1
|
Veatch JR, Singhi N, Srivastava S, Szeto JL, Jesernig B, Stull SM, Fitzgibbon M, Sarvothama M, Yechan-Gunja S, James SE, Riddell SR. A therapeutic cancer vaccine delivers antigens and adjuvants to lymphoid tissues using genetically modified T cells. J Clin Invest 2021; 131:e144195. [PMID: 34396986 PMCID: PMC8363286 DOI: 10.1172/jci144195] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 07/01/2021] [Indexed: 12/13/2022] Open
Abstract
Therapeutic vaccines that augment T cell responses to tumor antigens have been limited by poor potency in clinical trials. In contrast, the transfer of T cells modified with foreign transgenes frequently induces potent endogenous T cell responses to epitopes in the transgene product, and these responses are undesirable, because they lead to rejection of the transferred T cells. We sought to harness gene-modified T cells as a vaccine platform and developed cancer vaccines composed of autologous T cells modified with tumor antigens and additional adjuvant signals (Tvax). T cells expressing model antigens and a broad range of tumor neoantigens induced robust and durable T cell responses through cross-presentation of antigens by host DCs. Providing Tvax with signals such as CD80, CD137L, IFN-β, IL-12, GM-CSF, and FLT3L enhanced T cell priming. Coexpression of IL-12 and GM-CSF induced the strongest CD4+ and CD8+ T cell responses through complimentary effects on the recruitment and activation of DCs, mediated by autocrine IL-12 receptor signaling in the Tvax. Therapeutic vaccination with Tvax and adjuvants showed antitumor activity in subcutaneous and metastatic preclinical mouse models. Human T cells modified with neoantigens readily activated specific T cells derived from patients, providing a path for clinical translation of this therapeutic platform in cancer.
Collapse
Affiliation(s)
- Joshua R Veatch
- Clinical Research Division and Immunotherapy Integrated Research Center, Fred Hutchinson Cancer Research Center (FHCRC), Seattle, Washington, USA.,Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Naina Singhi
- Clinical Research Division and Immunotherapy Integrated Research Center, Fred Hutchinson Cancer Research Center (FHCRC), Seattle, Washington, USA
| | - Shivani Srivastava
- Clinical Research Division and Immunotherapy Integrated Research Center, Fred Hutchinson Cancer Research Center (FHCRC), Seattle, Washington, USA
| | - Julia L Szeto
- Clinical Research Division and Immunotherapy Integrated Research Center, Fred Hutchinson Cancer Research Center (FHCRC), Seattle, Washington, USA
| | - Brenda Jesernig
- Clinical Research Division and Immunotherapy Integrated Research Center, Fred Hutchinson Cancer Research Center (FHCRC), Seattle, Washington, USA
| | - Sylvia M Stull
- Clinical Research Division and Immunotherapy Integrated Research Center, Fred Hutchinson Cancer Research Center (FHCRC), Seattle, Washington, USA
| | | | - Megha Sarvothama
- Clinical Research Division and Immunotherapy Integrated Research Center, Fred Hutchinson Cancer Research Center (FHCRC), Seattle, Washington, USA
| | - Sushma Yechan-Gunja
- Clinical Research Division and Immunotherapy Integrated Research Center, Fred Hutchinson Cancer Research Center (FHCRC), Seattle, Washington, USA
| | - Scott E James
- Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Stanley R Riddell
- Clinical Research Division and Immunotherapy Integrated Research Center, Fred Hutchinson Cancer Research Center (FHCRC), Seattle, Washington, USA.,Department of Medicine, University of Washington, Seattle, Washington, USA
| |
Collapse
|
2
|
Huang MN, Nicholson LT, Batich KA, Swartz AM, Kopin D, Wellford S, Prabhakar VK, Woroniecka K, Nair SK, Fecci PE, Sampson JH, Gunn MD. Antigen-loaded monocyte administration induces potent therapeutic antitumor T cell responses. J Clin Invest 2020; 130:774-788. [PMID: 31661470 DOI: 10.1172/jci128267] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Accepted: 10/22/2019] [Indexed: 12/20/2022] Open
Abstract
Efficacy of dendritic cell (DC) cancer vaccines is classically thought to depend on their antigen-presenting cell (APC) activity. Studies show, however, that DC vaccine priming of cytotoxic T lymphocytes (CTLs) requires the activity of endogenous DCs, suggesting that exogenous DCs stimulate antitumor immunity by transferring antigens (Ags) to endogenous DCs. Such Ag transfer functions are most commonly ascribed to monocytes, implying that undifferentiated monocytes would function equally well as a vaccine modality and need not be differentiated to DCs to be effective. Here, we used several murine cancer models to test the antitumor efficacy of undifferentiated monocytes loaded with protein or peptide Ag. Intravenously injected monocytes displayed antitumor activity superior to DC vaccines in several cancer models, including aggressive intracranial glioblastoma. Ag-loaded monocytes induced robust CTL responses via Ag transfer to splenic CD8+ DCs in a manner independent of monocyte APC activity. Ag transfer required cell-cell contact and the formation of connexin 43-containing gap junctions between monocytes and DCs. These findings demonstrate the existence of an efficient gap junction-mediated Ag transfer pathway between monocytes and CD8+ DCs and suggest that administration of tumor Ag-loaded undifferentiated monocytes may serve as a simple and efficacious immunotherapy for the treatment of human cancers.
Collapse
Affiliation(s)
- Min-Nung Huang
- Department of Immunology.,Division of Cardiology, Department of Medicine
| | | | - Kristen A Batich
- School of Medicine.,Department of Pathology.,Preston Robert Tisch Brain Tumor Center
| | - Adam M Swartz
- Department of Pathology.,Preston Robert Tisch Brain Tumor Center
| | | | | | | | - Karolina Woroniecka
- School of Medicine.,Department of Pathology.,Preston Robert Tisch Brain Tumor Center
| | - Smita K Nair
- Department of Pathology.,Preston Robert Tisch Brain Tumor Center.,Department of Neurosurgery, and.,Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA
| | - Peter E Fecci
- Department of Pathology.,Preston Robert Tisch Brain Tumor Center.,Department of Neurosurgery, and
| | - John H Sampson
- Department of Pathology.,Preston Robert Tisch Brain Tumor Center.,Department of Neurosurgery, and
| | - Michael D Gunn
- Department of Immunology.,Division of Cardiology, Department of Medicine
| |
Collapse
|
3
|
Baldin AV, Savvateeva LV, Bazhin AV, Zamyatnin AA. Dendritic Cells in Anticancer Vaccination: Rationale for Ex Vivo Loading or In Vivo Targeting. Cancers (Basel) 2020; 12:cancers12030590. [PMID: 32150821 PMCID: PMC7139354 DOI: 10.3390/cancers12030590] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 02/29/2020] [Accepted: 03/02/2020] [Indexed: 12/16/2022] Open
Abstract
Dendritic cells (DCs) have shown great potential as a component or target in the landscape of cancer immunotherapy. Different in vivo and ex vivo strategies of DC vaccine generation with different outcomes have been proposed. Numerous clinical trials have demonstrated their efficacy and safety in cancer patients. However, there is no consensus regarding which DC-based vaccine generation method is preferable. A problem of result comparison between trials in which different DC-loading or -targeting approaches have been applied remains. The employment of different DC generation and maturation methods, antigens and administration routes from trial to trial also limits the objective comparison of DC vaccines. In the present review, we discuss different methods of DC vaccine generation. We conclude that standardized trial designs, treatment settings and outcome assessment criteria will help to determine which DC vaccine generation approach should be applied in certain cancer cases. This will result in a reduction in alternatives in the selection of preferable DC-based vaccine tactics in patient. Moreover, it has become clear that the application of a DC vaccine alone is not sufficient and combination immunotherapy with recent advances, such as immune checkpoint inhibitors, should be employed to achieve a better clinical response and outcome.
Collapse
Affiliation(s)
- Alexey V. Baldin
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (A.V.B.); (L.V.S.)
| | - Lyudmila V. Savvateeva
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (A.V.B.); (L.V.S.)
| | - Alexandr V. Bazhin
- Department of General, Visceral and Transplant Surgery, Ludwig-Maximilians University of Munich, 81377 Munich, Germany;
- German Cancer Consortium (DKTK), Partner Site Munich, 80336 Munich, Germany
| | - Andrey A. Zamyatnin
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (A.V.B.); (L.V.S.)
- Belozersky Institute of Physico-Chemical Biology, Department of Cell Signaling, Lomonosov Moscow State University, 119991 Moscow, Russia
- Correspondence: ; Tel.: +74-956-229-843
| |
Collapse
|
4
|
Lenti E, Bianchessi S, Proulx ST, Palano MT, Genovese L, Raccosta L, Spinelli A, Drago D, Andolfo A, Alfano M, Petrova TV, Mukenge S, Russo V, Brendolan A. Therapeutic Regeneration of Lymphatic and Immune Cell Functions upon Lympho-organoid Transplantation. Stem Cell Reports 2019; 12:1260-1268. [PMID: 31155505 PMCID: PMC6565831 DOI: 10.1016/j.stemcr.2019.04.021] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 04/29/2019] [Accepted: 04/30/2019] [Indexed: 12/14/2022] Open
Abstract
Lymph nodes (LNs) are secondary lymphoid tissues that play a critical role in filtering the lymph and promoting adaptive immune responses. Surgical resection of LNs, radiation therapy, or infections may damage lymphatic vasculature and compromise immune functions. Here, we describe the generation of functional synthetic lympho-organoids (LOs) using LN stromal progenitors and decellularized extracellular matrix-based scaffolds, two basic constituents of secondary lymphoid tissues. We show that upon transplantation at the site of resected LNs, LOs become integrated into the endogenous lymphatic vasculature and efficiently restore lymphatic drainage and perfusion. Upon immunization, LOs support the activation of antigen-specific immune responses, thus acquiring properties of native lymphoid tissues. These findings provide a proof-of-concept strategy for the development of functional lympho-organoids suitable for restoring lymphatic and immune cell functions.
Collapse
Affiliation(s)
- Elisa Lenti
- Unit of Lymphoid Organ Development, Division of Experimental Oncology, DIBIT-1 3A2, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132 Milan, Italy
| | - Silvia Bianchessi
- Unit of Lymphoid Organ Development, Division of Experimental Oncology, DIBIT-1 3A2, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132 Milan, Italy
| | - Steven T Proulx
- Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology, ETH Zurich, 8093 Zurich, Switzerland
| | - Maria Teresa Palano
- Unit of Lymphoid Organ Development, Division of Experimental Oncology, DIBIT-1 3A2, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132 Milan, Italy
| | - Luca Genovese
- Unit of Lymphoid Organ Development, Division of Experimental Oncology, DIBIT-1 3A2, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132 Milan, Italy
| | - Laura Raccosta
- Unit of Immuno-Biotherapy of Melanoma and Solid Tumors, Division of Experimental Oncology, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Antonello Spinelli
- Experimental Imaging Centre, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Denise Drago
- ProMiFa, Protein Microsequencing Facility, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Annapaola Andolfo
- ProMiFa, Protein Microsequencing Facility, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Massimo Alfano
- Division of Experimental Oncology/Unit of Urology, URI, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Tatiana V Petrova
- Department of Oncology, University of Lausanne, and Ludwig Institute for Cancer Research, 1066 Lausanne, Switzerland
| | - Sylvain Mukenge
- Department of Hepatobiliary Surgery, IRCCS San Raffaele Hospital, 20132 Milan, Italy
| | - Vincenzo Russo
- Unit of Immuno-Biotherapy of Melanoma and Solid Tumors, Division of Experimental Oncology, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Andrea Brendolan
- Unit of Lymphoid Organ Development, Division of Experimental Oncology, DIBIT-1 3A2, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132 Milan, Italy.
| |
Collapse
|
5
|
Kitazawa Y, Ueta H, Sawanobori Y, Katakai T, Yoneyama H, Ueha S, Matsushima K, Tokuda N, Matsuno K. Novel Targeting to XCR1 + Dendritic Cells Using Allogeneic T Cells for Polytopical Antibody Responses in the Lymph Nodes. Front Immunol 2019; 10:1195. [PMID: 31191552 PMCID: PMC6548820 DOI: 10.3389/fimmu.2019.01195] [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: 10/09/2018] [Accepted: 05/10/2019] [Indexed: 01/23/2023] Open
Abstract
Vaccination strategy that induce efficient antibody responses polytopically in most lymph nodes (LNs) against infections has not been established yet. Because donor-specific blood transfusion induces anti-donor class I MHC antibody production in splenectomized rats, we examined the mechanism and significance of this response. Among the donor blood components, T cells were the most efficient immunogens, inducing recipient T cell and B cell proliferative responses not only in the spleen, but also in the peripheral and gut LNs. Donor T cells soon migrated to the splenic T cell area and the LNs, with a temporary significant increase in recipient NK cells. XCR1+ resident dendritic cells (DCs), but not XCR1− DCs, selectively phagocytosed donor class I MHC+ fragments after 1 day. After 1.5 days, both DC subsets formed clusters with recipient CD4+ T cells, which proliferated within these clusters. Inhibition of donor T cell migration or depletion of NK cells by pretreatment with pertussis toxin or anti-asialoGM1 antibody, respectively, significantly suppressed DC phagocytosis and subsequent immune responses. Three allogeneic strains with different NK activities had the same response but with different intensity. Donor T cell proliferation was not required, indicating that the graft vs. host reaction is dispensable. Intravenous transfer of antigen-labeled and mitotic inhibitor-treated allogeneic, but not syngeneic, T cells induced a polytopical antibody response to labeled antigens in the LNs of splenectomized rats. These results demonstrate a novel mechanism of alloresponses polytopically in the secondary lymphoid organs (SLOs) induced by allogeneic T cells. Donor T cells behave as self-migratory antigen ferries to be delivered to resident XCR1+ DCs with negligible commitment of migratory DCs. Allogeneic T cells may be clinically applicable as vaccine vectors for polytopical prophylactic antibody production even in asplenic or hyposplenic individuals.
Collapse
Affiliation(s)
- Yusuke Kitazawa
- Department of Anatomy (Macro), School of Medicine, Dokkyo Medical University, Tochigi, Japan
| | - Hisashi Ueta
- Department of Anatomy (Macro), School of Medicine, Dokkyo Medical University, Tochigi, Japan
| | - Yasushi Sawanobori
- Department of Anatomy (Macro), School of Medicine, Dokkyo Medical University, Tochigi, Japan
| | - Tomoya Katakai
- Department of Immunology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | | | - Satoshi Ueha
- Division of Molecular Regulation of Inflammatory and Immune Diseases, Research Institute for Biomedical Sciences, Tokyo University of Science, Chiba, Japan
| | - Kouji Matsushima
- Division of Molecular Regulation of Inflammatory and Immune Diseases, Research Institute for Biomedical Sciences, Tokyo University of Science, Chiba, Japan
| | - Nobuko Tokuda
- Department of Anatomy (Macro), School of Medicine, Dokkyo Medical University, Tochigi, Japan
| | - Kenjiro Matsuno
- Department of Anatomy (Macro), School of Medicine, Dokkyo Medical University, Tochigi, Japan
| |
Collapse
|
6
|
O'Neil RT, Saha S, Veach RA, Welch RC, Woodard LE, Rooney CM, Wilson MH. Transposon-modified antigen-specific T lymphocytes for sustained therapeutic protein delivery in vivo. Nat Commun 2018; 9:1325. [PMID: 29636469 PMCID: PMC5893599 DOI: 10.1038/s41467-018-03787-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Accepted: 03/12/2018] [Indexed: 12/22/2022] Open
Abstract
A cell therapy platform permitting long-term delivery of peptide hormones in vivo would be a significant advance for patients with hormonal deficiencies. Here we report the utility of antigen-specific T lymphocytes as a regulatable peptide delivery platform for in vivo therapy. piggyBac transposon modification of murine cells with luciferase allows us to visualize T cells after adoptive transfer. Vaccination stimulates long-term T-cell engraftment, persistence, and transgene expression enabling detection of modified cells up to 300 days after adoptive transfer. We demonstrate adoptive transfer of antigen-specific T cells expressing erythropoietin (EPO) elevating the hematocrit in mice for more than 20 weeks. We extend our observations to human T cells demonstrating inducible EPO production from Epstein-Barr virus (EBV) antigen-specific T lymphocytes. Our results reveal antigen-specific T lymphocytes to be an effective delivery platform for therapeutic molecules such as EPO in vivo, with important implications for other diseases that require peptide therapy.
Collapse
Affiliation(s)
- Richard T O'Neil
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA
- The Vanderbilt Center for Kidney Disease, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA
| | - Sunandan Saha
- Interdepartmental Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Ruth Ann Veach
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA
- The Vanderbilt Center for Kidney Disease, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA
| | - Richard C Welch
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA
- The Vanderbilt Center for Kidney Disease, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA
| | - Lauren E Woodard
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA
- The Vanderbilt Center for Kidney Disease, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA
- Department of Veterans Affairs, Nashville, TN, 37212, USA
| | - Cliona M Rooney
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX, 77030, USA
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, 77030, USA
- Department of Immunology, Baylor College of Medicine, Houston, TX, 77030, USA
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Matthew H Wilson
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA.
- The Vanderbilt Center for Kidney Disease, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA.
- Department of Veterans Affairs, Nashville, TN, 37212, USA.
| |
Collapse
|
7
|
Lanterna C, Musumeci A, Raccosta L, Corna G, Moresco M, Maggioni D, Fontana R, Doglioni C, Bordignon C, Traversari C, Russo V. The administration of drugs inhibiting cholesterol/oxysterol synthesis is safe and increases the efficacy of immunotherapeutic regimens in tumor-bearing mice. Cancer Immunol Immunother 2016; 65:1303-1315. [PMID: 27520505 PMCID: PMC11029546 DOI: 10.1007/s00262-016-1884-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Accepted: 08/06/2016] [Indexed: 01/15/2023]
Abstract
Tumor-derived metabolites dampen tumor-infiltrating immune cells and antitumor immune responses. Among the various metabolites produced by tumors, we recently showed that cholesterol oxidized products, namely oxysterols, favor tumor growth through the inhibition of DC migration toward lymphoid organs and by promoting the recruitment of pro-tumor neutrophils within the tumor microenvironment. Here, we tested different drugs capable of blocking cholesterol/oxysterol formation. In particular, we tested efficacy and safety of different administration schedules, and of immunotherapy-based combination of a class of compounds, namely zaragozic acids, which inhibit cholesterol pathway downstream of mevalonate formation, thus leaving intact the formation of the isoprenoids, which are required for the maturation of proteins involved in the immune cell function. We show that zaragozic acids inhibit the in vivo growth of the RMA lymphoma and the Lewis lung carcinoma (LLC) without inducing side effects. Tumor growth inhibition requires an intact immune system, as immunodeficient tumor-bearing mice do not respond to zaragozic acid treatment. Of note, the effect of zaragozic acids is accompanied by a marked reduction in the LXR target genes Abcg1, Mertk, Scd1 and Srebp-1c in the tumor microenvironment. On the other hand, zoledronate, which blocks also isoprenoid formation, did not control the LLC tumor growth. Finally, we show that zaragozic acids potentiate the antitumor effects of active and adoptive immunotherapy, significantly prolonging the overall survival of tumor-bearing mice treated with the combo zaragozic acids and TAA-loaded DCs. This study identifies zaragozic acids as new antitumor compounds exploitable for the treatment of cancer patients.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents/therapeutic use
- Bridged Bicyclo Compounds, Heterocyclic/therapeutic use
- Carcinoma, Lewis Lung/immunology
- Carcinoma, Lewis Lung/therapy
- Cholesterol/metabolism
- Combined Modality Therapy
- Dendritic Cells/immunology
- Dendritic Cells/transplantation
- Gene Expression Regulation, Neoplastic/drug effects
- Humans
- Immunotherapy, Adoptive/methods
- Lymphoma, T-Cell/immunology
- Lymphoma, T-Cell/therapy
- Mice
- Mice, Inbred C57BL
- Mice, Inbred NOD
- Mice, SCID
- Oxysterols/metabolism
- Tricarboxylic Acids/therapeutic use
- Tumor Escape
- Tumor Microenvironment
Collapse
Affiliation(s)
- Claudia Lanterna
- Immuno-Biotherapy of Melanoma and Solid Tumors Unit, Division of Experimental Oncology, IRCCS Scientific Institute San Raffaele, Via Olgettina 58, 20132, Milan, Italy
- MolMed S.p.A., Milan, Italy
| | - Andrea Musumeci
- Immuno-Biotherapy of Melanoma and Solid Tumors Unit, Division of Experimental Oncology, IRCCS Scientific Institute San Raffaele, Via Olgettina 58, 20132, Milan, Italy
- Institute for Immunology, Munich, Germany
| | - Laura Raccosta
- Immuno-Biotherapy of Melanoma and Solid Tumors Unit, Division of Experimental Oncology, IRCCS Scientific Institute San Raffaele, Via Olgettina 58, 20132, Milan, Italy
| | - Gianfranca Corna
- Immuno-Biotherapy of Melanoma and Solid Tumors Unit, Division of Experimental Oncology, IRCCS Scientific Institute San Raffaele, Via Olgettina 58, 20132, Milan, Italy
| | - Marta Moresco
- Immuno-Biotherapy of Melanoma and Solid Tumors Unit, Division of Experimental Oncology, IRCCS Scientific Institute San Raffaele, Via Olgettina 58, 20132, Milan, Italy
| | - Daniela Maggioni
- Immuno-Biotherapy of Melanoma and Solid Tumors Unit, Division of Experimental Oncology, IRCCS Scientific Institute San Raffaele, Via Olgettina 58, 20132, Milan, Italy
| | - Raffaella Fontana
- Immuno-Biotherapy of Melanoma and Solid Tumors Unit, Division of Experimental Oncology, IRCCS Scientific Institute San Raffaele, Via Olgettina 58, 20132, Milan, Italy
| | - Claudio Doglioni
- Department of Pathology, IRCCS Scientific Institute San Raffaele, Milan, Italy
- Università Vita-Salute San Raffaele, Milan, Italy
| | - Claudio Bordignon
- Università Vita-Salute San Raffaele, Milan, Italy
- MolMed S.p.A., Milan, Italy
| | | | - Vincenzo Russo
- Immuno-Biotherapy of Melanoma and Solid Tumors Unit, Division of Experimental Oncology, IRCCS Scientific Institute San Raffaele, Via Olgettina 58, 20132, Milan, Italy.
| |
Collapse
|
8
|
Liu X, Barrett DM, Jiang S, Fang C, Kalos M, Grupp SA, June CH, Zhao Y. Improved anti-leukemia activities of adoptively transferred T cells expressing bispecific T-cell engager in mice. Blood Cancer J 2016; 6:e430. [PMID: 27258611 PMCID: PMC5141353 DOI: 10.1038/bcj.2016.38] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2016] [Accepted: 04/28/2016] [Indexed: 12/29/2022] Open
Abstract
Despite the impressive clinical efficacy of T cells engineered to express chimeric antigen receptors (CAR-Ts), the current applications of CAR-T cell therapy are limited by major treatment-related toxicity. Thus, safer yet effective alternative approaches must be developed. In this study, we compared CD19 bispecific T-cell engager (BiTE)-transferred T cells that had been transfected by RNA electroporation with CD19 CAR RNA-transferred T cells both in vitro and in an aggressive Nalm6 leukemia mouse model. BiTEs were secreted from the transferred T cells and enabled both the transferred and bystander T cells to specifically recognize CD19+ cell lines, with increased tumor killing ability, prolonged functional persistence, increased cytokine production and potent proliferation compared with the CAR-T cells. More interestingly, in comparison with CD3/CD28 bead-stimulated T cells, T cells that were expanded by a rapid T-cell expansion protocol (REP) showed enhanced anti-tumor activities for both CAR and BiTE RNA-electroporated T cells both in vitro and in a Nalm6 mouse model (P<0.01). Furthermore, the REP T cells with BiTE RNAs showed greater efficacy in the Nalm6 leukemia model compared with REP T cells with CAR RNA (P<0.05) and resulted in complete leukemia remission.
Collapse
Affiliation(s)
- X Liu
- Center for Cellular Immunotherapies, University of Pennsylvania Cancer Center, Philadelphia, PA, USA
| | - D M Barrett
- Division of Oncology, Department of Pathology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - S Jiang
- Center for Cellular Immunotherapies, University of Pennsylvania Cancer Center, Philadelphia, PA, USA
| | - C Fang
- Center for Cellular Immunotherapies, University of Pennsylvania Cancer Center, Philadelphia, PA, USA
| | - M Kalos
- Center for Cellular Immunotherapies, University of Pennsylvania Cancer Center, Philadelphia, PA, USA.,Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - S A Grupp
- Division of Oncology, Department of Pathology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - C H June
- Center for Cellular Immunotherapies, University of Pennsylvania Cancer Center, Philadelphia, PA, USA.,Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Y Zhao
- Center for Cellular Immunotherapies, University of Pennsylvania Cancer Center, Philadelphia, PA, USA.,Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
9
|
Abstract
The exploitation of the physiologic processing and presenting machinery of dendritic cells (DCs) by in vivo loading of tumor-associated antigens may improve the immunogenic potential and clinical efficacy of DC-based cancer vaccines. The approach developed by our group was based on the clinical observation that some patients treated with the infusion of donor lymphocytes transduced to express the HSV-TK suicide gene for relapse of hematologic malignancies, after allogeneic hematopoietic stem cell transplantation, developed a T cell-mediated immune response specifically directed against the HSV-TK gene product.We demonstrated that lymphocytes genetically modified to express HSV-TK as well as self/tumor antigens, acting as antigen carriers, efficiently target DCs in vivo in tumor-bearing mice. The infusion of TRP-2-transduced lymphocytes induced the establishment of protective immunity and long-term memory in tumor-bearing mice by cross-presentation of the antigen mediated by the CD11c(+)CD8a(+) DCs subset. A similar approach was applied in a clinical setting. Ten patients affected by MAGE-3(+) metastatic melanoma were treated with autologous lymphocytes retrovirally transduced to express the MAGE-3 tumor antigen. In three patients, the treatment led to the increase of MAGE-3 specific CD8+ and CD4+ effectors and the development of long-term memory, which ultimately correlated with a favorable clinical outcome. Transduced lymphocytes represent an efficient way for in vivo loading of tumor-associated antigens of DCs.
Collapse
Affiliation(s)
| | - Vincenzo Russo
- Cancer Gene Therapy Unit, IRCCS San Raffaele Hospital, Milano, Italy
| |
Collapse
|
10
|
CD4 T-cells transduced with CD80 and 4-1BBL mRNA induce long-term CD8 T-cell responses resulting in potent antitumor effects. Vaccine 2014; 32:6919-6926. [PMID: 25444817 DOI: 10.1016/j.vaccine.2014.10.042] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2014] [Revised: 09/16/2014] [Accepted: 10/25/2014] [Indexed: 11/21/2022]
Abstract
Therapeutic cancer vaccines are an attractive alternative to conventional therapies to treat malignant tumors, and more importantly, to prevent recurrence after primary therapy. However, the availability of professional antigen-presenting cells (APCs) has been restricted by difficulties encountered in obtaining sufficient professional APCs for clinical use. We have prepared an alternative cellular vaccine with CD4 T-cells that can be expanded easily to yield a pure and homogeneous population in vitro. To enhance their potency as a therapeutic vaccine, in vitro expanded CD4 T-cells were transfected with RNAs encoding the costimulatory ligands CD80, 4-1BBL, or both (CD80-T, 4-1BBL-T, and CD80/4-1BBL-T-cells, respectively). We observed augmented cell vitality in CD80/4-1BBL-T-cells in vitro and in vivo. Significant CD8 T-cell responses eliciting in vivo proliferation and cytotoxicity were obtained with CD80/4-1BBL-T-cell vaccination compared to CD80-T and 4-1BBL-T-cell vaccinations. In contrast, β2m-deficient CD80/4-1BBL-T-cells were not as effective as wile-type CD80/4-1BBL-T-cells in priming CD8 T-cells. Furthermore, CD80/4-1BBL-T-cell immunization resulted in curing established EG7 tumors, resulting in the generation of memory CD8 T-cell responses, and elicited therapeutic antitumor responses against B16 melanoma. These results suggest that CD4 T-cells endowed with costimulatory ligands allow the design of effective vaccination strategies against cancer.
Collapse
|
11
|
Phua KKL, Boczkowski D, Dannull J, Pruitt S, Leong KW, Nair SK. Whole blood cells loaded with messenger RNA as an anti-tumor vaccine. Adv Healthc Mater 2014; 3:837-42. [PMID: 24339387 DOI: 10.1002/adhm.201300512] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2013] [Revised: 11/18/2013] [Indexed: 01/07/2023]
Abstract
The use of a cell-based vaccine composed of autologous whole blood cells loaded with mRNA is described. Mice immunized with whole blood cells loaded with mRNA encoding antigen develop anti-tumor immunity comparable to DC-RNA immunization. This approach offers a simple and affordable alternative to RNA-based cellular therapy by circumventing complex, laborious and expensive ex vivo manipulations required for DC-based immunizations.
Collapse
Affiliation(s)
- Kyle K. L. Phua
- Department of Chemical & Biomolecular Engineering; National University of Singapore; Singapore 117576
- Department of Biomedical Engineering; Duke University; Durham NC 27708 USA
| | - David Boczkowski
- Department of Surgery; Duke University Medical Center; Durham NC 27710 USA
| | - Jens Dannull
- Department of Surgery; Duke University Medical Center; Durham NC 27710 USA
| | - Scott Pruitt
- Experimental Medicine; Merck Research Laboratories; Rahway NJ 07065 USA
| | - Kam W. Leong
- Department of Biomedical Engineering; Duke University; Durham NC 27708 USA
| | - Smita K. Nair
- Department of Surgery; Duke University Medical Center; Durham NC 27710 USA
| |
Collapse
|
12
|
Lamers CHJ, Debets R. Genetically modified T lymphocytes: more than just direct effectors. Immunotherapy 2013; 5:691-4. [DOI: 10.2217/imt.13.62] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Evaluation of: Russo V, Pilla L, Lunghi F et al. Clinical and immunologic responses in melanoma patients vaccinated with MAGE-A3-genetically modified lymphocytes. Int. J. Cancer 132(11), 2557–2566 (2012). When one mentions T lymphocytes, one easily recognizes the effective and antigen-specific manner by which T lymphocytes execute cellular immune responses towards pathogen-infected or cancerous cells. Russo and coworkers recognized the other side of the coin and exploited the potency of T cells to act as a cellular vaccine, to which end they used T cells transduced with the cancer–testis antigen MAGE-A3. Twenty-three patients with MAGE-A3-expressing melanoma were treated and six patients developed MAGE-A3-specific immune responses and showed clinical benefit, whereas patients without a MAGE-A3-specific immune response did not show clinical benefit. This report includes and extends on results from a pilot study including ten patients, of which three developed MAGE-A3-specific immune responses. The present study further explores a potential beneficial application of the observed immunogenicity of genetically modified T cells.
Collapse
Affiliation(s)
- Cor HJ Lamers
- Laboratory of Experimental Tumor Immunology, Department of Medical Oncology, Erasmus MC Cancer Institute, PO Box 5201, 3008 AE Rotterdam, The Netherlands.
| | - Reno Debets
- Laboratory of Experimental Tumor Immunology, Department of Medical Oncology, Erasmus MC Cancer Institute, PO Box 5201, 3008 AE Rotterdam, The Netherlands
| |
Collapse
|
13
|
Russo V, Pilla L, Lunghi F, Crocchiolo R, Greco R, Ciceri F, Maggioni D, Fontana R, Mukenge S, Rivoltini L, Rigamonti G, Mercuri SR, Nicoletti R, Maschio AD, Gianolli L, Fazio F, Marchianò A, Florio AD, Maio M, Salomoni M, Gallo-Stampino C, Fiacco MD, Lambiase A, Coulie PG, Patuzzo R, Parmiani G, Traversari C, Bordignon C, Santinami M, Bregni M. Clinical and immunologic responses in melanoma patients vaccinated with MAGE-A3-genetically modified lymphocytes. Int J Cancer 2012; 132:2557-66. [DOI: 10.1002/ijc.27939] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2012] [Accepted: 10/15/2012] [Indexed: 01/31/2023]
|
14
|
Russo V, Bondanza A, Ciceri F, Bregni M, Bordignon C, Traversari C, Bonini C. A dual role for genetically modified lymphocytes in cancer immunotherapy. Trends Mol Med 2012; 18:193-200. [DOI: 10.1016/j.molmed.2011.12.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2011] [Revised: 12/04/2011] [Accepted: 12/12/2011] [Indexed: 12/18/2022]
|
15
|
Bouvier I, Jusforgues-Saklani H, Lim A, Lemaître F, Lemercier B, Auriau C, Nicola MA, Leroy S, Law HK, Bandeira A, Moon JJ, Bousso P, Albert ML. Immunization route dictates cross-priming efficiency and impacts the optimal timing of adjuvant delivery. Front Immunol 2011; 2:71. [PMID: 22566860 PMCID: PMC3342317 DOI: 10.3389/fimmu.2011.00071] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2011] [Accepted: 11/17/2011] [Indexed: 11/29/2022] Open
Abstract
Delivery of cell-associated antigen represents an important strategy for vaccination. While many experimental models have been developed in order to define the critical parameters for efficient cross-priming, few have utilized quantitative methods that permit the study of the endogenous repertoire. Comparing different strategies of immunization, we report that local delivery of cell-associated antigen results in delayed T cell cross-priming due to the increased time required for antigen capture and presentation. In comparison, delivery of disseminated antigen resulted in rapid T cell priming. Surprisingly, local injection of cell-associated antigen, while slower, resulted in the differentiation of a more robust, polyfunctional, effector response. We also evaluated the combination of cell-associated antigen with poly I:C delivery and observed an immunization route-specific effect regarding the optimal timing of innate immune stimulation. These studies highlight the importance of considering the timing and persistence of antigen presentation, and suggest that intradermal injection with delayed adjuvant delivery is the optimal strategy for achieving CD8+ T cell cross-priming.
Collapse
Affiliation(s)
- Isabelle Bouvier
- Unité Immunobiologie des Cellules Dendritiques, Institut Pasteur Paris, France
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
T cells as vehicles for cancer vaccination. J Biomed Biotechnol 2011; 2011:417403. [PMID: 22131805 PMCID: PMC3205726 DOI: 10.1155/2011/417403] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2011] [Accepted: 08/30/2011] [Indexed: 12/20/2022] Open
Abstract
The success of cancer vaccines is dependent on the delivery of tumor-associated antigens (TAAs) within lymphoid tissue in the context of costimulatory molecules and immune stimulatory cytokines. Dendritic cells (DCs) are commonly utilized to elicit antitumor immune responses due to their attractive costimulatory molecule and cytokine expression profile. However, the efficacy of DC-based vaccines is limited by the poor viability and lymph-node migration of exogenously generated DCs in vivo. Alternatively, adoptively transferred T cells persist for long periods of time in vivo and readily migrate between the lymphoid and vascular compartments. In addition, T cells may be genetically modified to express both TAA and DC-activating molecules, suggesting that T cells may be ideal candidates to serve as cellular vehicles for antigen delivery to lymph node-resident DCs in vivo. This paper discusses the concept of using T cells to induce tumor-specific immunity for vaccination against cancer.
Collapse
|
17
|
Anderson LD, Cook DR, Yamamoto TN, Berger C, Maloney DG, Riddell SR. Identification of MAGE-C1 (CT-7) epitopes for T-cell therapy of multiple myeloma. Cancer Immunol Immunother 2011; 60:985-97. [PMID: 21461886 PMCID: PMC3183483 DOI: 10.1007/s00262-011-1009-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2010] [Accepted: 02/10/2011] [Indexed: 12/19/2022]
Abstract
Multiple myeloma is incurable with standard therapies but is susceptible to a T-cell-mediated graft versus myeloma effect after allogeneic stem cell transplantation. We sought to identify myeloma-specific antigens that might be used for T-cell immunotherapy of myeloma. MAGE-C1 (CT-7) is a cancer-testis antigen that is expressed by tumor cells in >70% of myeloma patients and elicits a humoral response in up to 93% of patients with CT-7(+) myeloma. No CD8(+) T-cell epitopes have been described for CT-7, so we used a combination of reverse immunology and immunization of HLA-A2 transgenic mice with a novel cell-based vaccine to identify three immunogenic epitopes of CT-7 that are recognized by human CD8(+) T-cells. CT-7-specific T-cells recognizing two of these peptides are able to recognize myeloma cells as well as CT-7 gene-transduced tumor cells, demonstrating that these epitopes are naturally processed and presented by tumor cells. This is the first report of the identification of immunogenic CD8(+) T-cell epitopes of MAGE-C1 (CT-7), which is the most commonly expressed cancer-testis antigen found in myeloma, and these epitopes may be promising candidate targets for vaccination or T-cell therapy of myeloma or other CT-7(+) malignancies.
Collapse
Affiliation(s)
- Larry D Anderson
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA.
| | | | | | | | | | | |
Collapse
|
18
|
Russo V, Lunghi F, Fontana R, Bregni M. A Clinical Study of a Cell-Based MAGE-A3 Active Immunotherapy in Advanced Melanoma Patients. J Cancer 2011; 2:329-30. [PMID: 21716850 PMCID: PMC3119396 DOI: 10.7150/jca.2.329] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2011] [Accepted: 05/26/2011] [Indexed: 11/05/2022] Open
Abstract
In this bi-institutional study, twenty-three stage IIIC-IV MAGE-A3(+) melanoma patients were vaccinated with M3TK-GML biweekly at three dose levels, with a subsequent phase of vaccinations at the maximum dose level. Anti-MAGE-A3 and anti-TK T cells were assessed by in vitro assay and delayed-type hypersensitivity skin testing.
Collapse
Affiliation(s)
- Vincenzo Russo
- 1. Cancer Gene Therapy Unit, San Raffaele Scientific Institute, via Olgettina 60, 20132, Milan, Italy
| | | | | | | |
Collapse
|
19
|
Abstract
The observation that T cells can recognize and specifically eliminate cancer cells has spurred interest in the development of efficient methods to generate large numbers of T cells with specificity for tumor antigens that can be harnessed for use in cancer therapy. Recent studies have demonstrated that during encounter with tumor antigen, the signals delivered to T cells by professional antigen-presenting cells can affect T-cell programming and their subsequent therapeutic efficacy. This has stimulated efforts to develop artificial antigen-presenting cells that allow optimal control over the signals provided to T cells. In this review, we will discuss the advantages and disadvantages of cellular and acellular artificial antigen-presenting cell systems and their use in T-cell adoptive immunotherapy for cancer.
Collapse
|
20
|
Cao Q, Jin Y, Jin M, He S, Gu Q, He S, Qiu Y, Ge H, Yoneyama H, Zhang Y. Therapeutic effect of MIP-1alpha-recruited dendritic cells on preestablished solid and metastatic tumors. Cancer Lett 2010; 295:17-26. [PMID: 20202744 DOI: 10.1016/j.canlet.2010.02.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2008] [Revised: 02/05/2010] [Accepted: 02/10/2010] [Indexed: 11/17/2022]
Abstract
We previously found that dendritic cell (DC) precursors could be recruited into the peripheral blood of B6 mice by administration of macrophage inflammatory protein (MIP)-1alpha. These MIP-1alpha-recruited DCs could induce anti-tumor protective immunity when pulsed with tumor cell lysate. In this study, MIP-1alpha-recruited DCs could not effectively suppress preestablished tumor when pulsed with B16 tumor cell lysate. However, inoculation with these DCs expressing MAGE-1 induced an anti-tumor immunity against preestablished solid and metastatic tumor from B16-MAGE-1 cells. These MIP-1alpha-recruited DCs expressed higher level of CCR7 and displayed a more significant chemotactic response toward secondary lymphoid tissue. Therefore, they are superior in the induction of cytotoxic T lymphocytes and the inhibition of tumor development and metastasis than bone marrow-derived DCs. This study established a novel approach to the treatment of preestablished solid and metastatic tumors using MIP-1alpha-recruited DCs transduced with tumor antigen gene.
Collapse
MESH Headings
- Animals
- Antigens, Neoplasm/genetics
- Cancer Vaccines/immunology
- Cancer Vaccines/therapeutic use
- Cell Line, Tumor
- Chemokine CCL3/pharmacology
- Chemotaxis
- Cytotoxicity, Immunologic
- Dendritic Cells/immunology
- Female
- Immunotherapy
- Lung Neoplasms/secondary
- Lung Neoplasms/therapy
- Lymphocyte Activation
- Melanoma, Experimental/immunology
- Melanoma, Experimental/prevention & control
- Melanoma, Experimental/secondary
- Melanoma, Experimental/therapy
- Melanoma-Specific Antigens
- Mice
- Mice, Inbred C57BL
- Neoplasm Proteins/genetics
- Receptors, CCR7/metabolism
- Recombinant Proteins/pharmacology
- T-Lymphocytes, Cytotoxic/immunology
- Transduction, Genetic
Collapse
Affiliation(s)
- Qi Cao
- Shanghai Institute of Immunology, Institutes of Medical Sciences, Shanghai Jiao Tong University School of Medicine and Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences & SJTUSM, Shanghai, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Guo S, Xu J, Denning W, Hel Z. Induction of protective cytotoxic T-cell responses by a B-cell-based cellular vaccine requires stable expression of antigen. Gene Ther 2009; 16:1300-13. [PMID: 19641529 PMCID: PMC2783822 DOI: 10.1038/gt.2009.93] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
B cell-based cellular vaccines represent a promising approach to active immunotherapy of cancer complementing the use of dendritic cells, especially in pediatric patients and patients with low bone marrow reserves. B cells can be easily prepared in large numbers and readily home to secondary lymphoid organs, the primary site of induction of cytotoxic T lymphocyte (CTL) responses. However, most B cell-based vaccines tested so far failed to induce functional and protective CTLs in in vivo models. Here we demonstrate that B cells activated via the Toll like receptor-9 (TLR-9) and CD40 up-regulate surface expression of MHC and costimulatory molecules, produce IL-12, and exhibit potent antigen-presenting properties in vitro. Importantly, while administration of peptide-coated or transiently transfected B cells fails to induce immune responses, therapeutic immunization with low numbers of genetically modified B cells stably expressing antigen results in an induction of functional CTLs and protection against the growth of tumor in an animal model. Following activation, B cells partially loose their ability to home to organized lymphoid tissue due to the shedding of CD62L; however, this property can be restored by expression of protease-resistant mutant of CD62L. In summary, the data presented in this report suggest that genetically modified activated B cells represent a promising candidate for a cancer vaccine eliciting functional systemic CTLs.
Collapse
Affiliation(s)
- S Guo
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294-2170, USA
| | | | | | | |
Collapse
|
22
|
Peripheral blood lymphocytes genetically modified to express the self/tumor antigen MAGE-A3 induce antitumor immune responses in cancer patients. Blood 2008; 113:1651-60. [PMID: 19074732 DOI: 10.1182/blood-2008-07-168666] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Dendritic cell (DC) targeting in vivo has recently been shown to confer strong and protective cytotoxic T lymphocyte (CTL)-based immunity in tumor murine models. Our group has recently demonstrated in preclinical models that the infusion of genetically modified lymphocytes (GMLs) expressing the self/tumor antigen TRP-2 is able to elicit functional TRP-2-specific effectors with antitumor activity by targeting DCs in vivo. Here we have analyzed vaccine- and tumor-specific immune responses of 10 melanoma patients treated with autologous GMLs expressing the cancer germline gene MAGE-A3. Three of 10 patients treated with MAGE-A3-GML showed an increase of circulating anti-MAGE-A3 T cells, and developed skin delayed-type hypersensitivity to MAGE-A3. Interestingly, in 2 of these patients, with progressive and measurable tumors at study entry, anti-MAGE-A3 T cells were detected not only in the blood but also within tumors resected after vaccination. These results demonstrate that the infusion of MAGE-A3-GML elicits antitumor T cells, which are capable of trafficking to inflamed tissues and of infiltrating tumors. Clinical studies on a larger group of patients are needed to evaluate the clinical efficacy of such a strategy.
Collapse
|
23
|
Park MY, Kim HS, Woo SJ, Kim CH, Park JS, Sohn HJ, Kim HJ, Oh ST, Kim TG. Efficient antitumor immunity in a murine colorectal cancer model induced by CEA RNA-electroporated B cells. Eur J Immunol 2008; 38:2106-17. [DOI: 10.1002/eji.200737960] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|