [Revised: 07/13/2001] [Accepted: 07/24/2001] [Indexed: 01/03/2023] Open
Abstract
B7H/B7RP (hereby called B7H) is a new member of the B7 family of costimulatory molecules and interacts with inducible costimulatory molecule (ICOS). Its function for CD8 T cells has not been reported. We report here that expression of B7H on the tumor cells reduced tumorigenicity and induced immunity to subsequent challenge with parental tumor cells. The immune protection correlates with an enhanced cytotoxic T lymphocyte (CTL) response against P1A, the major tumor antigen expressed in the J558 tumor. To understand the mechanism of immune protection, we adoptively transferred transgenic T cells specific for tumor antigen P1A into mice that bore P1A-expressing tumors. We found that while the transgenic T cells divided faster in mice bearing the B7H(+) tumors, optimal B7H-induced clonal expansion of P1CTL required costimulation by B7-1 and B7-2 on the endogenous host antigen-presenting cells (APCs). Interestingly, when B7H(+) and B7H(-) tumors were coinjected, P1CTL selectively eliminated the B7H(+) tumor cells. Moreover, B7H expressed on the tumor cells made them highly susceptible to destruction by CTL in vivo, even if the CTL was administrated into mice with large tumor burdens. Tumors that recurred in the P1CTL-treated mice lost transfected B7H and/or H-2L(d), the class I molecule that presents the P1A peptide. Taken together, our results reveal that B7H costimulates clonal expansion of, and cognate destruction by CD8(+) T lymphocytes in vivo.
Collapse
Affiliation(s)
- X Liu
- Department of Pathology and Comprehensive Cancer Center, Ohio State University Medical Center, Columbus, OH 43210, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Bai XF, Bender J, Liu J, Zhang H, Wang Y, Li O, Du P, Zheng P, Liu Y. Local costimulation reinvigorates tumor-specific cytolytic T lymphocytes for experimental therapy in mice with large tumor burdens. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2001; 167:3936-43. [PMID: 11564812 DOI: 10.4049/jimmunol.167.7.3936] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Cytotoxic T cells recognize tumor Ags and destroy cancer cells in vitro. Adoptive transfer studies with transgenic T cells specific for tumor Ags have demonstrated that CTL are effective only in mice with small tumor burdens and thus appear to have limited potential in cancer immunotherapy. Here we used transgenic mice that express the TCR specific for an unmutated tumor Ag P1A and multiple lineages of P1A-expressing tumors to address this critical issue. We found that local costimulation, either by expression of B7-1 on the tumor cells or by local administration of anti-CD28 mAb 37N, reinvigorated the function of CTL specific for the tumor Ag, as it substantially increased the efficacy of CTL therapy for mice with large tumor burdens. Our study suggests that CTL-based immunotherapy can be manipulated to deal with large tumors.
Collapse
MESH Headings
- Abatacept
- Animals
- Antibodies, Monoclonal/therapeutic use
- Antigens, CD
- Antigens, Differentiation/immunology
- Antigens, Neoplasm/immunology
- B7-1 Antigen/genetics
- B7-1 Antigen/physiology
- CD28 Antigens/immunology
- CTLA-4 Antigen
- Cell Division
- Cytokines/biosynthesis
- Cytokines/genetics
- Cytotoxicity Tests, Immunologic
- Immunoconjugates
- Immunotherapy, Adoptive
- Lymphocyte Activation
- Mice
- Mice, Transgenic
- Neoplasms, Experimental/immunology
- Neoplasms, Experimental/pathology
- Neoplasms, Experimental/therapy
- RNA, Messenger/biosynthesis
- Survival Rate
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Cytotoxic/transplantation
Collapse
Affiliation(s)
- X F Bai
- Department of Pathology and Comprehensive Cancer Center, Ohio State University Medical Center, 1645 Neil Avenue, Columbus, OH 43210, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Ochsenbein AF, Sierro S, Odermatt B, Pericin M, Karrer U, Hermans J, Hemmi S, Hengartner H, Zinkernagel RM. Roles of tumour localization, second signals and cross priming in cytotoxic T-cell induction. Nature 2001; 411:1058-64. [PMID: 11429607 DOI: 10.1038/35082583] [Citation(s) in RCA: 373] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The vertebrate immune system has evolved to protect against infections that threaten survival before reproduction. Clinically manifest tumours mostly arise after the reproductive years and somatic mutations allow even otherwise antigenic tumours to evade the attention of the immune system. Moreover, the lack of immunological co-stimulatory molecules on solid tumours could result in T-cell tolerance; that is, the failure of T cells to respond. However, this may not generally apply. Here we report several important findings regarding the immune response to tumours, on the basis of studies of several tumour types. First, tumour-specific induction of protective cytotoxic T cells (CTLs) depends on sufficient tumour cells reaching secondary lymphatic organs early and for a long enough duration. Second, diffusely invading systemic tumours delete CTLs. Third, tumours that stay strictly outside secondary lymphatic organs, or that are within these organs but separated from T cells by barriers, are ignored by T cells but do not delete them. Fourth, co-stimulatory molecules on tumour cells do not influence CTL priming but enhance primed CTL responses in peripheral solid tumours. Last, cross priming of CTLs by tumour antigens, mediated by major histocompatibility complex (MHC) class I molecules of antigen-presenting host cells, is inefficient and not protective. These rules of T-cell induction and maintenance not only change previous views but also rationales for anti-tumour immunotherapy.
Collapse
Affiliation(s)
- A F Ochsenbein
- Institute of Experimental Immunology, University Hospital, CH-8091 Zurich, Switzerland.
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Santori FR, Arsov I, Vukmanović S. Modulation of CD8+ T cell response to antigen by the levels of self MHC class I. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2001; 166:5416-21. [PMID: 11313378 DOI: 10.4049/jimmunol.166.9.5416] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The response of H-Y-specific TCR-transgenic CD8(+) T cells to Ag is characterized by poor proliferation, cytolytic activity, and IFN-gamma secretion. IFN-gamma secretion, but not cytotoxic function, can be rescued by the B7.1 molecule, suggesting that costimulation can selectively enhance some, but not all, effector CD8(+) T cell responses. Although the H-Y epitope binds H-2D(b) relatively less well than some other epitopes, it can induce potent CTL responses in nontransgenic mice, suggesting that the observed poor responsiveness of transgenic CD8(+) T cells cannot be ascribed to the epitope itself. Previously reported reactivity of this TCR to H-2A(b) is also not the cause of the poor responsiveness of the H-Y-specific CD8(+) T cells, as H-Y-specific CD8(+) T cells obtained from genetic backgrounds lacking H-2A(b) also responded poorly. Rather, reducing the levels of H-2(b) class I molecules by breeding the mice to (C57BL/6 x B10.D2)F(1) or TAP1(+/-) backgrounds partially restored cytotoxic activity and enhanced proliferative responses. These findings demonstrate that the self MHC class I gene dosage may regulate the extent of CD8(+) T cell responsiveness to Ag.
Collapse
Affiliation(s)
- F R Santori
- Michael Heidelberger Division of Immunology, Department of Pathology, and Kaplan Comprehensive Cancer Center, New York University School of Medicine, New York, NY 10016, USA
| | | | | |
Collapse
|
42
|
Srivastava PK, Amato RJ. Heat shock proteins: the 'Swiss Army Knife' vaccines against cancers and infectious agents. Vaccine 2001; 19:2590-7. [PMID: 11257397 DOI: 10.1016/s0264-410x(00)00492-8] [Citation(s) in RCA: 112] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
The ability of heat shock proteins to: (a) chaperone peptides, including antigenic peptides; (b) interact with antigen presenting cells through a receptor; (c) stimulate antigen presenting cells to secrete inflammatory cytokines; and (d) mediate maturation of dendritic cells, makes them a one-stop shop for the immune system. These properties also permit the utilization of heat shock proteins for development of a new generation of prophylactic and therapeutic vaccines against cancers and infectious diseases.
Collapse
Affiliation(s)
- P K Srivastava
- Center for Immunotherapy of Cancer and Infectious Diseases, University of Connecticut School of Medicine, Farmington, CT 06030-1601, USA.
| | | |
Collapse
|
43
|
Basu S, Srivastava PK. Heat shock proteins: the fountainhead of innate and adaptive immune responses. Cell Stress Chaperones 2001. [PMID: 11189450 DOI: 10.1379/1466-1268(2000)005<0443:hsptfo>2.0.co;2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The ability of heat shock proteins to (1) chaperone peptides, including antigenic peptides; (2) interact with antigen-presenting cells through a receptor; (3) stimulate antigen-presenting cells to secrete inflammatory cytokines; and (4) mediate maturation of dendritic cells, makes them a unique starting point for generation of immune responses. These properties also permit the use of heat shock proteins for development of a new generation of prophylactic and therapeutic vaccines against cancers and infectious diseases.
Collapse
Affiliation(s)
- S Basu
- Center for Immunotherapy of Cancer and Infectious Diseases, University of Connecticut School of Medicine, Farmington 06030-1601, USA
| | | |
Collapse
|
44
|
Marshall KW, Marks JD. Engineering and characterization of a novel fusion protein incorporating B7.2 and an anti-ErbB-2 single-chain antibody fragment for the activation of Jurkat T cells. J Immunother 2001; 24:27-36. [PMID: 11211146 DOI: 10.1097/00002371-200101000-00004] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The provision of the T-cell costimulatory molecule B7 to tumor cells can be an effective way to trigger a tumor-specific cytolytic T-cell response. One way to provide B7 to tumor cells would be to couple an antitumor antibody directly to B7. Such a molecule should target tumors displaying antigen and provide the costimulatory signal to T cells, resulting in the initiation of an antitumor T-cell response. To this end, a fusion protein was designed that incorporates a single-chain antibody fragment (scFv) to erbB-2 (Her2/neu), an oncogene product overexpressed by 30% to 50% of breast carcinomas, and the ECD of B7-2 (CD86). This fusion protein, expressed and purified from Pichia pastoris, was shown to retain binding activity to both counter receptors, erbB-2 and CD28. The fusion protein was also shown to target erbB-2-positive tumor cells and to deliver a CD28-specific T-cell costimulatory signal. These results suggest that a fusion protein engineered to target tumor cells and signal T cells for activation may be an effective means of cancer immunotherapy. Further studies should be performed to characterize the fusion protein in erbB-2 tumor-bearing mice for in vivo tumor targeting, biodistribution, and efficacy.
Collapse
Affiliation(s)
- K W Marshall
- Department of Anesthesia, University of California, San Francisco, USA
| | | |
Collapse
|
45
|
Schendel DJ, Frankenberger B, Jantzer P, Cayeux S, Nöbetaner E, Willimsky G, Maget B, Pohla H, Blankenstein T. Expression of B7.1 (CD80) in a renal cell carcinoma line allows expansion of tumor-associated cytotoxic T lymphocytes in the presence of an alloresponse. Gene Ther 2000; 7:2007-14. [PMID: 11175312 DOI: 10.1038/sj.gt.3301349] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2000] [Accepted: 09/14/2000] [Indexed: 11/08/2022]
Abstract
We have selected a well-characterized human renal cell carcinoma (RCC) line as the basis for development of a genetically engineered tumor cell vaccine to be applied in an allogeneic setting. This cell line was genetically modified by retroviral transduction to express B7.1 costimulatory molecules. The unmodified tumor cells and B7.1-expressing tumor cells were compared for their ability to induce tumor-associated responses in allogeneic peripheral blood mononuclear cells (PBMC) of two normal control donors having single MHC class I allele matches with the tumor cells. PBMC primed using B7.1-modified tumor cells showed a preponderance of CD3+CD8+ cytotoxic T lymphocytes (CTL) that proliferated over extended periods of time in mixed lymphocyte tumor cell (MLTC) cultures. Strong cytolytic activity developed in the primed populations and included allospecific CTL with specificity for mismatched HLA-A, -B and -C molecules. Nevertheless, it was possible to isolate CTL clones that were able to lyse tumor cells but not lymphoblastoid cells that expressed all the corresponding allospecificities. Thus, induction of complex allospecific responses did not hinder the development of tumor-associated CTL in vitro. These results support the use of this genetically modified allogeneic tumor cell line for vaccination of partial-MHC matched RCC patients.
Collapse
Affiliation(s)
- D J Schendel
- Institute of Molecular Immunology, GSF National Research Center for the Environment and Health, Munich, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Mogi S, Sakurai J, Kohsaka T, Enomoto S, Yagita H, Okumura K, Azuma M. Tumour rejection by gene transfer of 4-1BB ligand into a CD80(+) murine squamous cell carcinoma and the requirements of co-stimulatory molecules on tumour and host cells. Immunology 2000; 101:541-7. [PMID: 11122458 PMCID: PMC2327117 DOI: 10.1046/j.1365-2567.2000.t01-1-00138.x] [Citation(s) in RCA: 44] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
NRS1 is a murine squamous cell carcinoma that constitutively expresses the co-stimulatory molecule CD80 at a high level yet grows as a tumour in syngeneic C3H mice. We examined the effect of gene transfer of the 4-1BB ligand (4-1BBL) into NRS1 cells. Introduction of the 4-1BBL gene efficiently elicited anti-tumour immune responses in syngeneic mice which acquired specific immunity against wild-type tumour. T-cell depletion studies showed that CD8(+), but not CD4(+) T cells were essential for tumour eradication. Our results suggest that the transduced 4-1BBL is more effective than the spontaneously expressed CD80 for generation of primary anti-tumour CD8(+) T-cell responses. In addition to CD80 and CD86, the host-derived 4-1BBL is also involved in the secondary anti-tumour responses. This study indicates the complicated contribution of 4-1BBL, CD80 and CD86 on tumour and host cells in anti-tumour immune responses and a possible therapeutic application of 4-1BBL for human tumour vaccination and gene therapy.
Collapse
MESH Headings
- 4-1BB Ligand
- Animals
- Antigens, CD/immunology
- Antigens, Neoplasm/immunology
- B7-1 Antigen/immunology
- B7-2 Antigen
- CD8-Positive T-Lymphocytes/immunology
- Carcinoma, Squamous Cell/immunology
- Carcinoma, Squamous Cell/pathology
- Carcinoma, Squamous Cell/therapy
- Female
- Gene Transfer Techniques
- Graft Rejection/immunology
- Ligands
- Membrane Glycoproteins/immunology
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C3H
- Mice, Nude
- Neoplasm Transplantation
- Transduction, Genetic
- Transfection
- Tumor Cells, Cultured
- Tumor Necrosis Factor-alpha/genetics
- Tumor Necrosis Factor-alpha/immunology
Collapse
Affiliation(s)
- S Mogi
- Department of Immunology, National Children's Medical Research Center, Tokyo, Japan
| | | | | | | | | | | | | |
Collapse
|
47
|
Basu S, Srivastava PK. Heat shock proteins: the fountainhead of innate and adaptive immune responses. Cell Stress Chaperones 2000; 5:443-51. [PMID: 11189450 PMCID: PMC312875 DOI: 10.1379/1466-1268(2000)005<0443:hsptfo>2.0.co;2] [Citation(s) in RCA: 106] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2000] [Revised: 08/29/2000] [Accepted: 08/30/2000] [Indexed: 11/24/2022] Open
Abstract
The ability of heat shock proteins to (1) chaperone peptides, including antigenic peptides; (2) interact with antigen-presenting cells through a receptor; (3) stimulate antigen-presenting cells to secrete inflammatory cytokines; and (4) mediate maturation of dendritic cells, makes them a unique starting point for generation of immune responses. These properties also permit the use of heat shock proteins for development of a new generation of prophylactic and therapeutic vaccines against cancers and infectious diseases.
Collapse
Affiliation(s)
- Sreyashi Basu
- Center for Immunotherapy of Cancer and Infectious Diseases, University of Connecticut School of Medicine, Farmington, CT 06030-1601, USA
| | - Pramod K. Srivastava
- Center for Immunotherapy of Cancer and Infectious Diseases, University of Connecticut School of Medicine, Farmington, CT 06030-1601, USA
- Correspondence to: Pramod K. Srivastava, Tel: 860 679-4444; Fax: 860 679-4365; .
| |
Collapse
|
48
|
Lentiviral vectors for efficient delivery of CD80 and granulocyte-macrophage– colony-stimulating factor in human acute lymphoblastic leukemia and acute myeloid leukemia cells to induce antileukemic immune responses. Blood 2000. [DOI: 10.1182/blood.v96.4.1317.h8001317_1317_1326] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Cell vaccines engineered to express immunomodulators have shown feasibility in eliminating leukemia in murine models. Vectors for efficient gene delivery to primary human leukemia cells are required to translate this approach to clinical trials. In this study, second-generation lentiviral vectors derived from human immunodeficiency virus 1 were evaluated, with the cytomegalovirus (CMV) promoter driving expression of granulocyte-macrophage–colony-stimulating factor (GM-CSF) and CD80 in separate vectors or in a bicistronic vector. The vectors were pseudotyped with vesicular stomatitis virus G glycoprotein and concentrated to high titers (108-109 infective particles/mL). Human acute lymphoblastic leukemia (ALL), acute myeloid leukemia (AML), and chronic myeloid leukemia cell lines transduced with the monocistronic pHR-CD80 vector or the bicistronic pHR-GM/CD vector became 75% to 95% CD80 positive (CD80+). More important, transduction of primary human ALL and AML blasts with high-titer lentiviral vectors was consistently successful (40%-95% CD80+). The average amount of GM-CSF secretion by the leukemia cell lines transduced with the pHR-GM-CSF monocistronic vector was 2182.9 pg/106 cells per 24 hours. Secretion was markedly lower with the bicistronic pHR-GM/CD vector (average, 225.7 pg/106 cells per 24 hours). Lower amounts of CMV-driven messenger RNA were detected with the bicistronic vector, which may account for its poor expression of GM-CSF. Primary ALL cells transduced to express CD80 stimulated T-cell proliferation in an autologous mixed lymphocyte reaction. This stimulation was specifically blocked with monoclonal antibodies reactive against CD80 or by recombinant cytotoxic T-lymphocyte antigen 4–immunoglobulin fusion protein. These results show the feasibility of efficiently transducing primary leukemia cells with lentiviral vectors to express immunomodulators to elicit antileukemic immune responses.
Collapse
|
49
|
Lentiviral vectors for efficient delivery of CD80 and granulocyte-macrophage– colony-stimulating factor in human acute lymphoblastic leukemia and acute myeloid leukemia cells to induce antileukemic immune responses. Blood 2000. [DOI: 10.1182/blood.v96.4.1317] [Citation(s) in RCA: 69] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
Cell vaccines engineered to express immunomodulators have shown feasibility in eliminating leukemia in murine models. Vectors for efficient gene delivery to primary human leukemia cells are required to translate this approach to clinical trials. In this study, second-generation lentiviral vectors derived from human immunodeficiency virus 1 were evaluated, with the cytomegalovirus (CMV) promoter driving expression of granulocyte-macrophage–colony-stimulating factor (GM-CSF) and CD80 in separate vectors or in a bicistronic vector. The vectors were pseudotyped with vesicular stomatitis virus G glycoprotein and concentrated to high titers (108-109 infective particles/mL). Human acute lymphoblastic leukemia (ALL), acute myeloid leukemia (AML), and chronic myeloid leukemia cell lines transduced with the monocistronic pHR-CD80 vector or the bicistronic pHR-GM/CD vector became 75% to 95% CD80 positive (CD80+). More important, transduction of primary human ALL and AML blasts with high-titer lentiviral vectors was consistently successful (40%-95% CD80+). The average amount of GM-CSF secretion by the leukemia cell lines transduced with the pHR-GM-CSF monocistronic vector was 2182.9 pg/106 cells per 24 hours. Secretion was markedly lower with the bicistronic pHR-GM/CD vector (average, 225.7 pg/106 cells per 24 hours). Lower amounts of CMV-driven messenger RNA were detected with the bicistronic vector, which may account for its poor expression of GM-CSF. Primary ALL cells transduced to express CD80 stimulated T-cell proliferation in an autologous mixed lymphocyte reaction. This stimulation was specifically blocked with monoclonal antibodies reactive against CD80 or by recombinant cytotoxic T-lymphocyte antigen 4–immunoglobulin fusion protein. These results show the feasibility of efficiently transducing primary leukemia cells with lentiviral vectors to express immunomodulators to elicit antileukemic immune responses.
Collapse
|
50
|
Westerman LE, Sund SC, Selvaraj P, Jensen PE. Induction of tumor-specific immunity in mice by immunization with reconstituted tumor membrane liposomes containing recombinant B7-2 (CD86). J Immunother 2000; 23:456-63. [PMID: 10916755 DOI: 10.1097/00002371-200007000-00009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
There has been considerable interest in developing experimental vaccines using genetically modified tumor cells expressing cytokines or costimulatory molecules to enhance immunogenicity. The authors investigated an alternative approach of using protein transfer rather than gene transfer to introduce costimulatory molecules rapidly into tumor membranes. Immunization with a single dose of reconstituted tumor membrane liposomes containing purified recombinant B7-2 (CD86) induced tumor rejection in mice challenged with syngeneic tumors, including the poorly immunogenic AG104A fibrosarcoma. These findings support the possibility that cell-free vaccines composed of reconstituted tumor membrane liposomes containing additional immunostimulatory proteins may offer a practical and safe alternative to genetically modified tumor cells for treating human cancer.
Collapse
Affiliation(s)
- L E Westerman
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia 30322, USA
| | | | | | | |
Collapse
|