1
|
Vile R, Pulido J, Chen A, Kendall B, Tonne J, Metko M, Thompson J, Sangsuwannukul T, Yerovi MC, Diaz R, Webb M, Huff A, Moore M, Schuelke M, Irshad S, Appleton E, Melcher A. Cancer Immunotherapy Using AIRE Conditioning of the Tumor Epitopeome. RESEARCH SQUARE 2024:rs.3.rs-5411393. [PMID: 39606441 PMCID: PMC11601838 DOI: 10.21203/rs.3.rs-5411393/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
T cell immune tolerance is established in part through the activity of the Auto-immune Regulator (AIRE) transcription factor in the medullary Thymic Epithelial Cells (mTEC) of the thymus. AIRE induces expression of SELF peripheral tissue-specific antigens for presentation to naïve T cells to promote activation/deletion of potentially autoreactive T cells. We show, for the first time to our knowledge, that tumors mimic the role of AIRE in mTEC to evade immune rejection. Thus, by expressing a broad range of SELF epitopes against which minimal functional T cell reactivities exist because of thymic deletion, AIRE acts as a master controller of SELFNESS, effectively cloaking the tumor from T cell attack. Moreover, we describe a completely novel immunotherapy in which engineered changes in AIRE expression in tumor cells alters their profile of SELFNESS, exposing both AIRE-modified, and parental unmodified, tumor cells to T cell attack. Consistent with our studies, patient RNAseq shows expression of AIRE predicts response to immune therapies with a strong correlation between AIRE expression and markers of TCR signaling. Therefore, by re-setting the immunological SELFNESS of cancer cells, this novel AIRE-mediated immunotherapy 1). converts a highly tolerized T cell compartment into a heteroclitic tumor-reactive T cell population; 2) confers de novo sensitivity to immune checkpoint blockade upon non-immunogenic tumors; 3). completely removes the need to identify potentially immunogenic tumor-associated antigens as targets for generation of de novo CD8+ and helper CD4+ T cell responses; and 4) leads to potent T cell-mediated rejection of aggressive, immunologically cold, non-immunogenic tumors.
Collapse
|
2
|
Human CD141 + dendritic cells generated from adult peripheral blood monocytes. Cytotherapy 2019; 21:1049-1063. [PMID: 31447319 DOI: 10.1016/j.jcyt.2019.07.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 03/20/2019] [Accepted: 07/22/2019] [Indexed: 12/16/2022]
Abstract
Human CD141+ dendritic cells (DCs), specialized for cross-presentation, have been extensively studied in the development of DC-based therapy against cancer. A series of attempts was made to generate CD141+ DCs from cord blood CD34+ hematopoietic progenitors to overcome the practical limitation of in vivo rareness. In the present study, we identified a culture system that generates high CD141+ DCs. After culture of CD14+ monocytes in the presence of granulocyte macrophage colony-stimulating factor (GM-CSF) and interleukin (IL)-4 for 8 days, CD141 was detected on cells that adhered to the bottom of the culture plate. The attached cells exhibited typical features of immature monocyte-derived DCs (moDCs), except for higher CD86 expression, more dendrites and higher granularity compared with those that did not attach. With 3 additional days of culture, increased CD141 expression on the cells was retained along with adhesion ability and partial expression of CLEC9A, a c-type lectin receptor. Furthermore, the cells exhibited effective uptake of dead cells. Interestingly, the attached moDCs differently responded to polyinosinic:polycytidylic acid (poly I:C) stimulation as well as a mixed lymphocyte reaction. Collectively, our findings show that human CD141+ DCs can be sufficiently generated from peripheral blood CD14+ monocytes, potentiating further investigation into generation of higher yields of cross-priming human DCs in vitro.
Collapse
|
3
|
Single Domain Antibody-Mediated Blockade of Programmed Death-Ligand 1 on Dendritic Cells Enhances CD8 T-cell Activation and Cytokine Production. Vaccines (Basel) 2019; 7:vaccines7030085. [PMID: 31394834 PMCID: PMC6789804 DOI: 10.3390/vaccines7030085] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 08/01/2019] [Accepted: 08/03/2019] [Indexed: 12/22/2022] Open
Abstract
Dendritic cell [DC] vaccines can induce durable clinical responses, at least in a fraction of previously treated, late stage cancer patients. Several preclinical studies suggest that shielding programmed death-ligand 1 [PD-L1] on the DC surface may be an attractive strategy to extend such clinical benefits to a larger patient population. In this study, we evaluated the use of single domain antibody [sdAb] K2, a high affinity, antagonistic, PD-L1 specific sdAb, for its ability to enhance DC mediated T-cell activation and benchmarked it against the use of the monoclonal antibodies [mAbs], MIH1, 29E.2A3 and avelumab. Similar to mAbs, sdAb K2 enhanced antigen-specific T-cell receptor signaling in PD-1 positive (PD-1pos) reporter cells activated by DCs. We further showed that the activation and function of antigen-specific CD8 positive (CD8pos) T cells, activated by DCs, was enhanced by inclusion of sdAb K2, but not mAbs. The failure of mAbs to enhance T-cell activation might be explained by their low efficacy to bind PD-L1 on DCs when compared to binding of PD-L1 on non-immune cells, whereas sdAb K2 shows high binding to PD-L1 on immune as well as non-immune cells. These data provide a rationale for the inclusion of sdAb K2 in DC-based immunotherapy strategies.
Collapse
|
4
|
Taggart D, Andreou T, Scott KJ, Williams J, Rippaus N, Brownlie RJ, Ilett EJ, Salmond RJ, Melcher A, Lorger M. Anti-PD-1/anti-CTLA-4 efficacy in melanoma brain metastases depends on extracranial disease and augmentation of CD8 + T cell trafficking. Proc Natl Acad Sci U S A 2018; 115:E1540-E1549. [PMID: 29386395 PMCID: PMC5816160 DOI: 10.1073/pnas.1714089115] [Citation(s) in RCA: 168] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Inhibition of immune checkpoints programmed death 1 (PD-1) and cytotoxic T lymphocyte-associated protein 4 (CTLA-4) on T cells results in durable antitumor activity in melanoma patients. Despite high frequency of melanoma brain metastases (BrM) and associated poor prognosis, the activity and mechanisms of immune checkpoint inhibitors (ICI) in metastatic tumors that develop within the "immune specialized" brain microenvironment, remain elusive. We established a melanoma tumor transplantation model with intracranial plus extracranial (subcutaneous) tumor, mimicking the clinically observed coexistence of metastases inside and outside the brain. Strikingly, intracranial ICI efficacy was observed only when extracranial tumor was present. Extracranial tumor was also required for ICI-induced increase in CD8+ T cells, macrophages, and microglia in brain tumors, and for up-regulation of immune-regulatory genes. Combined PD-1/CTLA-4 blockade had a superior intracranial efficacy over the two monotherapies. Cell depletion studies revealed that NK cells and CD8+ T cells were required for intracranial anti-PD-1/anti-CTLA-4 efficacy. Rather than enhancing CD8+ T cell activation and expansion within intracranial tumors, PD-1/CTLA-4 blockade dramatically (∼14-fold) increased the trafficking of CD8+ T cells to the brain. This was mainly through the peripheral expansion of homing-competent effector CD8+ T cells and potentially further enhanced through up-regulation of T cell entry receptors intercellular adhesion molecule 1 and vascular adhesion molecule 1 on tumor vasculature. Our study indicates that extracranial activation/release of CD8+ T cells from PD-1/CTLA-4 inhibition and potentiation of their recruitment to the brain are paramount to the intracranial anti-PD-1/anti-CTLA-4 activity, suggesting augmentation of these processes as an immune therapy-enhancing strategy in metastatic brain cancer.
Collapse
Affiliation(s)
- David Taggart
- Leeds Institute of Cancer and Pathology, University of Leeds, Leeds LS9 7TF, United Kingdom
- MRC Centre for Inflammation Research, University of Edinburgh, Edinburgh EH8 9YL, United Kingdom
| | - Tereza Andreou
- Leeds Institute of Cancer and Pathology, University of Leeds, Leeds LS9 7TF, United Kingdom
| | - Karen J Scott
- Leeds Institute of Cancer and Pathology, University of Leeds, Leeds LS9 7TF, United Kingdom
| | - Jennifer Williams
- Leeds Institute of Cancer and Pathology, University of Leeds, Leeds LS9 7TF, United Kingdom
| | - Nora Rippaus
- Leeds Institute of Cancer and Pathology, University of Leeds, Leeds LS9 7TF, United Kingdom
| | - Rebecca J Brownlie
- Leeds Institute of Cancer and Pathology, University of Leeds, Leeds LS9 7TF, United Kingdom
| | - Elizabeth J Ilett
- Leeds Institute of Cancer and Pathology, University of Leeds, Leeds LS9 7TF, United Kingdom
| | - Robert J Salmond
- Leeds Institute of Cancer and Pathology, University of Leeds, Leeds LS9 7TF, United Kingdom
| | - Alan Melcher
- Leeds Institute of Cancer and Pathology, University of Leeds, Leeds LS9 7TF, United Kingdom
- The Institute of Cancer Research, The Royal Marsden NHS Foundation Trust, London SW3 6JJ, United Kingdom
| | - Mihaela Lorger
- Leeds Institute of Cancer and Pathology, University of Leeds, Leeds LS9 7TF, United Kingdom;
| |
Collapse
|
5
|
Liu D, Qiu Q, Zhang X, Dai M, Qin J, Hao J, Liao M, Cao W. Infection of chicken bone marrow mononuclear cells with subgroup J avian leukosis virus inhibits dendritic cell differentiation and alters cytokine expression. INFECTION GENETICS AND EVOLUTION 2016; 44:130-136. [DOI: 10.1016/j.meegid.2016.06.045] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Revised: 06/17/2016] [Accepted: 06/23/2016] [Indexed: 11/16/2022]
|
6
|
Mac Keon S, Ruiz MS, Gazzaniga S, Wainstok R. Dendritic cell-based vaccination in cancer: therapeutic implications emerging from murine models. Front Immunol 2015; 6:243. [PMID: 26042126 PMCID: PMC4438595 DOI: 10.3389/fimmu.2015.00243] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Accepted: 05/06/2015] [Indexed: 01/29/2023] Open
Abstract
Dendritic cells (DCs) play a pivotal role in the orchestration of immune responses, and are thus key targets in cancer vaccine design. Since the 2010 FDA approval of the first cancer DC-based vaccine (Sipuleucel-T), there has been a surge of interest in exploiting these cells as a therapeutic option for the treatment of tumors of diverse origin. In spite of the encouraging results obtained in the clinic, many elements of DC-based vaccination strategies need to be optimized. In this context, the use of experimental cancer models can help direct efforts toward an effective vaccine design. This paper reviews recent findings in murine models regarding the antitumoral mechanisms of DC-based vaccination, covering issues related to antigen sources, the use of adjuvants and maturing agents, and the role of DC subsets and their interaction in the initiation of antitumoral immune responses. The summary of such diverse aspects will highlight advantages and drawbacks in the use of murine models, and contribute to the design of successful DC-based translational approaches for cancer treatment.
Collapse
Affiliation(s)
- Soledad Mac Keon
- Laboratorio de Cancerología, Fundación Instituto Leloir, Instituto de Investigaciones Bioquímicas de Buenos Aires IIBBA-CONICET , Buenos Aires , Argentina
| | - María Sol Ruiz
- Centro de Investigaciones Oncológicas, Fundación para la Investigación, Docencia y Prevención del Cáncer (FUCA) , Buenos Aires , Argentina
| | - Silvina Gazzaniga
- Laboratorio de Biología Tumoral, Departamento de Química Biológica IQUIBICEN-CONICET, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires , Buenos Aires , Argentina
| | - Rosa Wainstok
- Laboratorio de Cancerología, Fundación Instituto Leloir, Instituto de Investigaciones Bioquímicas de Buenos Aires IIBBA-CONICET , Buenos Aires , Argentina ; Laboratorio de Biología Tumoral, Departamento de Química Biológica IQUIBICEN-CONICET, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires , Buenos Aires , Argentina
| |
Collapse
|
7
|
Harada Y, Okada-Nakanishi Y, Ueda Y, Tsujitani S, Saito S, Fuji-Ogawa T, Iida A, Hasegawa M, Ichikawa T, Yonemitsu Y. Cytokine-based high log-scale expansion of functional human dendritic cells from cord-blood CD34-positive cells. Sci Rep 2011; 1:174. [PMID: 22355689 PMCID: PMC3240956 DOI: 10.1038/srep00174] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2011] [Accepted: 11/14/2011] [Indexed: 11/09/2022] Open
Abstract
Dendritic cells (DCs) play a crucial role in maintaining the immune system. Though DC-based cancer immunotherapy has been suggested as a potential treatment for various kinds of malignancies, its clinical efficacies are still insufficient in many human trials. Issues that limit the clinical efficacy of DC-based immunotherapy, as well as the difficulty of the industrial production of DCs, are largely due to the limited number of autologous DCs available from each patient. We here established a possible breakthrough, a simple cytokine-based culture method to expand the log-scale order of functional human DCs. Floating cultivation of cord-blood CD34(+) cells under an optimized cytokine cocktail led these progenitor cells to stable log-scale proliferation and to DC differentiation. The expanded DCs had typical features of conventional myeloid DCs in vitro. Therefore, the concept of DC expansion should contribute significantly to the progress of DC immunotherapy.
Collapse
Affiliation(s)
- Yui Harada
- Department of Urology, Chiba University Graduate School ofMedicine, Chiba 260-8670, Japan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
8
|
WeiPeng, Zhao G, Ma Y, Yu H, Wang X. Dendritic cells transfected with PEG10 recombinant adenovirus elicit anti-tumor immune response in vitro and in vivo. Vaccine 2011; 29:3501-6. [DOI: 10.1016/j.vaccine.2011.02.027] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2010] [Revised: 02/04/2011] [Accepted: 02/09/2011] [Indexed: 12/12/2022]
|
9
|
Kondoh H, Okano S, Yoshida K, Yonemitsu Y, Tomita Y, Yoshikai Y, Wake N, Sueishi K. Semi-allogeneic dendritic cells injected via the intratumoural injection route show efficient antitumour effects in cooperation with host-derived professional antigen-presenting cells. Scand J Immunol 2010; 72:476-90. [PMID: 21044122 DOI: 10.1111/j.1365-3083.2010.02461.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Dendritic cells (DC)-based immunotherapy is a potent anticancer modality. In DC-based immunotherapy, allogeneic DC may be an alternative source, but the usefulness of allogeneic DC in DC-based immunotherapy is still controversial. When used for immunotherapy, three factors may affect the efficiency of an allogeneic DC-driven antitumour response: (1) survival time, which is affected by T-cell alloresponses; (2) major histocompatibility complex incompatibility with the host cells in the context of antigen presentation; and (3) the role of host-derived professional antigen-presenting cells (pAPC). In addition, it is unclear which injection route is preferable when using allogeneic DC. In this study, we demonstrate that semi-allogeneic DC, which share half of the genes of the recipient, are more effective when used via the intratumoural (i.t.) injection route, rather than the subcutaneous (s.c.) injection route, for the induction of efficient antitumour effects and the generation of a significant tumour-specific CD8(+) T-cell response. The i.t. route has the advantage of not requiring ex vivo pulsation with tumour lysates or tumour antigens, because the i.t.-injected DC can engulf tumour antigens in situ. Allogeneic bone marrow transplantation (BMT) models, which permit us to separately assess the three factors described previously, show that while all three factors are important for efficient antitumour effects, the control of the alloresponse to injected DC is the most crucial for host-derived pAPC to function well when DC are administered intratumourally. This information may be useful for DC-based cancer immunotherapy under circumstances that do not allow for the use of autologous DC.
Collapse
Affiliation(s)
- H Kondoh
- Division of Pathophysiological and Experimental Pathology, Department of Pathology, Graduate School of Medical Sciences, Kyushu University, Higashi-ku, Fukuoka, Japan
| | | | | | | | | | | | | | | |
Collapse
|
10
|
van den Ancker W, van Luijn MM, Westers TM, Bontkes HJ, Ruben JM, de Gruijl TD, Ossenkoppele GJ, van de Loosdrecht AA. Recent advances in antigen-loaded dendritic cell-based strategies for treatment of minimal residual disease in acute myeloid leukemia. Immunotherapy 2010; 2:69-83. [PMID: 20635890 DOI: 10.2217/imt.09.85] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Therapeutic vaccination with dendritic cells (DCs) is recognized as an important experimental therapy for the treatment of minimal residual disease in acute myeloid leukemia. Many sources of leukemia-associated antigens and different methods for antigen loading of DCs have been used in an attempt to optimize anti-tumor responses. For instance, monocyte-derived DCs have been loaded with apoptotic whole-cell suspensions, necrotic cell lysates, tumor-associated peptides, eluted peptides and cellular DNA or RNA. Furthermore, monocyte-derived DCs can be chemically or electrically fused with leukemic blasts, and DCs have been cultured out of leukemic blasts. However, it remains a challenge in cancer immunotherapy to identify which of these methods is the most optimal for antigen loading and activation of DCs. This review discusses recent advances in DC research and the application of this knowledge towards new strategies for antigen loading of DCs in the treatment of minimal residual disease in acute myeloid leukemia.
Collapse
Affiliation(s)
- Willemijn van den Ancker
- Department of Hematology, VU Institute for Cancer & Immunology, Cancer Center Amsterdam, VU University Medical Center, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands.
| | | | | | | | | | | | | | | |
Collapse
|
11
|
High-frequency alloreactive T cells augment effector function of low-frequency CD8+ T-cell responses under CD28/CD154 blockade. Transplantation 2010; 89:1208-17. [PMID: 20407401 DOI: 10.1097/tp.0b013e3181df53dc] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND Blockade of costimulatory molecules is a potent method of inducing long-term graft survival. We have previously addressed the issue of donor-reactive T-cell precursor frequency on relative costimulation dependence and found that the presence of a high precursor frequency of donor-reactive CD8 T cells resulted in costimulation blockade-resistant graft rejection, whereas the presence of a low-frequency donor-reactive population did not. To address the mechanisms by which high-frequency T cells obviated the requirement for costimulation, we asked whether a low-frequency population responding concomitantly with a high-frequency response also demonstrated costimulation independence. METHODS A model system was established in which B6 mice containing a low frequency of anti-membrane bound chicken ovalbumin (mOVA) responders and a high frequency of anti-BALB/c responders received a skin graft from B6.mOVAxBALB/c F1 donors in the presence or absence of cytotoxic T-lymphocyte antigen-4 Ig/anti-CD154 costimulatory blockade. RESULTS The results revealed that in the presence of costimulation blockade, high-frequency anti-BALB/c T cells augmented the effector activity of low-frequency anti-mOVA T cells, but it did not enhance the accumulation of anti-mOVA T cells capable of mediating graft rejection. CONCLUSIONS These results demonstrate that both antigen-specific and antigen-independent factors contribute to the relative costimulation independence of high-frequency T-cell responses.
Collapse
|
12
|
Gervais A, Eymard JC, Toulmonde E, Bernard J. Selected allogeneic dendritic cells markedly enhance human tumour antigen-specific T cell response in vitro. Cancer Immunol Immunother 2009; 58:1831-41. [PMID: 19330330 PMCID: PMC11030287 DOI: 10.1007/s00262-009-0694-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2008] [Accepted: 03/07/2009] [Indexed: 12/11/2022]
Abstract
BACKGROUND Alloreaction is known to accumulate several theoretical advantages that can improve dendritic cell (DC)-based anti-infective or antitumour strategies. Allogeneic DC have already been tested in experimental and clinical studies, but their efficacy compared with their autologous counterparts was rarely investigated and conclusions diverge. OBJECTIVE This study compared antigen-specific T cell responses following priming with autologous versus allogeneic DC and examined the possibility of screening these responses in order to select allogeneic DC that lead to a great amplification. RESULTS Allogeneic DC obtained from donors matched with the single HLA-A2 allele were efficient in generating in vitro peptide-specific T cell responses. When randomly chosen, allogeneic DC generated a broad range of antigen-specific T cell responses in comparison with autologous DC. When screened and selected, allogeneic DC markedly enhanced peptide-specific T cell priming and allowed a more efficient boosting of resulting T cells. These selected allogeneic DC provided a favourable cytokinic and cellular environment that can help concurrent antigen-specific responses. CONCLUSION Ex vivo selected allogeneic DC provide adjuvant effects that lead to amplification of concomitant antigen-specific T cell responses.
Collapse
Affiliation(s)
- Alban Gervais
- Institut Jean Godinot, Unité de Thérapie Cellulaire, Reims, France.
| | | | | | | |
Collapse
|
13
|
Dendritic cells and T cells deliver oncolytic reovirus for tumour killing despite pre-existing anti-viral immunity. Gene Ther 2009; 16:689-99. [PMID: 19282847 DOI: 10.1038/gt.2009.29] [Citation(s) in RCA: 98] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Reovirus is a naturally occurring oncolytic virus currently in early clinical trials. However, the rapid induction of neutralizing antibodies represents a major obstacle to successful systemic delivery. This study addresses, for the first time, the ability of cellular carriers in the form of T cells and dendritic cells (DC) to protect reovirus from systemic neutralization. In addition, the ability of these cellular carriers to manipulate the subsequent balance of anti-viral versus anti-tumour immune response is explored. Reovirus, either neat or loaded onto DC or T cells, was delivered intravenously into reovirus-naive or reovirus-immune C57Bl/6 mice bearing lymph node B16tk melanoma metastases. Three and 10 days after treatment, reovirus delivery, carrier cell trafficking, metastatic clearance and priming of anti-tumour/anti-viral immunity were assessed. In naive mice, reovirus delivered either neat or through cell carriage was detectable in the tumour-draining lymph nodes 3 days after treatment, though complete clearance of metastases was only obtained when the virus was delivered on T cells or mature DC (mDC); neat reovirus or loaded immature DC (iDC) gave only partial early tumour clearance. Furthermore, only T cells carrying reovirus generated anti-tumour immune responses and long-term tumour clearance; reovirus-loaded DC, in contrast, generated only an anti-viral immune response. In reovirus-immune mice, however, the results were different. Neat reovirus was completely ineffective as a therapy, whereas mDC--though not iDC--as well as T cells, effectively delivered reovirus to melanoma in vivo for therapy and anti-tumour immune priming. Moreover, mDC were more effective than T cells over a range of viral loads. These data show that systemically administered neat reovirus is not optimal for therapy, and that DC may be an appropriate vehicle for carriage of significant levels of reovirus to tumours. The pre-existing immune status against the virus is critical in determining the balance between anti-viral and anti-tumour immunity elicited when reovirus is delivered by cell carriage, and the viral dose and mode of delivery, as well as the immune status of patients, may profoundly affect the success of any clinical anti-tumour viral therapy. These findings are therefore of direct translational relevance for the future design of clinical trials.
Collapse
|
14
|
Prestwich RJ, Scott KJ, Brown J, Harnden P, Whelan P, Cartledge J, O’Donnell D, Pandha HS, Selby PJ, Banks RE, Merrick AE, Melcher AA. The feasibility of establishing a programme of adjuvant autologous vaccination for renal cell carcinoma. BJU Int 2009; 103:740-6. [DOI: 10.1111/j.1464-410x.2008.08113.x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
|
15
|
van den Ancker W, Westers TM, Ossenkoppele GJ, van de Loosdrecht AA. Back to basics: In search of the optimal dendritic cell for vaccination in AML. Leuk Res 2008; 32:1641-3. [DOI: 10.1016/j.leukres.2008.03.029] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2008] [Revised: 03/26/2008] [Accepted: 03/27/2008] [Indexed: 11/26/2022]
|