1
|
Tittarelli A, Pereda C, Gleisner MA, López MN, Flores I, Tempio F, Lladser A, Achour A, González FE, Durán-Aniotz C, Miranda JP, Larrondo M, Salazar-Onfray F. Long-Term Survival and Immune Response Dynamics in Melanoma Patients Undergoing TAPCells-Based Vaccination Therapy. Vaccines (Basel) 2024; 12:357. [PMID: 38675738 PMCID: PMC11053591 DOI: 10.3390/vaccines12040357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 01/05/2024] [Accepted: 01/06/2024] [Indexed: 04/28/2024] Open
Abstract
Cancer vaccines present a promising avenue for treating immune checkpoint blockers (ICBs)-refractory patients, fostering immune responses to modulate the tumor microenvironment. We revisit a phase I/II trial using Tumor Antigen-Presenting Cells (TAPCells) (NCT06152367), an autologous antigen-presenting cell vaccine loaded with heat-shocked allogeneic melanoma cell lysates. Initial findings showcased TAPCells inducing lysate-specific delayed-type hypersensitivity (DTH) reactions, correlating with prolonged survival. Here, we extend our analysis over 15 years, categorizing patients into short-term (<36 months) and long-term (≥36 months) survivors, exploring novel associations between clinical outcomes and demographic, genetic, and immunologic parameters. Notably, DTHpos patients exhibit a 53.1% three-year survival compared to 16.1% in DTHneg patients. Extended remissions are observed in long-term survivors, particularly DTHpos/M1cneg patients. Younger age, stage III disease, and moderate immune events also benefit short-term survivors. Immunomarkers like increased C-type lectin domain family 2 member D on CD4+ T cells and elevated interleukin-17A were detected in long-term survivors. In contrast, toll-like receptor-4 D229G polymorphism and reduced CD32 on B cells are associated with reduced survival. TAPCells achieved stable long remissions in 35.2% of patients, especially M1cneg/DTHpos cases. Conclusions: Our study underscores the potential of vaccine-induced immune responses in melanoma, emphasizing the identification of emerging biological markers and clinical parameters for predicting long-term remission.
Collapse
Affiliation(s)
- Andrés Tittarelli
- Programa Institucional de Fomento a la Investigación, Desarrollo e Innovación, Universidad Tecnológica Metropolitana, Santiago 8940577, Chile;
| | - Cristian Pereda
- Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago 8380453, Chile; (C.P.); (M.A.G.); (M.N.L.); (I.F.); (F.T.)
| | - María A. Gleisner
- Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago 8380453, Chile; (C.P.); (M.A.G.); (M.N.L.); (I.F.); (F.T.)
- Millennium Institute on Immunology and Immunotherapy, Faculty of Medicine, Universidad de Chile, Santiago 8380453, Chile
| | - Mercedes N. López
- Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago 8380453, Chile; (C.P.); (M.A.G.); (M.N.L.); (I.F.); (F.T.)
| | - Iván Flores
- Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago 8380453, Chile; (C.P.); (M.A.G.); (M.N.L.); (I.F.); (F.T.)
| | - Fabián Tempio
- Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago 8380453, Chile; (C.P.); (M.A.G.); (M.N.L.); (I.F.); (F.T.)
| | - Alvaro Lladser
- Centro Científico y Tecnológico de Excelencia Ciencia & Vida, Fundación Ciencia & Vida, Santiago 8580702, Chile;
- Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago 8580702, Chile
| | - Adnane Achour
- Science for Life Laboratory, Department of Medicine Solna, Karolinska Institute, 17176 Stockholm, Sweden;
- Division of Infectious Diseases, Karolinska University Hospital, 17176 Stockholm, Sweden
| | - Fermín E. González
- Laboratory of Experimental Immunology & Cancer, Faculty of Dentistry, Universidad de Chile, Santiago 8380000, Chile;
| | - Claudia Durán-Aniotz
- Latin American Brain Health Institute (BrainLat), Center for Social and Cognitive Neuroscience (CSCN), School of Psychology, Universidad Adolfo Ibañez, Santiago 7941169, Chile;
| | | | - Milton Larrondo
- Banco de Sangre, Hospital Clínico de la Universidad de Chile, Santiago 8380453, Chile;
| | - Flavio Salazar-Onfray
- Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago 8380453, Chile; (C.P.); (M.A.G.); (M.N.L.); (I.F.); (F.T.)
- Millennium Institute on Immunology and Immunotherapy, Faculty of Medicine, Universidad de Chile, Santiago 8380453, Chile
- Science for Life Laboratory, Department of Medicine Solna, Karolinska Institute, 17176 Stockholm, Sweden;
- Division of Infectious Diseases, Karolinska University Hospital, 17176 Stockholm, Sweden
| |
Collapse
|
2
|
Ebrahim T, Ebrahim AS, Kandouz M. Diversity of Intercellular Communication Modes: A Cancer Biology Perspective. Cells 2024; 13:495. [PMID: 38534339 PMCID: PMC10969453 DOI: 10.3390/cells13060495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 02/27/2024] [Accepted: 03/10/2024] [Indexed: 03/28/2024] Open
Abstract
From the moment a cell is on the path to malignant transformation, its interaction with other cells from the microenvironment becomes altered. The flow of molecular information is at the heart of the cellular and systemic fate in tumors, and various processes participate in conveying key molecular information from or to certain cancer cells. For instance, the loss of tight junction molecules is part of the signal sent to cancer cells so that they are no longer bound to the primary tumors and are thus free to travel and metastasize. Upon the targeting of a single cell by a therapeutic drug, gap junctions are able to communicate death information to by-standing cells. The discovery of the importance of novel modes of cell-cell communication such as different types of extracellular vesicles or tunneling nanotubes is changing the way scientists look at these processes. However, are they all actively involved in different contexts at the same time or are they recruited to fulfill specific tasks? What does the multiplicity of modes mean for the overall progression of the disease? Here, we extend an open invitation to think about the overall significance of these questions, rather than engage in an elusive attempt at a systematic repertory of the mechanisms at play.
Collapse
Affiliation(s)
- Thanzeela Ebrahim
- Department of Pathology, Wayne State University School of Medicine, Detroit, MI 48202, USA
| | - Abdul Shukkur Ebrahim
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI 48202, USA
| | - Mustapha Kandouz
- Department of Pathology, Wayne State University School of Medicine, Detroit, MI 48202, USA
- Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 48202, USA
| |
Collapse
|
3
|
Pérez-Baños A, Gleisner MA, Flores I, Pereda C, Navarrete M, Araya JP, Navarro G, Quezada-Monrás C, Tittarelli A, Salazar-Onfray F. Whole tumour cell-based vaccines: tuning the instruments to orchestrate an optimal antitumour immune response. Br J Cancer 2023; 129:572-585. [PMID: 37355722 PMCID: PMC10421921 DOI: 10.1038/s41416-023-02327-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 05/31/2023] [Accepted: 06/14/2023] [Indexed: 06/26/2023] Open
Abstract
Immunotherapy, particularly those based on immune checkpoint inhibitors (ICIs), has become a useful approach for many neoplastic diseases. Despite the improvements of ICIs in supporting tumour regression and prolonging survival, many patients do not respond or develop resistance to treatment. Thus, therapies that enhance antitumour immunity, such as anticancer vaccines, constitute a feasible and promising therapeutic strategy. Whole tumour cell (WTC) vaccines have been extensively tested in clinical studies as intact or genetically modified cells or tumour lysates, injected directly or loaded on DCs with distinct adjuvants. The essential requirements of WTC vaccines include the optimal delivery of a broad battery of tumour-associated antigens, the presence of tumour cell-derived molecular danger signals, and adequate adjuvants. These factors trigger an early and robust local innate inflammatory response that orchestrates an antigen-specific and proinflammatory adaptive antitumour response capable of controlling tumour growth by several mechanisms. In this review, the strengths and weaknesses of our own and others' experiences in studying WTC vaccines are revised to discuss the essential elements required to increase anticancer vaccine effectiveness.
Collapse
Affiliation(s)
- Amarilis Pérez-Baños
- Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
- Millennium Institute on Immunology and Immunotherapy, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - María Alejandra Gleisner
- Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
- Millennium Institute on Immunology and Immunotherapy, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Iván Flores
- Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
- Millennium Institute on Immunology and Immunotherapy, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Cristián Pereda
- Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
- Millennium Institute on Immunology and Immunotherapy, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Mariela Navarrete
- Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
- Millennium Institute on Immunology and Immunotherapy, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Juan Pablo Araya
- Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
- Millennium Institute on Immunology and Immunotherapy, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Giovanna Navarro
- Millennium Institute on Immunology and Immunotherapy, Faculty of Medicine, Universidad de Chile, Santiago, Chile
- Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, Valdivia, 5110566, Chile
| | - Claudia Quezada-Monrás
- Millennium Institute on Immunology and Immunotherapy, Faculty of Medicine, Universidad de Chile, Santiago, Chile
- Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, Valdivia, 5110566, Chile
| | - Andrés Tittarelli
- Programa Institucional de Fomento a la Investigación, Desarrollo e Innovación (PIDi), Universidad Tecnológica Metropolitana (UTEM), Santiago, Chile.
| | - Flavio Salazar-Onfray
- Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile.
- Millennium Institute on Immunology and Immunotherapy, Faculty of Medicine, Universidad de Chile, Santiago, Chile.
- Science for Life Laboratory, Department of Medicine Solna, Karolinska Institute and Section for Infectious Diseases, Karolinska University Hospital, 17176, Stockholm, Sweden.
| |
Collapse
|
4
|
Oliveira MMS, D'Aulerio R, Yong T, He M, Baptista MAP, Nylén S, Westerberg LS. Increased cross-presentation by dendritic cells and enhanced anti-tumour therapy using the Arp2/3 inhibitor CK666. Br J Cancer 2023; 128:982-991. [PMID: 36631633 PMCID: PMC10006228 DOI: 10.1038/s41416-022-02135-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 12/11/2022] [Accepted: 12/21/2022] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Dendritic cell (DC) vaccines for cancer therapy offer the possibility to let the patient's own immune system kill cancer cells. However, DC vaccines have shown less efficacy than expected due to failure to induce cancer cell killing and by activating T regulatory cells. METHODS We tested if inhibition of signalling via WASp and Arp2/3 using the small molecule CK666 would enhance DC-mediated killing of tumour cells in vitro and in vivo. RESULTS Using CK666 during the ex vivo phase of antigen processing of ovalbumin (OVA), murine and human DCs showed decreased phagosomal acidification, indicating activation of the cross-presentation pathway. When compared to untreated DCs, DCs treated with CK666 during uptake and processing of OVA-induced increased proliferation of OVA-specific CD8+ OT-I T cells in vitro and in vivo. Using the aggressive B16-mOVA melanoma tumour model, we show that mice injected with CK666-treated DCs and OVA-specific CD8+ OT-I T cells showed higher rejection of B16 melanoma cells when compared to mice receiving non-treated DCs. This resulted in the prolonged survival of tumour-bearing mice receiving CK666-treated DCs. Moreover, combining CK666-treated DCs with the checkpoint inhibitor anti-PD1 further prolonged survival. CONCLUSION Our data suggest that the small molecule inhibitor CK666 is a good candidate to enhance DC cross-presentation for cancer therapy.
Collapse
Affiliation(s)
- Mariana M S Oliveira
- Department of Microbiology Tumor and Cell biology, Karolinska Institutet, 17177, Stockholm, Sweden
| | - Roberta D'Aulerio
- Department of Microbiology Tumor and Cell biology, Karolinska Institutet, 17177, Stockholm, Sweden
| | - Tracer Yong
- Department of Microbiology Tumor and Cell biology, Karolinska Institutet, 17177, Stockholm, Sweden
| | - Minghui He
- Department of Microbiology Tumor and Cell biology, Karolinska Institutet, 17177, Stockholm, Sweden
| | - Marisa A P Baptista
- Institute for Virology and Immunobiology, Julius-Maximilians-University Würzburg, Würzburg, Germany
| | - Susanne Nylén
- Department of Microbiology Tumor and Cell biology, Karolinska Institutet, 17177, Stockholm, Sweden
| | - Lisa S Westerberg
- Department of Microbiology Tumor and Cell biology, Karolinska Institutet, 17177, Stockholm, Sweden.
| |
Collapse
|
5
|
Torres A, Vivanco S, Lavín F, Pereda C, Chernobrovkin A, Gleisner A, Alcota M, Larrondo M, López MN, Salazar-Onfray F, Zubarev RA, González FE. Haptoglobin Induces a Specific Proteomic Profile and a Mature-Associated Phenotype on Primary Human Monocyte-Derived Dendritic Cells. Int J Mol Sci 2022; 23:ijms23136882. [PMID: 35805888 PMCID: PMC9266681 DOI: 10.3390/ijms23136882] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Revised: 06/12/2022] [Accepted: 06/14/2022] [Indexed: 11/16/2022] Open
Abstract
Damage-associated molecular patterns (DAMPs) play a critical role in dendritic cells (DCs) ability to trigger a specific and efficient adaptive immune response for different physiological and pathological scenarios. We have previously identified constitutive DAMPs (HMGB1 and Calreticulin) as well as new putative inducible DAMPs such as Haptoglobin (HP), from a therapeutically used heat shock-conditioned melanoma cell lysate (called TRIMEL). Remarkably, HP was shown to be the most abundant protein in the proteomic profile of heat shock-conditioned TRIMEL samples. However, its relative contribution to the observed DCs phenotype has not been fully elucidated. Human DCs were generated from monocytes isolated from PBMC of melanoma patients and healthy donors. DC lineage was induced with rhIL-4 and rhGM-CSF. After additional stimulation with HP, the proteome of these HP-stimulated cells was characterized. In addition, DCs were phenotypically characterized by flow cytometry for canonical maturation markers and cytokine production. Finally, in vitro transmigration capacity was assessed using Transwell plates. Our results showed that the stimulation with HP was associated with the presence of exclusive and higher relative abundance of specific immune-; energy production-; lipid biosynthesis-; and DAMPs-related proteins. Importantly, HP stimulation enhanced the expression of specific DC maturation markers and pro-inflammatory and Th1-associated cytokines, and an in vitro transmigration of primary human DCs. Taken together, these data suggest that HP can be considered as a new inducible DAMP with an important role in in vitro DC activation for cancer immunotherapy.
Collapse
Affiliation(s)
- Alfredo Torres
- Laboratory of Experimental Immunology & Cancer, Faculty of Dentistry, University of Chile, Santiago 8380492, Chile; (A.T.); (S.V.); (F.L.)
- Department of Conservative Dentistry, Faculty of Dentistry, University of Chile, Santiago 8380492, Chile;
| | - Sheilah Vivanco
- Laboratory of Experimental Immunology & Cancer, Faculty of Dentistry, University of Chile, Santiago 8380492, Chile; (A.T.); (S.V.); (F.L.)
| | - Francisca Lavín
- Laboratory of Experimental Immunology & Cancer, Faculty of Dentistry, University of Chile, Santiago 8380492, Chile; (A.T.); (S.V.); (F.L.)
| | - Cristián Pereda
- Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (C.P.); (A.G.); (M.N.L.); (F.S.-O.)
| | - Alexey Chernobrovkin
- Department of Medical Biochemistry and Biophysics, Karolinska Institute, SE17177 Stockholm, Sweden; (A.C.); (R.A.Z.)
| | - Alejandra Gleisner
- Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (C.P.); (A.G.); (M.N.L.); (F.S.-O.)
| | - Marcela Alcota
- Department of Conservative Dentistry, Faculty of Dentistry, University of Chile, Santiago 8380492, Chile;
| | - Milton Larrondo
- Blood Bank Service, University of Chile Clinical Hospital, Santiago 8380453, Chile;
| | - Mercedes N. López
- Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (C.P.); (A.G.); (M.N.L.); (F.S.-O.)
- Millennium Institute on Immunology and Immunotherapy, Faculty of Medicine, University of Chile, Santiago 8380453, Chile
| | - Flavio Salazar-Onfray
- Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (C.P.); (A.G.); (M.N.L.); (F.S.-O.)
- Millennium Institute on Immunology and Immunotherapy, Faculty of Medicine, University of Chile, Santiago 8380453, Chile
| | - Roman A. Zubarev
- Department of Medical Biochemistry and Biophysics, Karolinska Institute, SE17177 Stockholm, Sweden; (A.C.); (R.A.Z.)
| | - Fermín E. González
- Laboratory of Experimental Immunology & Cancer, Faculty of Dentistry, University of Chile, Santiago 8380492, Chile; (A.T.); (S.V.); (F.L.)
- Department of Conservative Dentistry, Faculty of Dentistry, University of Chile, Santiago 8380492, Chile;
- Millennium Institute on Immunology and Immunotherapy, Faculty of Medicine, University of Chile, Santiago 8380453, Chile
- Correspondence: ; Tel.: +56-2-29781714
| |
Collapse
|
6
|
Ozaniak A, Vachtenheim J, Lischke R, Bartunkova J, Strizova Z. Novel Insights into the Immunotherapy of Soft Tissue Sarcomas: Do We Need a Change of Perspective? Biomedicines 2021; 9:biomedicines9080935. [PMID: 34440139 PMCID: PMC8393686 DOI: 10.3390/biomedicines9080935] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/19/2021] [Accepted: 07/27/2021] [Indexed: 12/19/2022] Open
Abstract
Soft tissue sarcomas (STSs) are rare mesenchymal tumors. With more than 80 histological subtypes of STSs, data regarding novel biomarkers of strong prognostic and therapeutic value are very limited. To date, the most important prognostic factor is the tumor grade, and approximately 50% of patients that are diagnosed with high-grade STSs die of metastatic disease within five years. Systemic chemotherapy represents the mainstay of metastatic STSs treatment for decades but induces response in only 15–35% of the patients, irrespective of the histological subtype. In the era of immunotherapy, deciphering the immune cell signatures within the STSs tumors may discriminate immunotherapy responders from non-responders and different immunotherapeutic approaches could be combined based on the predominant cell subpopulations infiltrating the STS tumors. Furthermore, understanding the immune diversity of the STS tumor microenvironment (TME) in different histological subtypes may provide a rationale for stratifying patients according to the TME immune parameters. In this review, we introduce the most important immune cell types infiltrating the STSs tumors and discuss different immunotherapies, as well as promising clinical trials, that would target these immune cells to enhance the antitumor immune responses and improve the prognosis of metastatic STSs patients.
Collapse
Affiliation(s)
- Andrej Ozaniak
- Third Department of Surgery, First Faculty of Medicine, Charles University and University Hospital Motol, 150 06 Prague, Czech Republic; (A.O.); (J.V.J.); (R.L.)
| | - Jiri Vachtenheim
- Third Department of Surgery, First Faculty of Medicine, Charles University and University Hospital Motol, 150 06 Prague, Czech Republic; (A.O.); (J.V.J.); (R.L.)
| | - Robert Lischke
- Third Department of Surgery, First Faculty of Medicine, Charles University and University Hospital Motol, 150 06 Prague, Czech Republic; (A.O.); (J.V.J.); (R.L.)
| | - Jirina Bartunkova
- Department of Immunology, Second Faculty of Medicine, Charles University and University Hospital Motol, 150 06 Prague, Czech Republic;
| | - Zuzana Strizova
- Department of Immunology, Second Faculty of Medicine, Charles University and University Hospital Motol, 150 06 Prague, Czech Republic;
- Correspondence: ; Tel.: +420-604712471
| |
Collapse
|
7
|
Danilova A, Misyurin V, Novik A, Girdyuk D, Avdonkina N, Nekhaeva T, Emelyanova N, Pipia N, Misyurin A, Baldueva I. Cancer/testis antigens expression during cultivation of melanoma and soft tissue sarcoma cells. Clin Sarcoma Res 2020; 10:3. [PMID: 32042403 PMCID: PMC6998350 DOI: 10.1186/s13569-020-0125-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 01/22/2020] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Autologous dendritic cells (DC) loaded with tumor-associated antigens (TAAs) are a promising approach for anticancer immunotherapy. Polyantigen lysates appear to be an excellent source of TAAs for loading onto the patient's dendritic cells. Cancer/testis antigens (CTA) are expressed by a wide range of tumors, but are minimally expressed on normal tissues, and could serve as a universal target for immunotherapy. However, CTA expression levels can vary significantly in patients with the same tumor type. We proposed that patients who do not respond to DC-based therapy may have distinct features of the CTA expression profile on tumor cells. PATIENTS AND METHODS We compared the gene expression of the principal families CTA in 22 melanoma and 27 soft tissue and bone sarcomas cell lines (STBS), received from patients and used for DC vaccine preparation. RESULTS The majority (47 of 49, 95.9%) cell lines showed CTA gene activity. The incidence of gene expression of GAGE, NYESO1, MAGEA1, PRAME's was significantly different (adj. p < 0.05) between melanoma and sarcoma cell lines. The expression of the SCP1 gene was detected neither in melanoma cells nor in the STBS cells. Clustering by the gene expression profile revealed four different expression patterns. We found three main patterns types: hyperexpression of multiple CTA, hyperexpression of one CTA with almost no expression of others, and no expression of CTA. All clusters types exist in melanoma and sarcoma cell lines. We observed dependence of killing efficacy from the PRAME (rho = 0.940, adj. p < 0.01) expression during real-time monitoring with the xCELLigence system of the interaction between melanoma or sarcoma cells with the T-lymphocytes activated by the lysate of selected allogenous melanoma cell lines with high expression of CTA. CONCLUSION Our results demonstrate that one can use lysates from allogeneic melanoma cell lines as a source of CTA for DC load during the production of anticancer vaccines for the STBS treatment. Patterns of CTA expression should be evaluated as biomarkers of response in prospective clinical trials.
Collapse
Affiliation(s)
- Anna Danilova
- N.N. Petrov’ National Medical Cancer Research Center, Leningradskaya str., 68, Pesochniy, Saint-Petersburg, 197758 Russian Federation
- Department of Oncoimmunology, N.N. Petrov’ National Medical Cancer Research Center, Leningradskaya str., 68, Pesochniy, Saint-Petersburg, 197758 Russian Federation
| | - Vsevolod Misyurin
- N.N. Blokhin’ National Medical Cancer Research Center, Kashirskoye sh. 24, Moscow, 115478 Russian Federation
| | - Aleksei Novik
- N.N. Petrov’ National Medical Cancer Research Center, Leningradskaya str., 68, Pesochniy, Saint-Petersburg, 197758 Russian Federation
| | - Dmitry Girdyuk
- N.N. Petrov’ National Medical Cancer Research Center, Leningradskaya str., 68, Pesochniy, Saint-Petersburg, 197758 Russian Federation
| | - Natalia Avdonkina
- N.N. Petrov’ National Medical Cancer Research Center, Leningradskaya str., 68, Pesochniy, Saint-Petersburg, 197758 Russian Federation
| | - Tatiana Nekhaeva
- N.N. Petrov’ National Medical Cancer Research Center, Leningradskaya str., 68, Pesochniy, Saint-Petersburg, 197758 Russian Federation
| | - Natalia Emelyanova
- N.N. Petrov’ National Medical Cancer Research Center, Leningradskaya str., 68, Pesochniy, Saint-Petersburg, 197758 Russian Federation
| | - Nino Pipia
- N.N. Petrov’ National Medical Cancer Research Center, Leningradskaya str., 68, Pesochniy, Saint-Petersburg, 197758 Russian Federation
| | - Andrey Misyurin
- N.N. Blokhin’ National Medical Cancer Research Center, Kashirskoye sh. 24, Moscow, 115478 Russian Federation
| | - Irina Baldueva
- N.N. Petrov’ National Medical Cancer Research Center, Leningradskaya str., 68, Pesochniy, Saint-Petersburg, 197758 Russian Federation
| |
Collapse
|
8
|
Dendritic Cells Loaded with Heat Shock-Conditioned Ovarian Epithelial Carcinoma Cell Lysates Elicit T Cell-Dependent Antitumor Immune Responses In Vitro. J Immunol Res 2019; 2019:9631515. [PMID: 31886313 PMCID: PMC6899292 DOI: 10.1155/2019/9631515] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 09/08/2019] [Accepted: 10/16/2019] [Indexed: 12/13/2022] Open
Abstract
Ovarian epithelial carcinoma (OEC) is the most frequent ovarian tumor, characterized by a high mortality in advanced stages where conventional therapies are not effective. Based on the role of the immune system in the progression of this disease, immunotherapy using checkpoint blockade has been considered as a therapeutic alternative. Nevertheless, its results do not match up to the positive results in entities like melanoma and other malignancies, suggesting the need to find other therapies to be used alone or in combination. Dendritic cell- (DC-) based vaccines have shown promising results in several types of cancer, such as melanoma, prostate, and lung cancers, due to the essential role played by DCs in the activation of specific T cells, thus using other ways of activating the immune response than immune checkpoint blockade. During the last decade, we have used DC-based vaccines loaded with an allogeneic heat shock-conditioned melanoma cell lysate in the treatment of advanced stage patients in a series of clinical trials. In these studies, 60% of treated patients showed immunological responses which correlated positively with improved survival. Considering the relevance of ovarian cancer and the promising results of our DC-based vaccine, we show here that heat shock-conditioned cell lysates derived from ovarian epithelial carcinoma cell lines have the potential to induce the phenotypic and functional maturation of human DC, which in turn, is able to induce an efficient CD4+ and CD8+ T cell-mediated immune responses against ovarian cancer cell lines in vitro. In summary, OEC heat shock-conditioned cell lysate-loaded DCs may be considered for future combined immunotherapy approaches against ovarian tumors.
Collapse
|
9
|
Robles-Planells C, Michelson SA, Mena J, Escrig D, Rojas JL, Sanchez-Guerrero G, Hernández R, Barrera-Avalos C, Rojo LE, Sauma D, Kalergis AM, Imarai M, Fernández R, Robles CA, Leiva-Salcedo E, Santander R, Escobar A, Acuña-Castillo C. Lithraea caustic (Litre) Extract Promotes an Antitumor Response Against B16 Melanoma. Front Pharmacol 2019; 10:1201. [PMID: 31695610 PMCID: PMC6817581 DOI: 10.3389/fphar.2019.01201] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Accepted: 09/17/2019] [Indexed: 11/17/2022] Open
Abstract
Melanoma immunotherapy, specifically the autotransplant of dendritic cells charged with tumors antigens, has shown promising results in clinical trials. The positive clinical effects of this therapy have been associated to increased Th17 response and delayed-type hypersensitivity (DTH) against to tumor antigens. Some synthetic compounds, such as diphenylcyclopropenone (DPCP), are capable of triggering a DTH response in cutaneous malignancies and also to induce clinically relevant effects against melanoma. In this work, we evaluated Litre extract (LExT), a standardized extract of a Chilean stinging plant, Lithraea caustic (Litre). As Litre plant is known to induce DTH, we used a murine B16 melanoma model to compare the topical and intratumor efficacy of LExT with synthetic DTH inducers (DPCP and 2,4-dinitrochlorobenzene [DNCB]). LExt contained mainly long chain catechols and sesquiterpenes. The intratumor injection of LExT induced a significant delay in tumor growth, similarly topical treatment of an established tumor with 0.1% LExT ointment induced a growth delay and even tumor regression in 15% of treated animals. No significant changes were observed on the T-cell populations associated to LExT treatment, and neither DNCB nor DPCP were capable to induce none of the LExT-induced antitumoral effects. Interestingly, our results indicate that LExT induces an antitumor response against melanoma in a mouse model and could bring a new –and affordable- treatment for melanoma in humans.
Collapse
Affiliation(s)
- Claudia Robles-Planells
- Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, Chile.,Centro de Biotecnología Acuícola, Universidad de Santiago de Chile, Santiago, Chile
| | - Sofia A Michelson
- Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, Chile
| | - Javier Mena
- Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, Chile
| | - Daniela Escrig
- Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, Chile
| | - Juan L Rojas
- Escuela de Medicina, Facultad de Ciencias Médicas, Universidad de Santiago de Chile, Santiago, Chile
| | - Giselle Sanchez-Guerrero
- Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, Chile
| | - Ronny Hernández
- Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, Chile
| | - Carlos Barrera-Avalos
- Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, Chile.,Centro de Biotecnología Acuícola, Universidad de Santiago de Chile, Santiago, Chile
| | - Leonel E Rojo
- Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, Chile.,Centro de Biotecnología Acuícola, Universidad de Santiago de Chile, Santiago, Chile
| | - Daniela Sauma
- Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
| | - Alexis M Kalergis
- Millennium Institute Immunology and Immunotherapy, FOCIS Center of Excellence, Facultad de Ciencias Biológicas, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Mónica Imarai
- Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, Chile.,Centro de Biotecnología Acuícola, Universidad de Santiago de Chile, Santiago, Chile
| | | | - Carolina A Robles
- Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, Chile
| | - Elías Leiva-Salcedo
- Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, Chile
| | - Rocio Santander
- Departamento de Ciencias del Ambiente, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, Chile
| | - Alejandro Escobar
- Laboratorio Biología Celular y Molecular, Instituto de Investigación en Ciencias Odontológicas, Facultad de Odontología, Universidad de Chile, Santiago, Chile
| | - Claudio Acuña-Castillo
- Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, Chile.,Centro de Biotecnología Acuícola, Universidad de Santiago de Chile, Santiago, Chile
| |
Collapse
|
10
|
Rojas-Sepúlveda D, Tittarelli A, Gleisner MA, Ávalos I, Pereda C, Gallegos I, González FE, López MN, Butte JM, Roa JC, Fluxá P, Salazar-Onfray F. Tumor lysate-based vaccines: on the road to immunotherapy for gallbladder cancer. Cancer Immunol Immunother 2018; 67:1897-1910. [PMID: 29600445 PMCID: PMC6244977 DOI: 10.1007/s00262-018-2157-5] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 03/26/2018] [Indexed: 12/14/2022]
Abstract
Immunotherapy based on checkpoint blockers has proven survival benefits in patients with melanoma and other malignancies. Nevertheless, a significant proportion of treated patients remains refractory, suggesting that in combination with active immunizations, such as cancer vaccines, they could be helpful to improve response rates. During the last decade, we have used dendritic cell (DC) based vaccines where DCs loaded with an allogeneic heat-conditioned melanoma cell lysate were tested in a series of clinical trials. In these studies, 60% of stage IV melanoma DC-treated patients showed immunological responses correlating with improved survival. Further studies showed that an essential part of the clinical efficacy was associated with the use of conditioned lysates. Gallbladder cancer (GBC) is a high-incidence malignancy in South America. Here, we evaluated the feasibility of producing effective DCs using heat-conditioned cell lysates derived from gallbladder cancer cell lines (GBCCL). By characterizing nine different GBCCLs and several fresh tumor tissues, we found that they expressed some tumor-associated antigens such as CEA, MUC-1, CA19-9, Erb2, Survivin, and several carcinoembryonic antigens. Moreover, heat-shock treatment of GBCCLs induced calreticulin translocation and release of HMGB1 and ATP, both known to act as danger signals. Monocytes stimulated with combinations of conditioned lysates exhibited a potent increase of DC-maturation markers. Furthermore, conditioned lysate-matured DCs were capable of strongly inducing CD4+ and CD8+ T cell activation, in both allogeneic and autologous cell co-cultures. Finally, in vitro stimulated CD8+ T cells recognize HLA-matched GBCCLs. In summary, GBC cell lysate-loaded DCs may be considered for future immunotherapy approaches.
Collapse
Affiliation(s)
- Daniel Rojas-Sepúlveda
- Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Independencia 1027, building H, Third floor, 8380453, Santiago, Chile.,Millennium Institute on Immunology and Immunotherapy, Universidad de Chile, 8380453, Santiago, Chile.,Faculty of Science, Universidad San Sebastián, Lota 2465, 7510157, Santiago, Chile
| | - Andrés Tittarelli
- Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Independencia 1027, building H, Third floor, 8380453, Santiago, Chile.,Millennium Institute on Immunology and Immunotherapy, Universidad de Chile, 8380453, Santiago, Chile
| | - María Alejandra Gleisner
- Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Independencia 1027, building H, Third floor, 8380453, Santiago, Chile.,Millennium Institute on Immunology and Immunotherapy, Universidad de Chile, 8380453, Santiago, Chile
| | - Ignacio Ávalos
- Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Independencia 1027, building H, Third floor, 8380453, Santiago, Chile.,Millennium Institute on Immunology and Immunotherapy, Universidad de Chile, 8380453, Santiago, Chile
| | - Cristián Pereda
- Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Independencia 1027, building H, Third floor, 8380453, Santiago, Chile.,Millennium Institute on Immunology and Immunotherapy, Universidad de Chile, 8380453, Santiago, Chile
| | - Iván Gallegos
- Pathological Anatomy Service, Clinic Hospital, Universidad de Chile, 8380456, Santiago, Chile
| | - Fermín Eduardo González
- Millennium Institute on Immunology and Immunotherapy, Universidad de Chile, 8380453, Santiago, Chile.,Department of Conservative Dentistry, Faculty of Dentistry, Universidad de Chile, 8380492, Santiago, Chile
| | - Mercedes Natalia López
- Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Independencia 1027, building H, Third floor, 8380453, Santiago, Chile.,Millennium Institute on Immunology and Immunotherapy, Universidad de Chile, 8380453, Santiago, Chile
| | - Jean Michel Butte
- Department of Surgery, Fundación Arturo López Pérez, Institute of Oncology, 7500921, Santiago, Chile
| | - Juan Carlos Roa
- Department of Pathology, School of Medicine, Pontificia Universidad Católica de Chile, 8330023, Santiago, Chile.,Center for Investigation in Translational Oncology (CITO), Advanced Center for Chronic Diseases (ACCDiS), School of Medicine, Pontificia Universidad Católica de Chile, 8330023, Santiago, Chile
| | - Paula Fluxá
- Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Independencia 1027, building H, Third floor, 8380453, Santiago, Chile.,Millennium Institute on Immunology and Immunotherapy, Universidad de Chile, 8380453, Santiago, Chile
| | - Flavio Salazar-Onfray
- Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Independencia 1027, building H, Third floor, 8380453, Santiago, Chile. .,Millennium Institute on Immunology and Immunotherapy, Universidad de Chile, 8380453, Santiago, Chile.
| |
Collapse
|
11
|
Proteomic Identification of Heat Shock-Induced Danger Signals in a Melanoma Cell Lysate Used in Dendritic Cell-Based Cancer Immunotherapy. J Immunol Res 2018; 2018:3982942. [PMID: 29744371 PMCID: PMC5878886 DOI: 10.1155/2018/3982942] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Revised: 11/28/2017] [Accepted: 12/11/2017] [Indexed: 12/17/2022] Open
Abstract
Autologous dendritic cells (DCs) loaded with cancer cell-derived lysates have become a promising tool in cancer immunotherapy. During the last decade, we demonstrated that vaccination of advanced melanoma patients with autologous tumor antigen presenting cells (TAPCells) loaded with an allogeneic heat shock- (HS-) conditioned melanoma cell-derived lysate (called TRIMEL) is able to induce an antitumor immune response associated with a prolonged patient survival. TRIMEL provides not only a broad spectrum of potential melanoma-associated antigens but also danger signals that are crucial in the induction of a committed mature DC phenotype. However, potential changes induced by heat conditioning on the proteome of TRIMEL are still unknown. The identification of newly or differentially expressed proteins under defined stress conditions is relevant for understanding the lysate immunogenicity. Here, we characterized the proteomic profile of TRIMEL in response to HS treatment. A quantitative label-free proteome analysis of over 2800 proteins was performed, with 91 proteins that were found to be regulated by HS treatment: 18 proteins were overexpressed and 73 underexpressed. Additionally, 32 proteins were only identified in the HS-treated TRIMEL and 26 in non HS-conditioned samples. One protein from the overexpressed group and two proteins from the HS-exclusive group were previously described as potential damage-associated molecular patterns (DAMPs). Some of the HS-induced proteins, such as haptoglobin, could be also considered as DAMPs and candidates for further immunological analysis in the establishment of new putative danger signals with immunostimulatory functions.
Collapse
|
12
|
Gleisner MA, Navarrete M, Hofmann F, Salazar-Onfray F, Tittarelli A. Mind the Gaps in Tumor Immunity: Impact of Connexin-Mediated Intercellular Connections. Front Immunol 2017; 8:1067. [PMID: 28919895 PMCID: PMC5585150 DOI: 10.3389/fimmu.2017.01067] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Accepted: 08/16/2017] [Indexed: 12/22/2022] Open
Abstract
Gap junctions (GJs)-mediated intercellular communications (GJICs) are connexin (Cx)-formed plasma membrane channels that allow for the passage of small molecules between adjacent cells, and are involved in several physiopathological processes, including immune responses against cancer. In general, tumor cells are poorly coupled through GJs, mainly due to low Cx expression or reduced channel activity, suggesting that Cxs may have tumor suppressor roles. However, more recent data indicate that Cxs and/or GJICs may also in some cases promote tumor progression. This dual role of Cx channels in tumor outcome may be due, at least partially, to the fact that GJs not only interconnect cells from the same type, such as cancer cells, but also promote the intercellular communication of tumor cells with different types of cells from their microenvironment, and such diverse intercellular interactions have distinctive impact on tumor development. For example, whereas GJ-mediated interactions among tumor cells and microglia have been implicated in promotion of tumor growth, tumor cells delivery to dendritic cells of antigenic peptides through GJs have been associated with enhanced immune-mediated tumor elimination. In this review, we provide an updated overview on the role of GJICs in tumor immunity, focusing on the pro-tumor and antitumor effect of GJs occurring among tumor and immune cells. Accumulated data suggest that GJICs may act as tumor suppressors or enhancers depending on whether tumor cells interact predominantly with antitumor immune cells or with stromal cells. The complex modulation of immune-tumor cell GJICs should be taken into consideration in order to potentiate current cancer immunotherapies.
Collapse
Affiliation(s)
- María Alejandra Gleisner
- Disciplinary Program of Immunology, Faculty of Medicine, Institute of Biomedical Sciences, Universidad de Chile, Santiago, Chile.,Faculty of Medicine, Millennium Institute on Immunology and Immunotherapy, Universidad de Chile, Santiago, Chile
| | - Mariela Navarrete
- Disciplinary Program of Immunology, Faculty of Medicine, Institute of Biomedical Sciences, Universidad de Chile, Santiago, Chile.,Faculty of Medicine, Millennium Institute on Immunology and Immunotherapy, Universidad de Chile, Santiago, Chile
| | - Francisca Hofmann
- Disciplinary Program of Immunology, Faculty of Medicine, Institute of Biomedical Sciences, Universidad de Chile, Santiago, Chile.,Faculty of Medicine, Millennium Institute on Immunology and Immunotherapy, Universidad de Chile, Santiago, Chile
| | - Flavio Salazar-Onfray
- Disciplinary Program of Immunology, Faculty of Medicine, Institute of Biomedical Sciences, Universidad de Chile, Santiago, Chile.,Faculty of Medicine, Millennium Institute on Immunology and Immunotherapy, Universidad de Chile, Santiago, Chile
| | - Andrés Tittarelli
- Disciplinary Program of Immunology, Faculty of Medicine, Institute of Biomedical Sciences, Universidad de Chile, Santiago, Chile.,Faculty of Medicine, Millennium Institute on Immunology and Immunotherapy, Universidad de Chile, Santiago, Chile
| |
Collapse
|
13
|
Markov OV, Mironova NL, Vlassov VV, Zenkova MA. Antitumor Vaccines Based on Dendritic Cells: From Experiments using Animal Tumor Models to Clinical Trials. Acta Naturae 2017; 9:27-38. [PMID: 29104773 PMCID: PMC5662271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Indexed: 11/07/2022] Open
Abstract
The routine methods used to treat oncological diseases have a number of drawbacks, including non-specific action and severe side effects for patients. Furthermore, tumor diseases are associated with a suppression of the immune system that often leads to the inefficiency of standard treatment methods. The development of novel immunotherapeutic approaches having specific antitumor action and that activate the immune system is of crucial importance. Vaccines based on dendritic cells (DCs) loaded with tumor antigens ex vivo that can activate antitumor cytotoxic T-cell responses stand out among different antitumor immunotherapeutic approaches. This review is focused on analyzing different methods of DC-based vaccine preparation and current research in antitumor DC-based vaccines using animal tumor models and in clinical trials.
Collapse
Affiliation(s)
- O. V. Markov
- Institute of Chemical Biology and Fundamental Medicine SB RAS, Lavrentieva Ave. 8, Novosibirsk, 630090 , Russia
| | - N. L. Mironova
- Institute of Chemical Biology and Fundamental Medicine SB RAS, Lavrentieva Ave. 8, Novosibirsk, 630090 , Russia
| | - V. V. Vlassov
- Institute of Chemical Biology and Fundamental Medicine SB RAS, Lavrentieva Ave. 8, Novosibirsk, 630090 , Russia
| | - M. A. Zenkova
- Institute of Chemical Biology and Fundamental Medicine SB RAS, Lavrentieva Ave. 8, Novosibirsk, 630090 , Russia
| |
Collapse
|
14
|
Tao Z, Li S, Ichim TE, Yang J, Riordan N, Yenugonda V, Babic I, Kesari S. Cellular immunotherapy of cancer: an overview and future directions. Immunotherapy 2017; 9:589-606. [DOI: 10.2217/imt-2016-0086] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The clinical success of checkpoint inhibitors has led to a renaissance of interest in cancer immunotherapies. In particular, the possibility of ex vivo expanding autologous lymphocytes that specifically recognize tumor cells has attracted much research and clinical trial interest. In this review, we discuss the historical background of tumor immunotherapy using cell-based approaches, and provide some rationale for overcoming current barriers to success of autologous immunotherapy. An overview of adoptive transfer of lymphocytes, tumor infiltrating lymphocytes and dendritic cell therapies is provided. We conclude with discussing the possibility of gene-manipulating immune cells in order to augment therapeutic activity, including silencing of the immune-suppressive zinc finger orphan nuclear receptor, NR2F6, as an attractive means of overcoming tumor-associated immune suppression.
Collapse
Affiliation(s)
- Ziqi Tao
- The Affiliated XuZhou Center Hospital of Nanjing University of Chinese Medicine, The Affiliated XuZhou Hospital of Medical College of Southeast University, Jiangsu, China
| | - Shuang Li
- Department of Endocrinology, the Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | | | - Junbao Yang
- Department of Translational Neurosciences and Neurotherapeutics, Pacific Neuroscience Institute, John Wayne Cancer Institute, Providence Saint John’s Health Center, Santa Monica, CA 90404, USA
| | - Neil Riordan
- Medistem Panama, Inc., City of Knowledge, Clayton, Republic of Panama
| | - Venkata Yenugonda
- Department of Translational Neurosciences and Neurotherapeutics, Pacific Neuroscience Institute, John Wayne Cancer Institute, Providence Saint John’s Health Center, Santa Monica, CA 90404, USA
| | - Ivan Babic
- Department of Translational Neurosciences and Neurotherapeutics, Pacific Neuroscience Institute, John Wayne Cancer Institute, Providence Saint John’s Health Center, Santa Monica, CA 90404, USA
| | - Santosh Kesari
- Department of Translational Neurosciences and Neurotherapeutics, Pacific Neuroscience Institute, John Wayne Cancer Institute, Providence Saint John’s Health Center, Santa Monica, CA 90404, USA
- John Wayne Cancer Institute, 2200 Santa Monica Blvd, Santa Monica, CA 90404, USA
| |
Collapse
|
15
|
Adjuvant Autologous Melanoma Vaccine for Macroscopic Stage III Disease: Survival, Biomarkers, and Improved Response to CTLA-4 Blockade. J Immunol Res 2016; 2016:8121985. [PMID: 27294163 PMCID: PMC4887652 DOI: 10.1155/2016/8121985] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Revised: 03/30/2016] [Accepted: 04/17/2016] [Indexed: 11/30/2022] Open
Abstract
Background. There is not yet an agreed adjuvant treatment for melanoma patients with American Joint Committee on Cancer stages III B and C. We report administration of an autologous melanoma vaccine to prevent disease recurrence. Patients and Methods. 126 patients received eight doses of irradiated autologous melanoma cells conjugated to dinitrophenyl and mixed with BCG. Delayed type hypersensitivity (DTH) response to unmodified melanoma cells was determined on the vaccine days 5 and 8. Gene expression analysis was performed on 35 tumors from patients with good or poor survival. Results. Median overall survival was 88 months with a 5-year survival of 54%. Patients attaining a strong DTH response had a significantly better (p = 0.0001) 5-year overall survival of 75% compared with 44% in patients without a strong response. Gene expression array linked a 50-gene signature to prognosis, including a cluster of four cancer testis antigens: CTAG2 (NY-ESO-2), MAGEA1, SSX1, and SSX4. Thirty-five patients, who received an autologous vaccine, followed by ipilimumab for progressive disease, had a significantly improved 3-year survival of 46% compared with 19% in nonvaccinated patients treated with ipilimumab alone (p = 0.007). Conclusion. Improved survival in patients attaining a strong DTH and increased response rate with subsequent ipilimumab suggests that the autologous vaccine confers protective immunity.
Collapse
|
16
|
Bol KF, Schreibelt G, Gerritsen WR, de Vries IJM, Figdor CG. Dendritic Cell-Based Immunotherapy: State of the Art and Beyond. Clin Cancer Res 2016; 22:1897-906. [PMID: 27084743 DOI: 10.1158/1078-0432.ccr-15-1399] [Citation(s) in RCA: 272] [Impact Index Per Article: 30.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Accepted: 02/23/2016] [Indexed: 11/16/2022]
Abstract
Dendritic cell (DC) vaccination in cancer patients aims to induce or augment an effective antitumor immune response against tumor antigens and was first explored in a clinical trial in the 1990s. More than two decades later, numerous clinical trials have been performed or are ongoing with a wide variety of DC subsets, culture protocols, and treatment regimens. The safety of DC vaccination and its ability to induce antitumor responses have clearly been established; however, although scattered patients with long-term benefit were reported, DC vaccines have not yet fulfilled their promise, perhaps mainly due to the lack of large-scale well-conducted phase II/III trials. To allow meaningful multicenter phase III trials, the production of DC vaccines should be standardized between centers which is now becoming feasible. To improve the efficacy of DC-based immunotherapy, it could be combined with other treatments.
Collapse
Affiliation(s)
- Kalijn F Bol
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Nijmegen, the Netherlands. Department of Medical Oncology, Radboud University Medical Centre, Nijmegen, the Netherlands
| | - Gerty Schreibelt
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Nijmegen, the Netherlands
| | - Winald R Gerritsen
- Department of Medical Oncology, Radboud University Medical Centre, Nijmegen, the Netherlands
| | - I Jolanda M de Vries
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Nijmegen, the Netherlands. Department of Medical Oncology, Radboud University Medical Centre, Nijmegen, the Netherlands
| | - Carl G Figdor
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Nijmegen, the Netherlands.
| |
Collapse
|
17
|
Wojas-Turek J, Szczygieł A, Kicielińska J, Rossowska J, Piasecki E, Pajtasz-Piasecka E. Treatment with cyclophosphamide supported by various dendritic cell-based vaccines induces diversification in CD4⁺ T cell response against MC38 colon carcinoma. Int J Oncol 2015; 48:493-505. [PMID: 26648160 PMCID: PMC4725454 DOI: 10.3892/ijo.2015.3278] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Accepted: 09/26/2015] [Indexed: 12/18/2022] Open
Abstract
The present study shows that an application of cyclophosphamide (CY) supported by dendritic cell (DC)-based vaccines affected differentiation of the activity of CD4+ T cell subpopulations accompanied by an alteration in CD8+ cell number. Vaccines were composed of bone marrow-derived DCs activated with tumor cell lysate (BM-DC/TAgTNF-α) and/or genetically modified DCs of JAWS II line (JAWS II/ Neo or JAWS II/IL-2 cells). Compared to untreated or CY-treated mice, the combined treatment of MC38 colon carcinoma-bearing mice resulted in significant tumor growth inhibition associated with an increase in influx of CD4+ and CD8+ T cells into tumor tissue. Whereas, the division of these cell population in spleen was not observed. Depending on the nature of DC-based vaccines and number of their applications, both tumor infiltrating cells and spleen cells were able to produce various amount of IFN-γ, IL-4 and IL-10 after mitogenic ex vivo stimulation. The administration of CY followed by BM-DC/TAgTNF-α and genetically modified JAWS II cells, increased the percentage of CD4+T-bet+ and CD4+GATA3+ cells and decreased the percentage of CD4+RORγt+ and CD4+FoxP3+ lymphocytes. However, the most intensive response against tumor was noted after the ternary treatment with CY + BM-DC/TAgTNF-α + JAWS II/IL-2 cells. Thus, the administration of various DC-based vaccines was responsible for generation of the diversified antitumor response. These findings demonstrate that the determination of the size of particular CD4+ T cell subpopulations may become a prognostic factor and be the basis for future development of anticancer therapy.
Collapse
Affiliation(s)
- Justyna Wojas-Turek
- Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wroclaw, Poland
| | - Agnieszka Szczygieł
- Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wroclaw, Poland
| | - Jagoda Kicielińska
- Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wroclaw, Poland
| | - Joanna Rossowska
- Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wroclaw, Poland
| | - Egbert Piasecki
- Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wroclaw, Poland
| | - Elżbieta Pajtasz-Piasecka
- Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wroclaw, Poland
| |
Collapse
|
18
|
Kosmaczewska A, Ciszak L, Swierkot J, Szteblich A, Kosciow K, Frydecka I. Exogenous IL-2 controls the balance in Th1, Th17, and Treg cell distribution in patients with progressive rheumatoid arthritis treated with TNF-alpha inhibitors. Inflammation 2015; 38:765-74. [PMID: 25145773 PMCID: PMC4344954 DOI: 10.1007/s10753-014-9987-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Interleukin-2 (IL-2) has been suggested to control Treg/Th17 balance. Recently, we reported a relationship of rheumatoid arthritis (RA) activity/progression with irreversible systemic Treg and Th1 defects including serum IL-2 shortage. Herein, we explore the role of in vitro stimulation with rIL-2 in the observed immune alterations reversal. Patients with stable or progressive RA were assigned to methotrexate (MTX) group or to TNF-alpha inhibitors (iTNF) group, respectively. Flow cytometric analyses were performed before and after 6 months of treatment. Circulating Th1, Th17, and Treg cells were determined before and after 72-h culture with anti-CD3 + rIL-2. Before therapy, 72-h stimulation restored recently observed phenotypic Th cell alterations, except for the enriched Th17 subset normalized as late as after therapy in all patients. Under 6-month therapy, anti-CD3 stimulation changed the Th cell distribution only in progressive RA; despite Th1 enrichment, it revealed Treg population defects, which were completely reversed by exogenous IL-2 added to the stimulating culture. Our paper shows that in aggressive RA patients exhibiting serum IL-2 shortage despite iTNF therapy, exogenous rIL-2 is capable of promoting Treg differentiation affected by chronic activation, thus supporting its use in the combined strategy of biologic treatment of the progressive form of RA.
Collapse
Affiliation(s)
- Agata Kosmaczewska
- Department of Immunopathology, Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, R. Weigla St. 12, 53-114, Wroclaw, Poland,
| | | | | | | | | | | |
Collapse
|
19
|
González FE, Gleisner A, Falcón-Beas F, Osorio F, López MN, Salazar-Onfray F. Tumor cell lysates as immunogenic sources for cancer vaccine design. Hum Vaccin Immunother 2015; 10:3261-9. [PMID: 25625929 DOI: 10.4161/21645515.2014.982996] [Citation(s) in RCA: 112] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Autologous dendritic cells (DCs) loaded with tumor-associated antigens (TAAs) are a promising immunological tool for cancer therapy. These stimulate the antitumor response and immunological memory generation. Nevertheless, many patients remain refractory to DC approaches. Antigen (Ag) delivery to DCs is relevant to vaccine success, and antigen peptides, tumor-associated proteins, tumor cells, autologous tumor lysates, and tumor-derived mRNA have been tested as Ag sources. Recently, DCs loaded with allogeneic tumor cell lysates were used to induce a potent immunological response. This strategy provides a reproducible pool of almost all potential Ags suitable for patient use, independent of MHC haplotypes or autologous tumor tissue availability. However, optimizing autologous tumor cell lysate preparation is crucial to enhancing efficacy. This review considers the role of cancer cell-derived lysates as a relevant source of antigens and as an activating factor for ex vivo therapeutic DCs capable of responding to neoplastic cells. These promising therapies are associated with the prolonged survival of advanced cancer patients.
Collapse
Key Words
- AM, Cytokine-activated monocytes
- Ags, Antigens
- CDAMs, Cell death-associated molecules
- CRT, Calreticulin
- CTLs, Cytotoxic T lymphocytes
- DAMPs
- DAMPs, Damage-associated molecular patterns
- DCs, Dendritic cells
- DTH, Delayed-type IV hypersensitivity
- GM-CSF, Granulocyte and macrophage colony stimulating factor
- HMGB1, High-mobility group box 1 protein
- HSPs, Heat shock proteins
- ICD, Immunogenic cell death
- MAAs, Melanoma-associated antigens
- MHC, Major histocompatibility complex
- MM, Malignant melanoma
- NKT, Natural killer T cell
- PAMPs, Pathogen-associated molecular patterns
- PBMCs, Peripheral blood mononuclear cells
- PCCL, Prostate cancer cell lysate
- PD1, Programmed cell death protein 1
- PRRs, Pattern recognition receptors
- PSA, Prostate specific antigen
- RAGE, Receptor for advanced glycation endproducts
- SNPs, Single nucleotide polymorphisms
- TAAs, Tumor-associated antigens
- TAPCells, Tumor antigen presenting cells
- TCRs, T cell receptors
- TLRs, Toll-like receptors
- TNF, Tumor necrosis factor
- TRIMEL, Allogeneic melanoma cell lysate
- TRIPRO, Allogeneic prostate cell lysate
- Toll-like receptors
- Tregs, Regulatory T lymphocytes
- cancer immunotherapy
- dendritic cells
- immunogenic cell death
Collapse
Affiliation(s)
- Fermín E González
- a Millennium Institute on Immunology and Immunotherapy; Institute of Biomedical Sciences; Faculty of Medicine ; University of Chile ; Santiago , Chile
| | | | | | | | | | | |
Collapse
|
20
|
Pellegrino P, Falvella FS, Cheli S, Perrotta C, Clementi E, Radice S. The role of Toll-like receptor 4 polymorphisms in vaccine immune response. THE PHARMACOGENOMICS JOURNAL 2015; 16:96-101. [PMID: 25823688 DOI: 10.1038/tpj.2015.21] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Revised: 12/22/2014] [Accepted: 01/28/2015] [Indexed: 12/22/2022]
Abstract
Toll-like receptors (TLRs) are a class of pattern recognition receptors that are deputed to recognise a range of molecular structures in pathogens. One of the most studied members of this family is the TLR4, which is essential for the signalling of lipopolysaccharide. The gene encoding for TLR4 is highly polymorphic and this genetic variability may explain in part the interindividual variability observed in several clinical setting, including the response to vaccination. Herein, we review and systematise the available scientific evidence about the effect of TLR4 polymorphisms on vaccine response, including approved prophylactic, new therapeutic cancer vaccines and recently approved vaccine adjuvants. Data reviewed in this analysis indicate that TLR4 polymorphisms significantly affect vaccine response. If these results are confirmed by further analyses, the use of these genetic biomarkers may become a useful tool to tailor vaccination in specific subsets of patients.
Collapse
Affiliation(s)
- P Pellegrino
- Unit of Clinical Pharmacology, Department of Biomedical and Clinical Sciences, University Hospital "Luigi Sacco", Università di Milano, Milan, Italy
| | - F S Falvella
- Unit of Clinical Pharmacology, Department of Biomedical and Clinical Sciences, University Hospital "Luigi Sacco", Università di Milano, Milan, Italy
| | - S Cheli
- Unit of Clinical Pharmacology, Department of Biomedical and Clinical Sciences, University Hospital "Luigi Sacco", Università di Milano, Milan, Italy
| | - C Perrotta
- Unit of Clinical Pharmacology, Department of Biomedical and Clinical Sciences, University Hospital "Luigi Sacco", Università di Milano, Milan, Italy
| | - E Clementi
- Scientific Institute, IRCCS E. Medea, Lecco, Italy.,Unit of Clinical Pharmacology, Department of Biomedical and Clinical Sciences, Consiglio Nazionale delle Ricerche Institute of Neuroscience, University Hospital "Luigi Sacco", Università di Milano, Milan, Italy
| | - S Radice
- Unit of Clinical Pharmacology, Department of Biomedical and Clinical Sciences, University Hospital "Luigi Sacco", Università di Milano, Milan, Italy
| |
Collapse
|
21
|
Teulings HE, Limpens J, Jansen SN, Zwinderman AH, Reitsma JB, Spuls PI, Luiten RM. Vitiligo-like depigmentation in patients with stage III-IV melanoma receiving immunotherapy and its association with survival: a systematic review and meta-analysis. J Clin Oncol 2015; 33:773-81. [PMID: 25605840 DOI: 10.1200/jco.2014.57.4756] [Citation(s) in RCA: 453] [Impact Index Per Article: 45.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
PURPOSE Vitiligo-like depigmentation in patients with melanoma may be associated with more favorable clinical outcome. We conducted a systematic review of patients with stage III to IV melanoma treated with immunotherapy to determine the cumulative incidence of vitiligo-like depigmentation and the prognostic value of vitiligo development on survival. METHODS We systemically searched and selected all studies on melanoma immunotherapy that reported on autoimmune toxicity and/or vitiligo between 1995 and 2013. Methodologic quality of each study was appraised using adapted criteria for systematic reviews in prognostic studies. Random-effect models were used to calculate summary estimates of the cumulative incidence of vitiligo-like depigmentation across studies. The prognostic value of vitiligo-like depigmentation on survival outcome was assessed using random-effects Cox regression survival analyses. RESULTS One hundred thirty-seven studies were identified comprising 139 treatment arms (11 general immune stimulation, 84 vaccine, 28 antibody-based, and 16 adoptive transfer) including a total of 5,737 patients. The overall cumulative incidence of vitiligo was 3.4% (95% CI, 2.5% to 4.5%). In 27 studies reporting individual patient data, vitiligo development was significantly associated with both progression-free-survival (hazard ratio [HR], 0.51; 95% CI, 0.32 to 0.82; P < .005) and overall survival (HR, 0.25; 95% CI, 0.10 to 0.61; P < .003), indicating that these patients have two to four times less risk of disease progression and death, respectively, compared with patients without vitiligo development. CONCLUSION Although vitiligo occurs only in a low percentage of patients with melanoma treated with immunotherapy, our findings suggest clear survival benefit in these patients. Awareness of vitiligo induction in patients with melanoma is important as an indicator of robust antimelanoma immunity and associated improved survival.
Collapse
Affiliation(s)
- Hansje-Eva Teulings
- Hansje-Eva Teulings, Jacqueline Limpens, Sophia N. Jansen, Aeilko H. Zwinderman, Johannes B. Reitsma, Phyllis I. Spuls, and Rosalie M. Luiten, Academic Medical Centre, University of Amsterdam, Amsterdam; Johannes B. Reitsma, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, the Netherlands.
| | - Jacqueline Limpens
- Hansje-Eva Teulings, Jacqueline Limpens, Sophia N. Jansen, Aeilko H. Zwinderman, Johannes B. Reitsma, Phyllis I. Spuls, and Rosalie M. Luiten, Academic Medical Centre, University of Amsterdam, Amsterdam; Johannes B. Reitsma, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Sophia N Jansen
- Hansje-Eva Teulings, Jacqueline Limpens, Sophia N. Jansen, Aeilko H. Zwinderman, Johannes B. Reitsma, Phyllis I. Spuls, and Rosalie M. Luiten, Academic Medical Centre, University of Amsterdam, Amsterdam; Johannes B. Reitsma, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Aeilko H Zwinderman
- Hansje-Eva Teulings, Jacqueline Limpens, Sophia N. Jansen, Aeilko H. Zwinderman, Johannes B. Reitsma, Phyllis I. Spuls, and Rosalie M. Luiten, Academic Medical Centre, University of Amsterdam, Amsterdam; Johannes B. Reitsma, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Johannes B Reitsma
- Hansje-Eva Teulings, Jacqueline Limpens, Sophia N. Jansen, Aeilko H. Zwinderman, Johannes B. Reitsma, Phyllis I. Spuls, and Rosalie M. Luiten, Academic Medical Centre, University of Amsterdam, Amsterdam; Johannes B. Reitsma, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Phyllis I Spuls
- Hansje-Eva Teulings, Jacqueline Limpens, Sophia N. Jansen, Aeilko H. Zwinderman, Johannes B. Reitsma, Phyllis I. Spuls, and Rosalie M. Luiten, Academic Medical Centre, University of Amsterdam, Amsterdam; Johannes B. Reitsma, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Rosalie M Luiten
- Hansje-Eva Teulings, Jacqueline Limpens, Sophia N. Jansen, Aeilko H. Zwinderman, Johannes B. Reitsma, Phyllis I. Spuls, and Rosalie M. Luiten, Academic Medical Centre, University of Amsterdam, Amsterdam; Johannes B. Reitsma, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, the Netherlands
| |
Collapse
|
22
|
Datta J, Terhune JH, Lowenfeld L, Cintolo JA, Xu S, Roses RE, Czerniecki BJ. Optimizing dendritic cell-based approaches for cancer immunotherapy. THE YALE JOURNAL OF BIOLOGY AND MEDICINE 2014; 87:491-518. [PMID: 25506283 PMCID: PMC4257036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Dendritic cells (DC) are professional antigen-presenting cells uniquely suited for cancer immunotherapy. They induce primary immune responses, potentiate the effector functions of previously primed T-lymphocytes, and orchestrate communication between innate and adaptive immunity. The remarkable diversity of cytokine activation regimens, DC maturation states, and antigen-loading strategies employed in current DC-based vaccine design reflect an evolving, but incomplete, understanding of optimal DC immunobiology. In the clinical realm, existing DC-based cancer immunotherapy efforts have yielded encouraging but inconsistent results. Despite recent U.S. Federal and Drug Administration (FDA) approval of DC-based sipuleucel-T for metastatic castration-resistant prostate cancer, clinically effective DC immunotherapy as monotherapy for a majority of tumors remains a distant goal. Recent work has identified strategies that may allow for more potent "next-generation" DC vaccines. Additionally, multimodality approaches incorporating DC-based immunotherapy may improve clinical outcomes.
Collapse
Affiliation(s)
- Jashodeep Datta
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Julia H. Terhune
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania,Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland
| | - Lea Lowenfeld
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Jessica A. Cintolo
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Shuwen Xu
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Robert E. Roses
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Brian J. Czerniecki
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania,To whom all correspondence should be addressed: Brian J. Czerniecki, MD PhD, Department of Surgery, University of Pennsylvania Perelman School of Medicine, 3400 Civic Center Drive, Philadelphia, PA 19104; Tele: 215-615-1696; Fax: 215-615-0555;
| |
Collapse
|
23
|
González FE, Ortiz C, Reyes M, Dutzan N, Patel V, Pereda C, Gleisner MA, López MN, Gutkind JS, Salazar-Onfray F. Melanoma cell lysate induces CCR7 expression and in vivo migration to draining lymph nodes of therapeutic human dendritic cells. Immunology 2014; 142:396-405. [PMID: 24673602 DOI: 10.1111/imm.12264] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2013] [Revised: 01/29/2014] [Accepted: 01/30/2014] [Indexed: 02/04/2023] Open
Abstract
We have previously reported a novel method for the production of tumour-antigen-presenting cells (referred to as TAPCells) that are currently being used in cancer therapy, using an allogeneic melanoma-derived cell lysate (referred to as TRIMEL) as an antigen provider and activation factor. It was recently demonstrated that TAPCell-based immunotherapy induces T-cell-mediated immune responses resulting in improved long-term survival of stage IV melanoma patients. Clinically, dendritic cell (DC) migration from injected sites to lymph nodes is an important requirement for an effective anti-tumour immunization. This mobilization of DCs is mainly driven by the C-C chemokine receptor type 7 (CCR7), which is up-regulated on mature DCs. Using flow cytometry and immunohistochemistry, we investigated if TRIMEL was capable of inducing the expression of the CCR7 on TAPCells and enhancing their migration in vitro, as well as their in vivo relocation to lymph nodes in an ectopic xenograft animal model. Our results confirmed that TRIMEL induces a phenotypic maturation and increases the expression of surface CCR7 on melanoma patient-derived DCs, and also on the monocytic/macrophage cell line THP-1. Moreover, in vitro assays showed that TRIMEL-stimulated DCs and THP-1 cells were capable of migrating specifically in the presence of the CCR7 ligand CCL19. Finally, we demonstrated that TAPCells could migrate in vivo from the injection site into the draining lymph nodes. This work contributes to an increased understanding of the biology of DCs produced ex vivo allowing the design of new strategies for effective DC-based vaccines for treating aggressive melanomas.
Collapse
Affiliation(s)
- Fermín E González
- Millennium Institute on Immunology and Immunotherapy, University of Chile, Santiago, Chile; Department of Conservative Dentistry, Faculty of Dentistry, University of Chile, Santiago, Chile
| | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Hargadon KM. Murine and Human Model Systems for the Study of Dendritic Cell Immunobiology. Int Rev Immunol 2014; 35:85-115. [DOI: 10.3109/08830185.2014.952413] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
25
|
Anguille S, Smits EL, Lion E, van Tendeloo VF, Berneman ZN. Clinical use of dendritic cells for cancer therapy. Lancet Oncol 2014; 15:e257-67. [PMID: 24872109 DOI: 10.1016/s1470-2045(13)70585-0] [Citation(s) in RCA: 537] [Impact Index Per Article: 48.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Since the mid-1990s, dendritic cells have been used in clinical trials as cellular mediators for therapeutic vaccination of patients with cancer. Dendritic cell-based immunotherapy is safe and can induce antitumour immunity, even in patients with advanced disease. However, clinical responses have been disappointing, with classic objective tumour response rates rarely exceeding 15%. Paradoxically, findings from emerging research indicate that dendritic cell-based vaccination might improve survival, advocating implementation of alternative endpoints to assess the true clinical potency of dendritic cell-based vaccination. We review the clinical effectiveness of dendritic cell-based vaccine therapy in melanoma, prostate cancer, malignant glioma, and renal cell carcinoma, and summarise the most important lessons from almost two decades of clinical studies of dendritic cell-based immunotherapy in these malignant disorders. We also address how the specialty is evolving, and which new therapeutic concepts are being translated into clinical trials to leverage the clinical effectiveness of dendritic cell-based cancer immunotherapy. Specifically, we discuss two main trends: the implementation of the next-generation dendritic cell vaccines that have improved immunogenicity, and the emerging paradigm of combination of dendritic cell vaccination with other cancer therapies.
Collapse
Affiliation(s)
- Sébastien Anguille
- Center for Cell Therapy and Regenerative Medicine, Antwerp University Hospital, Edegem, Belgium; Laboratory of Experimental Hematology, Tumor Immunology Group (TIGR), Vaccine and Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Faculty of Medicine and Health Sciences, Antwerp, Belgium.
| | - Evelien L Smits
- Center for Cell Therapy and Regenerative Medicine, Antwerp University Hospital, Edegem, Belgium; Center for Oncological Research, University of Antwerp, Faculty of Medicine and Health Sciences, Antwerp, Belgium
| | - Eva Lion
- Laboratory of Experimental Hematology, Tumor Immunology Group (TIGR), Vaccine and Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Faculty of Medicine and Health Sciences, Antwerp, Belgium
| | - Viggo F van Tendeloo
- Laboratory of Experimental Hematology, Tumor Immunology Group (TIGR), Vaccine and Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Faculty of Medicine and Health Sciences, Antwerp, Belgium
| | - Zwi N Berneman
- Center for Cell Therapy and Regenerative Medicine, Antwerp University Hospital, Edegem, Belgium; Laboratory of Experimental Hematology, Tumor Immunology Group (TIGR), Vaccine and Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Faculty of Medicine and Health Sciences, Antwerp, Belgium
| |
Collapse
|
26
|
Escobar A, Aguirre A, Guzmán MA, González R, Catalán D, Acuña-Castillo C, Larrondo M, López M, Pesce B, Rolland J, O’Hehir R, Aguillón JC. Tolerogenic dendritic cells derived from donors with natural rubber latex allergy modulate allergen-specific T-cell responses and IgE production. PLoS One 2014; 9:e85930. [PMID: 24465795 PMCID: PMC3899084 DOI: 10.1371/journal.pone.0085930] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2013] [Accepted: 12/03/2013] [Indexed: 11/25/2022] Open
Abstract
Natural rubber latex (NRL; Hevea brasiliensis) allergy is an IgE-mediated reaction to latex proteins. When latex glove exposure is the main sensitizing agent, Hev b 5 is one of the major allergens. Dendritic cells (DC), the main antigen presenting cells, modulated with pharmacological agents can restore tolerance in several experimental models, including allergy. In the current study, we aimed to generate DC with tolerogenic properties from NRL-allergic patients and evaluate their ability to modulate allergen-specific T and B cell responses. Here we show that dexamethasone-treated DC (dxDC) differentiated into a subset of DC, characterized by low expression of MHC class II, CD40, CD80, CD86 and CD83 molecules. Compared with LPS-matured DC, dxDC secreted lower IL-12 and higher IL-10 after CD40L activation, and induced lower alloantigenic T cell proliferation. We also show that dxDC pulsed with the dominant Hev b 5 T-cell epitope peptide, Hev b 546–65, inhibited both proliferation of Hev b 5-specific T-cell lines and the production of Hev b 5-specific IgE. Additionally, dxDC induced a subpopulation of IL-10-producing regulatory T cells that suppressed proliferation of Hev b 5-primed T cells. In conclusion, dxDC generated from NRL-allergic patients can modulate allergen-specific T-cell responses and IgE production, supporting their potential use in allergen-specific immunotherapy.
Collapse
Affiliation(s)
- Alejandro Escobar
- Research Institute of Dental Science, Faculty of Dentistry, University of Chile, Santiago, Chile
- * E-mail: (AE); (JCA)
| | - Adam Aguirre
- Department of Pharmacy, Faculty of Chemistry, Catholic University of Chile, Santiago, Chile
| | - María Antonieta Guzmán
- Allergy Center, Clinical Hospital of University of Chile, University of Chile, Santiago, Chile
| | - Rodrigo González
- Blood bank Clinical Hospital of University of Chile, University of Chile, Santiago, Chile
| | - Diego Catalán
- Immunology Program, Faculty of Medicine, University of Chile Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
| | - Claudio Acuña-Castillo
- Department of Biology, Faculty of Chemistry and Biology, University of Santiago, Santiago, Chile
| | - Milton Larrondo
- Blood bank Clinical Hospital of University of Chile, University of Chile, Santiago, Chile
| | - Mercedes López
- Immunology Program, Faculty of Medicine, University of Chile Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
| | - Barbara Pesce
- Immunology Program, Faculty of Medicine, University of Chile Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
| | - Jennifer Rolland
- Department of Immunology, AMREP, Monash University, Melbourne, Australia
| | - Robyn O’Hehir
- Department of Immunology, AMREP, Monash University, Melbourne, Australia
| | - Juan Carlos Aguillón
- Immunology Program, Faculty of Medicine, University of Chile Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- * E-mail: (AE); (JCA)
| |
Collapse
|
27
|
Reyes D, Salazar L, Espinoza E, Pereda C, Castellón E, Valdevenito R, Huidobro C, Inés Becker M, Lladser A, López MN, Salazar-Onfray F. Tumour cell lysate-loaded dendritic cell vaccine induces biochemical and memory immune response in castration-resistant prostate cancer patients. Br J Cancer 2013; 109:1488-97. [PMID: 23989944 PMCID: PMC3777003 DOI: 10.1038/bjc.2013.494] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2013] [Revised: 07/24/2013] [Accepted: 07/30/2013] [Indexed: 12/16/2022] Open
Abstract
Background: Recently, we produced a tumour antigen-presenting cells (TAPCells) vaccine using a melanoma cell lysate, called TRIMEL, as an antigen source and an activation factor. Tumour antigen-presenting cells induced immunological responses and increased melanoma patient survival. Herein, we investigated the effect of TAPCells loaded with prostate cancer cell lysates (PCCL) as an antigen source, and TRIMEL as a dendritic cell (DC) activation factor; which were co-injected with the Concholepas concholepas haemocyanin (CCH) as an adjuvant on castration-resistant prostate cancer (CRPC) patients. Methods: The lysate mix capacity, for inducing T-cell activation, was analysed by flow cytometry and Elispot. Delayed-type hypersensitivity (DTH) reaction against PCCL, frequency of CD8+ memory T cells (Tm) in blood and prostate-specific antigen (PSA) levels in serum were measured in treated patients. Results: The lysate mix induced functional mature DCs that were capable of activating PCCL-specific T cells. No relevant adverse reactions were observed. Six out of 14 patients showed a significant decrease in levels of PSA. DTH+ patients showed a prolonged PSA doubling-time after treatment. Expansion of functional central and effector CD8+ Tm were detected. Conclusion: Treatment of CRPC patients with lysate-loaded TAPCells and CCH as an adjuvant is safe: generating biochemical and memory immune responses. However, the limited number of cases requires confirmation in a phase II clinical trial.
Collapse
Affiliation(s)
- D Reyes
- 1] Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago 8380453, Chile [2] Service of Urology, University of Chile Clinical Hospital, Santiago 8380453, Chile [3] Millennium Institute on Immunology and Immunotherapy, University of Chile, Santiago 8380453 Chile
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Durán-Aniotz C, Segal G, Salazar L, Pereda C, Falcón C, Tempio F, Aguilera R, González R, Pérez C, Tittarelli A, Catalán D, Nervi B, Larrondo M, Salazar-Onfray F, López MN. The immunological response and post-treatment survival of DC-vaccinated melanoma patients are associated with increased Th1/Th17 and reduced Th3 cytokine responses. Cancer Immunol Immunother 2013; 62:761-72. [PMID: 23242374 PMCID: PMC11028820 DOI: 10.1007/s00262-012-1377-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2012] [Accepted: 11/14/2012] [Indexed: 12/20/2022]
Abstract
INTRODUCTION Immunization with autologous dendritic cells (DCs) loaded with a heat shock-conditioned allogeneic melanoma cell lysate caused lysate-specific delayed type hypersensitivity (DTH) reactions in a number of patients. These responses correlated with a threefold prolonged long-term survival of DTH(+) with respect to DTH(-) unresponsive patients. Herein, we investigated whether the immunological reactions associated with prolonged survival were related to dissimilar cellular and cytokine responses in blood. MATERIALS AND METHODS Healthy donors and melanoma patient's lymphocytes obtained from blood before and after vaccinations and from DTH biopsies were analyzed for T cell population distribution and cytokine release. RESULTS/DISCUSSION Peripheral blood lymphocytes from melanoma patients have an increased proportion of Th3 (CD4(+) TGF-β(+)) regulatory T lymphocytes compared with healthy donors. Notably, DTH(+) patients showed a threefold reduction of Th3 cells compared with DTH(-) patients after DCs vaccine treatment. Furthermore, DCs vaccination resulted in a threefold augment of the proportion of IFN-γ releasing Th1 cells and in a twofold increase of the IL-17-producing Th17 population in DTH(+) with respect to DTH(-) patients. Increased Th1 and Th17 cell populations in both blood and DTH-derived tissues suggest that these profiles may be related to a more effective anti-melanoma response. CONCLUSIONS Our results indicate that increased proinflammatory cytokine profiles are related to detectable immunological responses in vivo (DTH) and to prolonged patient survival. Our study contributes to the understanding of immunological responses produced by DCs vaccines and to the identification of follow-up markers for patient outcome that may allow a closer individual monitoring of patients.
Collapse
Affiliation(s)
- Claudia Durán-Aniotz
- Faculty of Medicine, Institute of Biomedical Sciences, University of Chile, 8380453, Santiago, Chile.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Tittarelli A, González FE, Pereda C, Mora G, Muñoz L, Saffie C, García T, Díaz D, Falcón C, Hermoso M, López MN, Salazar-Onfray F. Toll-like receptor 4 gene polymorphism influences dendritic cell in vitro function and clinical outcomes in vaccinated melanoma patients. Cancer Immunol Immunother 2012; 61:2067-77. [PMID: 22552381 PMCID: PMC11029707 DOI: 10.1007/s00262-012-1268-7] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2012] [Accepted: 04/11/2012] [Indexed: 12/22/2022]
Abstract
Toll-like receptor 4 (TLR4) is expressed on dendritic cells (DCs), sensing environmental danger molecules that induce their activation and maturation. Recently, we reported a method for the production of therapeutic DCs against melanoma, called tumor antigen-presenting cells (TAPCells), using a heat-shocked allogeneic melanoma cell lysate (TRIMEL) as an activation factor and antigen provider. Since TRIMEL contains endogenous TLR4 ligands, we evaluated the role of TLR4 in TAPCells differentiation by antibody neutralization and the association of a Tlr4 polymorphism (896A/G) (Asp299Gly), determined by PCR-RFLP, with the in vitro activation capacity and the clinical outcome of TAPCells-vaccinated patients. Antibody blocking of monocyte TLR4 inhibited surface expression, determined by flow cytometry, of the major histocompatibility complex class I, CCR7, CD80, CD83 and CD86 on TAPCells, reduced interleukin (IL)-6 and tumor necrosis factor -α gene expression evaluated by qRT-PCR, and also inhibited the TAPCells-mediated interferon-γ (IFN-γ) secretion of melanoma-specific CD8(+) T cells determined by ELISpot (p < 0.01). Moreover, CD8(+) T-cell activation capacity was significantly reduced in TAPCells bearing the TLR4 Asp299Gly receptor (p < 0.05). Finally, TAPCells-vaccinated stage-IV melanoma patients bearing the Tlr4 896G allele showed a shortened post-therapy median survival rate compared with those carrying the Tlr4 896A allele (p < 0.05; log-rank test). Our results indicate that TLR4 is a key receptor for the tumor lysate-mediated in vitro generation of clinically efficient antigen-presenting cells. Further analysis of patients included in different vaccine protocols is necessary for definitively establishing a role for TLR4 polymorphism in clinical responses.
Collapse
Affiliation(s)
- Andrés Tittarelli
- Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, 8380453, Santiago Chile, Chile.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
NAKAMURA IZUMI, KANAZAWA MASASHI, SATO YU, IRISAWA ATSUSHI, TAKAGI TADAYUKI, OGATA TAKASHI, KASHIMURA SHOGO, KENJO AKIRA, SUZUKI HIROYUKI, SHIBATA MASAHIKO, SHIMURA TATSUO, OHIRA HIROMASA, GOTO MITSUKAZU, TAKENOSHITA SEIICHI, OHTO HITOSHI. CLINICAL EVALUATION OF DENDRITIC CELLS VACCINATION FOR ADVANCED CANCER PATIENTS AT FUKUSHIMA MEDICAL UNIVERSITY. Fukushima J Med Sci 2012; 58:40-8. [DOI: 10.5387/fms.58.40] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|
31
|
Chiang CLL, Maier DA, Kandalaft LE, Brennan AL, Lanitis E, Ye Q, Levine BL, Czerniecki BJ, Powell DJ, Coukos G. Optimizing parameters for clinical-scale production of high IL-12 secreting dendritic cells pulsed with oxidized whole tumor cell lysate. J Transl Med 2011; 9:198. [PMID: 22082029 PMCID: PMC3283529 DOI: 10.1186/1479-5876-9-198] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2011] [Accepted: 11/14/2011] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Dendritic cells (DCs) are the most potent antigen-presenting cell population for activating tumor-specific T cells. Due to the wide range of methods for generating DCs, there is no common protocol or defined set of criteria to validate the immunogenicity and function of DC vaccines. METHODS Monocyte-derived DCs were generated during 4 days of culture with recombinant granulocyte-macrophage colony stimulating factor and interleukin-4, and pulsed with tumor lysate produced by hypochlorous acid oxidation of tumor cells. Different culture parameters for clinical-scale DC preparation were investigated, including: 1) culture media; 2) culture surface; 3) duration of activating DCs with lipopolysaccharide (LPS) and interferon (IFN)-gamma; 4) method of DC harvest; and 5) cryomedia and final DC product formulation. RESULTS DCs cultured in CellGenix DC media containing 2% human AB serum expressed higher levels of maturation markers following lysate-loading and maturation compared to culturing with serum-free CellGenix DC media or AIM-V media, or 2% AB serum supplemented AIM-V media. Nunclon™Δ surface, but not Corning(®) tissue-culture treated surface and Corning(®) ultra-low attachment surface, were suitable for generating an optimal DC phenotype. Recombinant trypsin resulted in reduced major histocompatibility complex (MHC) Class I and II expression on mature lysate-loaded DCs, however presentation of MHC Class I peptides by DCs was not impaired and cell viability was higher compared to cell scraping. Preservation of DCs with an infusible cryomedia containing Plasma-Lyte A, dextrose, sodium chloride injection, human serum albumin, and DMSO yielded higher cell viability compared to using human AB serum containing 10% DMSO. Finally, activating DCs for 16 hours with LPS and IFN-γ stimulated robust mixed leukocyte reactions (MLRs), and high IL-12p70 production in vitro that continued for 24 hours after the cryopreserved DCs were thawed and replated in fresh media. CONCLUSIONS This study examined criteria including DC phenotype, viability, IL-12p70 production and the ability to stimulate MLR as metrics of whole oxidized tumor lysate-pulsed DC immunogenicity and functionality. Development and optimization of this unique method is now being tested in a clinical trial of autologous oxidized tumor lysate-pulsed DC in clinical-scale in recurrent ovarian, primary peritoneal or fallopian tube cancer (NCT01132014).
Collapse
Affiliation(s)
- Cheryl L-L Chiang
- Ovarian Cancer Research Center, University of Pennsylvania, Philadelphia, 19104, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Rahma OE, Ashtar E, Czystowska M, Szajnik ME, Wieckowski E, Bernstein S, Herrin VE, Shams MA, Steinberg SM, Merino M, Gooding W, Visus C, Deleo AB, Wolf JK, Bell JG, Berzofsky JA, Whiteside TL, Khleif SN. A gynecologic oncology group phase II trial of two p53 peptide vaccine approaches: subcutaneous injection and intravenous pulsed dendritic cells in high recurrence risk ovarian cancer patients. Cancer Immunol Immunother 2011; 61:373-84. [PMID: 21927947 DOI: 10.1007/s00262-011-1100-9] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2011] [Accepted: 08/16/2011] [Indexed: 12/22/2022]
Abstract
PURPOSE Peptide antigens have been administered by different approaches as cancer vaccine therapy, including direct injection or pulsed onto dendritic cells; however, the optimal delivery method is still debatable. In this study, we describe the immune response elicited by two vaccine approaches using the wild-type (wt) p53 vaccine. EXPERIMENTAL DESIGN Twenty-one HLA-A2.1 patients with stage III, IV, or recurrent ovarian cancer overexpressing the p53 protein with no evidence of disease were treated in two cohorts. Arm A received SC wt p53:264-272 peptide admixed with Montanide and GM-CSF. Arm B received wt p53:264-272 peptide-pulsed dendritic cells IV. Interleukin-2 (IL-2) was administered to both cohorts in alternative cycles. RESULTS Nine of 13 patients (69%) in arm A and 5 of 6 patients (83%) in arm B developed an immunologic response as determined by ELISPOT and tetramer assays. The vaccine caused no serious systemic side effects. IL-2 administration resulted in grade 3 and 4 toxicities in both arms and directly induced the expansion of T regulatory cells. The median overall survival was 40.8 and 29.6 months for arm A and B, respectively; the median progression-free survival was 4.2 and. 8.7 months, respectively. CONCLUSION We found that using either vaccination approach generates comparable specific immune responses against the p53 peptide with minimal toxicity. Accordingly, our findings suggest that the use of less demanding SC approach may be as effective. Furthermore, the use of low-dose SC IL-2 as an adjuvant might have interfered with the immune response. Therefore, it may not be needed in future trials.
Collapse
Affiliation(s)
- Osama E Rahma
- Vaccine Branch, CCR, NCI, 41 Medlars Dr., Building 41 Room B900, Bethesda, MD 20892, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Autologous peripheral blood mononuclear cell recognition of autologous proliferating tumor cells in the context of a patient-specific vaccine trial. J Biomed Biotechnol 2011; 2011:635850. [PMID: 21541189 PMCID: PMC3085493 DOI: 10.1155/2011/635850] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2010] [Revised: 02/10/2011] [Accepted: 02/27/2011] [Indexed: 11/17/2022] Open
Abstract
Metastatic melanoma patients who were treated with patient-specific vaccines consisting of dendritic cells loaded with autologous tumor cells had a 5-year survival of over 50%. Enzyme-linked immunospot (ELISPOT) has been used to detect antigen reactive T cells as a means of determining immune response. We wished to determine whether IFN-gamma secretion in an ELISPOT assay was prognostic or predictive for survival following treatment. Peripheral blood mononuclear cells (PBMCs) collected at weeks 0 and 4 were evaluated by ELISPOT assay for response to autologous tumor cells. Overall, there was slight increase in the number of tumor reactive lymphocytes from week 0 to week 4. Using >5 spots/100 K PBMC as the cutoff, a log-rank analysis revealed only a slight statistical significance in overall survival for patients who lacked tumor reactive PBMCs at week 4. The sensitivity of ELISPOT in the context of patient-specific cellular vaccines is unclear.
Collapse
|
34
|
Aguilera R, Saffie C, Tittarelli A, González FE, Ramírez M, Reyes D, Pereda C, Hevia D, García T, Salazar L, Ferreira A, Hermoso M, Mendoza-Naranjo A, Ferrada C, Garrido P, López MN, Salazar-Onfray F. Heat-Shock Induction of Tumor-Derived Danger Signals Mediates Rapid Monocyte Differentiation into Clinically Effective Dendritic Cells. Clin Cancer Res 2011; 17:2474-83. [DOI: 10.1158/1078-0432.ccr-10-2384] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
35
|
Lotem M, Machlenkin A, Hamburger T, Nissan A, Kadouri L, Frankenburg S, Gimmon Z, Elias O, David IB, Kuznetz A, Shiloni E, Peretz T. Autologous Melanoma Vaccine Induces Antitumor and Self-Reactive Immune Responses That Affect Patient Survival and Depend on MHC Class II Expression on Vaccine Cells. Clin Cancer Res 2009; 15:4968-77. [DOI: 10.1158/1078-0432.ccr-08-3320] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
36
|
Cohen S, Haimovich J, Hollander N. Dendritic cell-based therapeutic vaccination against myeloma: vaccine formulation determines efficacy against light chain myeloma. THE JOURNAL OF IMMUNOLOGY 2009; 182:1667-73. [PMID: 19155516 DOI: 10.4049/jimmunol.182.3.1667] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Multiple myeloma is an incurable plasma cell malignancy. Immunotherapy in myeloma patients had limited success to date. We have previously demonstrated that dendritic cells (DCs) pulsed with autologous Ig Id induced Id-reactive CD8(+) T cells and protection against a myeloma tumor challenge. In this work, we studied the therapeutic efficacy of chemotherapy combined with different formulations of DC-based vaccines in mice bearing large plasma cell tumors. The comparative study demonstrated that s.c. injection of DCs loaded with Id coupled to keyhole limpet hemocyanin, s.c. injection of DCs loaded with irradiated tumor cells, and intratumoral injection of naive DCs were similarly effective in mediating tumor regression and long-term survival. However, whereas the Id-keyhole limpet hemocyanin-DC vaccine was inefficient against myeloma cells that lost expression of the Ig H chain, intratumoral injection of naive DCs and s.c. injection of DCs loaded with irradiated tumor cells were highly effective against cells producing L chains only. This may be of particular importance for patients with L chain myeloma. Given that T cells respond primarily to peptides derived from H chain CDRs, attempts to treat L chain disease with myeloma protein-pulsed DCs may be futile. Vaccination with tumor cell-loaded DCs may, however, induce an effective antitumor response.
Collapse
Affiliation(s)
- Sharon Cohen
- Department of Human Microbiology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | | | | |
Collapse
|
37
|
López MN, Pereda C, Segal G, Muñoz L, Aguilera R, González FE, Escobar A, Ginesta A, Reyes D, González R, Mendoza-Naranjo A, Larrondo M, Compán A, Ferrada C, Salazar-Onfray F. Prolonged Survival of Dendritic Cell–Vaccinated Melanoma Patients Correlates With Tumor-Specific Delayed Type IV Hypersensitivity Response and Reduction of Tumor Growth Factor β-Expressing T Cells. J Clin Oncol 2009; 27:945-52. [DOI: 10.1200/jco.2008.18.0794] [Citation(s) in RCA: 112] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PurposeThe aim of this work was to assess immunologic response, disease progression, and post-treatment survival of melanoma patients vaccinated with autologous dendritic cells (DCs) pulsed with a novel allogeneic cell lysate (TRIMEL) derived from three melanoma cell lines.Patients and MethodsForty-three stage IV and seven stage III patients were vaccinated four times with TRIMEL/DC vaccine. Specific delayed type IV hypersensitivity (DTH) reaction, ex vivo cytokine production, and regulatory T-cell populations were determined. Overall survival and disease progression rates were analyzed using Kaplan-Meier curves and compared with historical records.ResultsThe overall survival for stage IV patients was 15 months. More than 60% of patients showed DTH-positive reaction against the TRIMEL. Stage IV/DTH-positive patients displayed a median survival of 33 months compared with 11 months observed for DTH-negative patients (P = .0014). All stage III treated patients were DTH positive and remained alive and tumor free for a median follow-up period of 48 months (range, 33 to 64 months). DTH-positive patients showed a marked reduction in the proportion of CD4+ transforming growth factor (TGF) β+ regulatory T cells compared to DTH-negative patients (1.54% v 5.78%; P < .0001).ConclusionOur findings strongly suggest that TRIMEL-pulsed DCs provide a standardized and widely applicable source of melanoma antigens, very effective in evoking antimelanoma immune response. To our knowledge, this is the first report describing a correlation between vaccine-induced reduction of CD4+TGFβ+ regulatory T cells and in vivo antimelanoma immune response associated to improved patient survival and disease stability.
Collapse
Affiliation(s)
- Mercedes N. López
- From the Millennium Nucleus on Immunology and Immunotherapy, Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile; Research Support Office, Clinical Hospital of the University of Chile, Santiago; and the Regional Hospital of Concepción, Concepción, Chile
| | - Cristian Pereda
- From the Millennium Nucleus on Immunology and Immunotherapy, Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile; Research Support Office, Clinical Hospital of the University of Chile, Santiago; and the Regional Hospital of Concepción, Concepción, Chile
| | - Gabriela Segal
- From the Millennium Nucleus on Immunology and Immunotherapy, Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile; Research Support Office, Clinical Hospital of the University of Chile, Santiago; and the Regional Hospital of Concepción, Concepción, Chile
| | - Leonel Muñoz
- From the Millennium Nucleus on Immunology and Immunotherapy, Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile; Research Support Office, Clinical Hospital of the University of Chile, Santiago; and the Regional Hospital of Concepción, Concepción, Chile
| | - Raquel Aguilera
- From the Millennium Nucleus on Immunology and Immunotherapy, Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile; Research Support Office, Clinical Hospital of the University of Chile, Santiago; and the Regional Hospital of Concepción, Concepción, Chile
| | - Fermín E. González
- From the Millennium Nucleus on Immunology and Immunotherapy, Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile; Research Support Office, Clinical Hospital of the University of Chile, Santiago; and the Regional Hospital of Concepción, Concepción, Chile
| | - Alejandro Escobar
- From the Millennium Nucleus on Immunology and Immunotherapy, Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile; Research Support Office, Clinical Hospital of the University of Chile, Santiago; and the Regional Hospital of Concepción, Concepción, Chile
| | - Alexandra Ginesta
- From the Millennium Nucleus on Immunology and Immunotherapy, Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile; Research Support Office, Clinical Hospital of the University of Chile, Santiago; and the Regional Hospital of Concepción, Concepción, Chile
| | - Diego Reyes
- From the Millennium Nucleus on Immunology and Immunotherapy, Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile; Research Support Office, Clinical Hospital of the University of Chile, Santiago; and the Regional Hospital of Concepción, Concepción, Chile
| | - Rodrigo González
- From the Millennium Nucleus on Immunology and Immunotherapy, Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile; Research Support Office, Clinical Hospital of the University of Chile, Santiago; and the Regional Hospital of Concepción, Concepción, Chile
| | - Ariadna Mendoza-Naranjo
- From the Millennium Nucleus on Immunology and Immunotherapy, Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile; Research Support Office, Clinical Hospital of the University of Chile, Santiago; and the Regional Hospital of Concepción, Concepción, Chile
| | - Milton Larrondo
- From the Millennium Nucleus on Immunology and Immunotherapy, Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile; Research Support Office, Clinical Hospital of the University of Chile, Santiago; and the Regional Hospital of Concepción, Concepción, Chile
| | - Alvaro Compán
- From the Millennium Nucleus on Immunology and Immunotherapy, Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile; Research Support Office, Clinical Hospital of the University of Chile, Santiago; and the Regional Hospital of Concepción, Concepción, Chile
| | - Carlos Ferrada
- From the Millennium Nucleus on Immunology and Immunotherapy, Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile; Research Support Office, Clinical Hospital of the University of Chile, Santiago; and the Regional Hospital of Concepción, Concepción, Chile
| | - Flavio Salazar-Onfray
- From the Millennium Nucleus on Immunology and Immunotherapy, Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile; Research Support Office, Clinical Hospital of the University of Chile, Santiago; and the Regional Hospital of Concepción, Concepción, Chile
| |
Collapse
|
38
|
Capitini CM, Fry TJ, Mackall CL. Cytokines as Adjuvants for Vaccine and Cellular Therapies for Cancer. AMERICAN JOURNAL OF IMMUNOLOGY 2009; 5:65-83. [PMID: 20182648 PMCID: PMC2826803 DOI: 10.3844/ajisp.2009.65.83] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
PROBLEM STATEMENT: The development of a potent vaccine that can help treat tumors resistant to conventional cytotoxic therapies remains elusive. While part of the problem may be that trials have focused on patients with bulky residual disease, the desire to maximize responses to the vaccine remains. APPROACH: The gamma(c) family of cytokines offer a unique opportunity to support the expansion and effector potential of vaccine-responding T-cells, as well as stimulate other effectors, such as natural killer (NK) cells, to become activated. RESULTS: Combining vaccines with cytokines seems logical but can bring unwanted toxicity, as has been observed with interleukin (IL)-2. In addition, the nonspecific activation or expansion of unwanted cell subsets, such as regulatory T-cells, can contribute to global immunosuppression and limit vaccine responses. The development of IL-7 and IL-21 for the clinic offers the promise of enhancing anti-tumor responses but with far less systemic toxicity and no expansion of regulatory T cells. Preclinical studies demonstrate that IL-15 could also improve T-cell, and especially NK-cell, responses as well. CONCLUSIONS/RECOMMENDATIONS: Future work should expand the use of vaccines with IL-7, IL-21 and hopefully IL-15 in high-risk patients, and consider treatment while in a state of minimal residual disease to maximize benefit. Identifying tumors that can signal through gamma(c) cytokines will also be essential so that induction of relapse will be avoided.
Collapse
Affiliation(s)
- Christian M. Capitini
- Immunology Section, Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892
| | - Terry J. Fry
- Immunology Section, Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892
- Center for Cancer and Blood Disorders, Children’s National Medical Center, Washington, DC 20010
| | - Crystal L. Mackall
- Immunology Section, Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892
| |
Collapse
|
39
|
Salazar L, Aravena O, Abello P, Escobar A, Contreras-Levicoy J, Rojas-Colonelli N, Catalán D, Aguirre A, Zúñiga R, Pesce B, González C, Cepeda R, Cuchacovich M, Molina MC, Salazar-Onfray F, Delgado M, Toes RE, Aguillón JC. Modulation of established murine collagen-induced arthritis by a single inoculation of short-term lipopolysaccharide-stimulated dendritic cells. Ann Rheum Dis 2008; 67:1235-41. [PMID: 18056756 DOI: 10.1136/ard.2007.072199] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
BACKGROUND The use of regulatory or immature dendritic cells (DCs) as tools for modulating experimental rheumatoid arthritis is very recent. Tumour necrosis factor (TNF)-stimulated DCs have been shown to restore tolerance in experimental autoimmune encephalomyelitis and collagen-induced arthritis (CIA). OBJECTIVE We investigated the capacity of short-term lipopolysaccharide (LPS)-stimulated DCs pulsed with type II collagen (CII) to induce tolerance against established CIA. METHODS Bone marrow-derived DCs were generated in the presence of granulocyte monocyte colony-stimulating factor (GM-CSF). After CIA induction, mice were injected at day 35 with a single dose of 4- or 24-h LPS-stimulated DCs that had been loaded with CII (4hLPS/CII/DCs or 24hLPS/CII/DCs). Arthritis progression was monitored by clinical and histological evaluations. RESULTS Flow cytometry of 4hLPS/CII/DCs showed intermediate CD40 and CD86 expression, lower than that of 24hLPS/CII/DCs (fully mature) and higher than that of CII/DCs (immature). A functional assay showed that 4hLPS/CII/DCs display increased endocytosis ability with respect to 24hLPS/CII/DCs, indicating a semimature state. The single inoculation of 4hLPS/CII/DCs in mice with established CIA reduced disease severity significantly over time. Histological evaluation of mice treated with 4hLPS/CII/DCs revealed diminished inflammatory synovitis, cartilage damage and fibrosis. Co-cultures of DCs with splenocytes from CIA mice showed that collagen-specific interferon (IFN)gamma production was dramatically inhibited by 4hLPS/CII/DCs. 4hLPS/CII/DCs were high IL10 producers, which could explain the inhibition of arthritis progression in mice receiving this treatment because neither antibodies nor regulatory CD4+CD25+Foxp3+ T lymphocytes were demonstrated to be involved. CONCLUSION Short-term LPS-modulated DCs inoculation interferes with CIA progression when loaded with CII.
Collapse
Affiliation(s)
- L Salazar
- Programa Disciplinario de Inmunología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Phase Ib trial assessing autologous, tumor-pulsed dendritic cells as a vaccine administered with or without IL-2 in patients with metastatic melanoma. J Immunother 2008; 31:591-8. [PMID: 18528294 DOI: 10.1097/cji.0b013e31817fd90b] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Twenty-four subjects with metastatic melanoma were treated on a randomized Phase Ib trial evaluating an autologous tumor lysate-pulsed dendritic cell (DC) vaccine with or without interleukin (IL)-2. The vaccine consisted of autologous DCs obtained from peripheral blood mononuclear cells (PBMCs) cultured in granulocyte macrophage-colony stimulating factor and IL-4 then pulsed with autologous tumor cell lysate and keyhole limpet hemocyanin (KLH). The primary end points of the trial were safety and immune response to vaccine. Subjects were randomized to vaccine administered every other week times 3, vaccine x 3 followed by low-dose IL-2, or vaccine x 3 followed by high-dose IL-2. Immune response was monitored pretreatment and at 2 and 4 weeks after the third vaccine administration. Disease evaluation was performed at 4 weeks after the third vaccination. Therapy was well tolerated with no local vaccine toxicity greater than grade 1 in any arm. IL-2 toxicity was as expected without additional toxicity from the addition of IL-2 to vaccine. Immune response defined as delayed-type hypersensitivity, PBMC interferon-gamma enzyme-linked immunosorbent spot, and PBMC proliferation, to both autologous tumor and KLH were detected in all arms. Interferon-gamma enzyme-linked immunosorbent spot response to KLH (7 of 10 patients) and autologous tumor (4 of 10 patients) were also detected in subjects with available vaccine draining lymph node cells. There were no differences in immune response between treatment arms. No clinical responses were seen. Autologous tumor lysate-pulsed DC vaccine with or without IL-2 was well tolerated and immunogenic but failed to induce clinical response in patients with advanced melanoma.
Collapse
|
41
|
von Euw EM, Barrio MM, Furman D, Levy EM, Bianchini M, Peguillet I, Lantz O, Vellice A, Kohan A, Chacón M, Yee C, Wainstok R, Mordoh J. A phase I clinical study of vaccination of melanoma patients with dendritic cells loaded with allogeneic apoptotic/necrotic melanoma cells. Analysis of toxicity and immune response to the vaccine and of IL-10 -1082 promoter genotype as predictor of disease progression. J Transl Med 2008; 6:6. [PMID: 18221542 PMCID: PMC2265680 DOI: 10.1186/1479-5876-6-6] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2007] [Accepted: 01/25/2008] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Sixteen melanoma patients (1 stage IIC, 8 stage III, and 7 stage IV) were treated in a Phase I study with a vaccine (DC/Apo-Nec) composed of autologous dendritic cells (DCs) loaded with a mixture of apoptotic/necrotic allogeneic melanoma cell lines (Apo-Nec), to evaluate toxicity and immune responses. Also, IL-10 1082 genotype was analyzed in an effort to predict disease progression. METHODS PBMC were obtained after leukapheresis and DCs were generated from monocytes cultured in the presence of GM-CSF and IL-4 in serum-free medium. Immature DCs were loaded with gamma-irradiated Apo-Nec cells and injected id without adjuvant. Cohorts of four patients were given four vaccines each with 5, 10, 15, or 20 x 106 DC/Apo-Nec cell per vaccine, two weeks apart. Immune responses were measured by ELISpot and tetramer analysis. Il-10 genotype was measured by PCR and corroborated by IL-10 production by stimulated PBMC. RESULTS Immature DCs efficiently phagocytosed melanoma Apo-Nec cells and matured after phagocytosis as evidenced by increased expression of CD83, CD80, CD86, HLA class I and II, and 75.2 +/- 16% reduction in Dextran-FITC endocytosis. CCR7 was also up-regulated upon Apo-Nec uptake in DCs from all patients, and accordingly DC/Apo-Nec cells were able to migrate in vitro toward MIP-3 beta. The vaccine was well tolerated in all patients. The DTH score increased significantly in all patients after the first vaccination (Mann-Whitney Test, p < 0.05). The presence of CD8+T lymphocytes specific to gp100 and Melan A/MART-1 Ags was determined by ELISpot and tetramer analysis in five HLA-A*0201 patients before and after vaccination; one patient had stable elevated levels before and after vaccination; two increased their CD8 + levels, one had stable moderate and one had negligible levels. The analysis of IL-10 promoter -1082 polymorphism in the sixteen patients showed a positive correlation between AA genotype, accompanied by lower in vitro IL-10 production by stimulated PBMC, and faster melanoma progression after lymph nodes surgery (p = 0.04). With a mean follow-up of 49.5 months post-surgery, one stage IIC patient and 7/8 stage III patients remain NED but 7/7 stage IV patients have progressed. CONCLUSION We conclude that DC/Apo-Nec vaccine is safe, well tolerated and it may induce specific immunity against melanoma Ags. Patients with a low-producing IL-10 polymorphism appear to have a worst prognosis. TRIAL REGISTRATION Clinicaltrials.gov (NHI) NCT00515983.
Collapse
Affiliation(s)
- Erika M von Euw
- Centro de Investigaciones Oncológicas FUCA, Cramer 1180, (1426) Buenos Aires, Argentina.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Dendritic cells loaded with apoptotic antibody-coated tumor cells provide protective immunity against B-cell lymphoma in vivo. Blood 2007; 111:1504-11. [PMID: 17993615 DOI: 10.1182/blood-2007-03-080507] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The in vitro priming of tumor-specific T cells by dendritic cells (DCs) phagocytosing killed tumor cells can be augmented in the presence of antitumor monoclonal antibody (mAb). We investigated whether DCs phagocytosing killed lymphoma cells coated with tumor-specific antibody could elicit antitumor immunity in vivo. Irradiated murine 38C13 lymphoma cells were cocultured with bone marrow-derived DCs in the presence or absence of tumor-specific mAb. Mice vaccinated with DCs cocultured with mAb-coated tumor cells were protected from tumor challenge (60% long-term survival), whereas DCs loaded with tumor cells alone were much less effective. The opsonized whole tumor cell-DC vaccine elicited significantly better tumor protection than a traditional lymphoma idiotype (Id) protein vaccine, and in combination with chemotherapy could eradicate preexisting tumor. Moreover, the DC vaccine protected animals from both wild-type and Id-negative variant tumor cells, indicating that Id is not a major target of the induced tumor immunity. Protection was critically dependent upon CD8(+) T cells, with lesser contribution by CD4(+) T cells. Importantly, opsonized whole tumor cell-DC vaccination did not result in tissue-specific autoimmunity. Since opsonized whole tumor cell-DC and Id vaccines appear to target distinct tumor antigens, optimal antilymphoma immunity might be achieved by combining these approaches.
Collapse
|
43
|
Mendoza-Naranjo A, Saéz PJ, Johansson CC, Ramírez M, Mandakovic D, Pereda C, López MN, Kiessling R, Sáez JC, Salazar-Onfray F. Functional gap junctions facilitate melanoma antigen transfer and cross-presentation between human dendritic cells. THE JOURNAL OF IMMUNOLOGY 2007; 178:6949-57. [PMID: 17513744 DOI: 10.4049/jimmunol.178.11.6949] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Previously, we found that human dendritic cells (hDCs) pulsed with a melanoma cell lysate (MCL) and stimulated with TNF-alpha (MCL/TNF) acquire a mature phenotype in vitro and are able to trigger tumor-specific immune responses when they are used in melanoma immunotherapy in patients. In this study, we describe that MCL/TNF induces gap junction (GJ)-mediated intercellular communications and promotes melanoma Ag transfer between ex vivo produced hDCs from melanoma patients. hDCs also exhibit increased expression of the GJ-related protein connexin 43, which contributes to GJ plaque formation after MCL/TNF stimulation. The addition of GJ inhibitors suppresses intercellular tumor Ag transfer between hDCs, thus reducing melanoma-specific T cell activation. In summary, we demonstrate that MCL/TNF-stimulated hDCs can establish functional GJ channels that participate in melanoma Ag transfer, facilitating Ag cross-presentation and an effective dendritic cell-mediated melanoma-specific T cell response. These results suggest that GJs formed between hDCs used in cancer vaccination protocols could be essentials for the establishment of a more efficient antitumor response.
Collapse
Affiliation(s)
- Ariadna Mendoza-Naranjo
- Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Affiliation(s)
- Gavin Melmed
- Department of Internal Medicine, Baylor University Medical Center, Dallas, Texas, USA.
| |
Collapse
|
45
|
Ridolfi R, Petrini M, Fiammenghi L, Stefanelli M, Ridolfi L, Ballardini M, Migliori G, Riccobon A. Improved overall survival in dendritic cell vaccination-induced immunoreactive subgroup of advanced melanoma patients. J Transl Med 2006; 4:36. [PMID: 16914047 PMCID: PMC1562447 DOI: 10.1186/1479-5876-4-36] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2006] [Accepted: 08/16/2006] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND We present our experience of therapeutic vaccination using dendritic cells (DC) pulsed with autologous tumor antigens in patients with advanced melanoma. METHODS Twenty-one pretreated advanced melanoma patients were vaccinated with autologous DC pulsed with 100 microg/ml of autologous-tumor-lysate (ATL) or -homogenate (ATH) and 50 microg/ml of keyhole limpet hemocyanin (KLH). The first 8 patients were treated subcutaneously or intradermally with immature-DC (iDC) (range 4.5-82 x 10(6)) and the remaining 13 intradermally with in vitro matured DC (mDC) (range 1.2-26 x 10(6)). Subcutaneous interleukin-2 (3 x 10(6) IU) was administered from days 3 to 7 of each treatment cycle. RESULTS Three of the 8 iDC patients obtained stabilizations (SD), each of 6 months' duration. The 13 mDC patients showed 1 complete response (8 months), 1 partial response (3 months), 2 mixed responses (6 and 12 months) and 3 SD (9, 7+, and 3+ months). Overall responses (OR) were observed in 4/21 (19%) patients, or 4/13 (30.7%) considering mDC treatment only. 10/21 (47.6%) patients showed non progressive disease (NPD), with 7/13 (53.8%) cases of NPD for mDC-treated patients. No major toxicities were observed. The positive delayed-type hypersensitivity (DTH) test to ATL/ATH and/or KLH correlated with increased overall survival (OS). Median OS was 24 months (range 3-45) for the 10 DTH-positive (1 iDC and 9 mDC) and 5 months (range 3-14) for the 11 DTH-negative patients (P < 0.001). The in vitro evaluation of gamma IFN-secreting T-cells in 10 patients showed good correlation with both DTH (75%) and clinical outcome (70%). CONCLUSION Vaccination using DC pulsed with ATL/ATH and KLH in advanced melanoma patients is well tolerated and can induce a clinical response, especially when mDC are used. Successful immunization, verified by positive DTH, leads to longer survival.
Collapse
Affiliation(s)
- Ruggero Ridolfi
- Department of Medical Oncology, Morgagni-Pierantoni Hospital, Via Forlanini 34, 47100 Forlì, Italy
| | | | - Laura Fiammenghi
- Istituto Oncologico Romagnolo, Via Forlanini 34, 47100 Forlì, Italy
| | - Monica Stefanelli
- Department of Medical Oncology, Morgagni-Pierantoni Hospital, Via Forlanini 34, 47100 Forlì, Italy
| | - Laura Ridolfi
- Department of Medical Oncology, Morgagni-Pierantoni Hospital, Via Forlanini 34, 47100 Forlì, Italy
| | | | - Giuseppe Migliori
- Blood Transfusion Unit, Morgagni-Pierantoni Hospital, Via Forlanini 34, 47100 Forlì, Italy
| | - Angela Riccobon
- Department of Medical Oncology, Morgagni-Pierantoni Hospital, Via Forlanini 34, 47100 Forlì, Italy
| |
Collapse
|
46
|
Bellavite P, Ortolani R, Conforti A. Immunology and homeopathy. 3. Experimental studies on animal models. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2006; 3:171-86. [PMID: 16786046 PMCID: PMC1475939 DOI: 10.1093/ecam/nel016] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/05/2005] [Accepted: 03/16/2006] [Indexed: 01/23/2023]
Abstract
A search of the literature and the experiments carried out by the authors of this review show that there are a number of animal models where the effect of homeopathic dilutions or the principles of homeopathic medicine have been tested. The results relate to the immunostimulation by ultralow doses of antigens, the immunological models of the ‘simile’, the regulation of acute or chronic inflammatory processes and the use of homeopathic medicines in farming. The models utilized by different research groups are extremely etherogeneous and differ as the test medicines, the dilutions and the outcomes are concerned. Some experimental lines, particularly those utilizing mice models of immunomodulation and anti-inflammatory effects of homeopathic complex formulations, give support to a real effect of homeopathic high dilutions in animals, but often these data are of preliminary nature and have not been independently replicated. The evidence emerging from animal models is supporting the traditional ‘simile’ rule, according to which ultralow doses of compounds, that in high doses are pathogenic, may have paradoxically a protective or curative effect. Despite a few encouraging observational studies, the effectiveness of the homeopathic prevention or therapy of infections in veterinary medicine is not sufficiently supported by randomized and controlled trials.
Collapse
Affiliation(s)
- Paolo Bellavite
- Department of Scienze Morfologico-Biomediche, University of Verona, Piazza L.A. Scuro, 37134 Verona, Italy.
| | | | | |
Collapse
|
47
|
Zorn E, Nelson EA, Mohseni M, Porcheray F, Kim H, Litsa D, Bellucci R, Raderschall E, Canning C, Soiffer RJ, Frank DA, Ritz J. IL-2 regulates FOXP3 expression in human CD4+CD25+ regulatory T cells through a STAT-dependent mechanism and induces the expansion of these cells in vivo. Blood 2006; 108:1571-9. [PMID: 16645171 PMCID: PMC1895505 DOI: 10.1182/blood-2006-02-004747] [Citation(s) in RCA: 584] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
IL-2 plays a critical role in the maintenance of CD4+CD25+ FOXP3(+) regulatory T cells (Tregs) in vivo. We examined the effects of IL-2 signaling in human Tregs. In vitro, IL-2 selectively up-regulated the expression of FOXP3 in purified CD4+CD25+ T cells but not in CD4+CD25- cells. This regulation involved the binding of STAT3 and STAT5 proteins to a highly conserved STAT-binding site located in the first intron of the FOXP3 gene. We also examined the effects of low-dose IL-2 treatment in 12 patients with metastatic cancer and 9 patients with chronic myelogenous leukemia after allogeneic hematopoietic stem cell transplantation. Overall, IL-2 treatment resulted in a 1.9 median fold increase in the frequency of CD4+CD25+ cells in peripheral blood as well as a 9.7 median fold increase in FOXP3 expression in CD3+ T cells. CD56+CD3- natural killer (NK) cells also expanded during IL-2 therapy but did not express FOXP3. In vitro treatment of NK cells with 5-aza-2'-deoxycytidine restored the IL-2 signaling pathway leading to FOXP3 expression, suggesting that this gene was constitutively repressed by DNA methylation in these cells. Our findings support the clinical evaluation of low-dose IL-2 to selectively modulate CD4+CD25+ Tregs and increase expression of FOXP3 in vivo.
Collapse
MESH Headings
- Antigens, CD/blood
- CD4-Positive T-Lymphocytes/cytology
- CD4-Positive T-Lymphocytes/drug effects
- CD4-Positive T-Lymphocytes/immunology
- Cell Division
- Forkhead Transcription Factors/genetics
- Gene Expression Regulation/drug effects
- Humans
- Interleukin-2/pharmacology
- Interleukin-2/therapeutic use
- Killer Cells, Natural/drug effects
- Killer Cells, Natural/immunology
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/immunology
- Neoplasms/drug therapy
- Neoplasms/immunology
- Receptors, Interleukin-2/blood
- STAT Transcription Factors/physiology
- Stem Cell Transplantation
- T-Lymphocytes/drug effects
- T-Lymphocytes/immunology
Collapse
Affiliation(s)
- Emmanuel Zorn
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute, 44 Binney St, Boston, MA 02115, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Copier J, Whelan M, Dalgleish A. Biomarkers for the development of cancer vaccines: current status. Mol Diagn Ther 2006; 10:337-43. [PMID: 17154650 DOI: 10.1007/bf03256210] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Significant improvements in our knowledge of tumor immunology have resulted in more sophisticated vaccine approaches for the treatment of cancer. However, research into biomarkers that correlate with the clinical outcome of immunotherapy has lagged behind vaccine development. To this extent, very few immunological or other markers exist that can be used in clinical trials for immunotherapy. In this review, we discuss the current status of biomarker development specifically for the monitoring and development of cancer vaccines. This includes immunological biomarkers (measurement of T-cell and cytokine responses), autoimmunity as a correlate for treatment outcome, and the possible development of multiple biomarkers using high-throughput proteomics technologies. The generation of such biomarkers will allow us to make clinical decisions about patient treatment at an earlier stage and should aid in shortening the development time for vaccines.
Collapse
Affiliation(s)
- John Copier
- Department of Oncology, Division of Cellular and Molecular Medicine, St George's University of London, London, UK
| | | | | |
Collapse
|