1
|
Breast Cancer Vaccine Containing a Novel Toll-like Receptor 7 Agonist and an Aluminum Adjuvant Exerts Antitumor Effects. Int J Mol Sci 2022; 23:ijms232315130. [PMID: 36499455 PMCID: PMC9741412 DOI: 10.3390/ijms232315130] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 11/26/2022] [Accepted: 11/28/2022] [Indexed: 12/03/2022] Open
Abstract
Mucin 1 (MUC1) has received increasing attention due to its high expression in breast cancer, in which MUC1 acts as a cancer antigen. Our group has been committed to the development of small-molecule TLR7 (Toll-like receptor 7) agonists, which have been widely investigated in the field of tumor immunotherapy. In the present study, we constructed a novel tumor vaccine (SZU251 + MUC1 + Al) containing MUC1 and two types of adjuvants: a TLR7 agonist (SZU251) and an aluminum adjuvant (Al). Immunostimulatory responses were first verified in vitro, where the vaccine promoted the release of cytokines and the expression of costimulatory molecules in mouse BMDCs (bone marrow dendritic cells) and spleen lymphocytes. Then, we demonstrated that SZU251 + MUC1 + Al was effective and safe against a tumor expressing the MUC1 antigen in both prophylactic and therapeutic schedules in vivo. The immune responses in vivo were attributed to the increase in specific humoral and cellular immunity, including antibody titers, CD4+, CD8+ and activated CD8+ T cells. Therefore, our vaccine candidate may have beneficial effects in the prevention and treatment of breast cancer patients.
Collapse
|
2
|
Disis ML, Cecil DL. Breast cancer vaccines for treatment and prevention. Breast Cancer Res Treat 2021; 191:481-489. [PMID: 34846625 DOI: 10.1007/s10549-021-06459-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 11/15/2021] [Indexed: 12/22/2022]
Abstract
Breast cancer is immunogenic and a variety of vaccines have been designed to boost immunity directed against the disease. The components of a breast cancer vaccine, the antigen, the delivery system, and the adjuvant, can have a significant impact on vaccine immunogenicity. There have been numerous immunogenic proteins identified in all subtypes of breast cancer. The majority of these antigens are weakly immunogenic nonmutated tumor-associated proteins. Mutated proteins and neoantigen epitopes are found only in a small minority of patients and are enriched in the triple negative subtype. Several vaccines have advanced to large randomized Phase II or Phase III clinical trials. None of these trials met their primary endpoint of either progression-free or overall survival. Despite these set-backs investigators have learned important lessons regarding the clinical application of breast cancer vaccines from the type of immune response needed for tumor eradication, Type I T-cell immunity, to the patient populations most likely to benefit from vaccination. Many therapeutic breast cancer vaccines are now being tested in combination with other forms of immune therapy or chemotherapy and radiation. Breast cancer vaccines as single agents are now studied in the context of the prevention of relapse or development of disease. Newer approaches are designing vaccines to prevent breast cancer by intercepting high-risk lesions such as ductal carcinoma in situ to limit the progression of these tumors to invasive cancer. There are also several efforts to develop vaccines for the primary prevention of breast cancer by targeting antigens expressed during breast cancer initiation.
Collapse
Affiliation(s)
- Mary L Disis
- Cancer Vaccine Institute, University of Washington, Seattle, WA, USA.
| | - Denise L Cecil
- Cancer Vaccine Institute, University of Washington, Seattle, WA, USA
| |
Collapse
|
3
|
Formenti SC, Lee P, Adams S, Goldberg JD, Li X, Xie MW, Ratikan JA, Felix C, Hwang L, Faull KF, Sayre JW, Hurvitz S, Glaspy JA, Comin-Anduix B, Demaria S, Schaue D, McBride WH. Focal Irradiation and Systemic TGFβ Blockade in Metastatic Breast Cancer. Clin Cancer Res 2018; 24:2493-2504. [PMID: 29476019 PMCID: PMC5999326 DOI: 10.1158/1078-0432.ccr-17-3322] [Citation(s) in RCA: 199] [Impact Index Per Article: 28.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Revised: 01/03/2018] [Accepted: 02/19/2018] [Indexed: 12/16/2022]
Abstract
Purpose: This study examined the feasibility, efficacy (abscopal effect), and immune effects of TGFβ blockade during radiotherapy in metastatic breast cancer patients.Experimental Design: Prospective randomized trial comparing two doses of TGFβ blocking antibody fresolimumab. Metastatic breast cancer patients with at least three distinct metastatic sites whose tumor had progressed after at least one line of therapy were randomized to receive 1 or 10 mg/kg of fresolimumab, every 3 weeks for five cycles, with focal radiotherapy to a metastatic site at week 1 (three doses of 7.5 Gy), that could be repeated to a second lesion at week 7. Research bloods were drawn at baseline, week 2, 5, and 15 to isolate PBMCs, plasma, and serum.Results: Twenty-three patients were randomized, median age 57 (range 35-77). Seven grade 3/4 adverse events occurred in 5 of 11 patients in the 1 mg/kg arm and in 2 of 12 patients in the 10 mg/kg arm, respectively. Response was limited to three stable disease. At a median follow up of 12 months, 20 of 23 patients are deceased. Patients receiving the 10 mg/kg had a significantly higher median overall survival than those receiving 1 mg/kg fresolimumab dose [hazard ratio: 2.73 with 95% confidence interval (CI), 1.02-7.30; P = 0.039]. The higher dose correlated with improved peripheral blood mononuclear cell counts and a striking boost in the CD8 central memory pool.Conclusions: TGFβ blockade during radiotherapy was feasible and well tolerated. Patients receiving the higher fresolimumab dose had a favorable systemic immune response and experienced longer median overall survival than the lower dose group. Clin Cancer Res; 24(11); 2493-504. ©2018 AACR.
Collapse
Affiliation(s)
- Silvia C Formenti
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY.
| | - Percy Lee
- Department of Radiation Oncology, University of California, Los Angeles, California
- Jonsson Compressive Cancer Center, University of California, Los Angeles, California
| | - Sylvia Adams
- Department of Medicine, New York University School of Medicine, New York, NY
| | - Judith D Goldberg
- Department of Population Health, New York University School of Medicine, New York, NY
- Department of Environmental Medicine, New York University School of Medicine, New York, NY
| | - Xiaochun Li
- Department of Population Health, New York University School of Medicine, New York, NY
- Department of Environmental Medicine, New York University School of Medicine, New York, NY
| | - Mike W Xie
- Department of Radiation Oncology, University of California, Los Angeles, California
| | - Josephine A Ratikan
- Department of Radiation Oncology, University of California, Los Angeles, California
| | - Carol Felix
- Department of Radiation Oncology, University of California, Los Angeles, California
| | - Lin Hwang
- Jonsson Compressive Cancer Center, University of California, Los Angeles, California
| | - Kym F Faull
- Pasarow Mass Spectrometry Laboratory at University of California, Los Angeles, California
| | - James W Sayre
- Public Health Biostatistics at University of California, Los Angeles, California
| | - Sara Hurvitz
- Jonsson Compressive Cancer Center, University of California, Los Angeles, California
- Medicine, Hematology & Oncology at University of California, Los Angeles, California
| | - John A Glaspy
- Jonsson Compressive Cancer Center, University of California, Los Angeles, California
- Medicine, Hematology & Oncology at University of California, Los Angeles, California
| | - Begoña Comin-Anduix
- Jonsson Compressive Cancer Center, University of California, Los Angeles, California
- Medicine, Hematology & Oncology at University of California, Los Angeles, California
| | - Sandra Demaria
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, NY
| | - Dörthe Schaue
- Department of Radiation Oncology, University of California, Los Angeles, California
- Jonsson Compressive Cancer Center, University of California, Los Angeles, California
| | - William H McBride
- Department of Radiation Oncology, University of California, Los Angeles, California.
- Jonsson Compressive Cancer Center, University of California, Los Angeles, California
| |
Collapse
|
4
|
Scheikl-Gatard T, Tosch C, Lemonnier F, Rooke R. Identification of new MUC1 epitopes using HLA-transgenic animals: implication for immunomonitoring. J Transl Med 2017; 15:154. [PMID: 28679396 PMCID: PMC5499006 DOI: 10.1186/s12967-017-1254-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2017] [Accepted: 06/24/2017] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The success of immunotherapeutics in oncology and the search for further improvements has prompted revisiting the use of cancer vaccines. In this context, knowledge of the immunogenic epitopes and the monitoring of the immune response cancer vaccines generate are essential. MUC1 has been considered one of the most important tumor associated antigen for decades. METHODS To identify HLA-restricted MUC1 peptides we used eight human MHC class I transgenic mouse lines, together covering more than 80% of the human population. MUC1 peptides were identified by vaccinating each line with full length MUC1 coding sequences and using an IFNγ ELIspot restimulation assay. Relevant peptides were tested in a flow cytometry-based tetramer assay and for their capacity to serve as target in an in vivo killing assay. RESULTS Four previously identified MUC1 peptides were confirmed and five are described here for the first time. These nine peptide-MHC combinations were further characterized. Six gave above-background tetramer staining of splenocytes from immunized animals and three peptides were induced more than 5% specific in vivo killing. CONCLUSIONS These data describe for the first time five new HLA class I-restricted peptides and revisit some that were previously described. They also emphasize the importance of using in vivo/ex vivo models to screen for immunogenic peptides and define the functions for individual peptide-HLA combinations.
Collapse
Affiliation(s)
| | - Caroline Tosch
- Transgene SA, 400 Bld Gonthier d'Andernach, 67400, Illkirch Graffenstaden, France
| | - François Lemonnier
- Unité INSERM 1016, Département Endocrinologie, Métabolisme et Diabète. Equipe Immunologie des Diabètes, Bâtiment Cassini, 123 Bd Port Royal, 75014, Paris, France
| | - Ronald Rooke
- Institut de Recherche Servier, 125 Chemin de Ronde, 78290, Croissy, France.
| |
Collapse
|
5
|
In Vitro Assessment of the Expression and T Cell Immunogenicity of the Tumor-Associated Antigens BORIS, MUC1, hTERT, MAGE-A3 and Sp17 in Uterine Cancer. Int J Mol Sci 2016; 17:ijms17091525. [PMID: 27618037 PMCID: PMC5037800 DOI: 10.3390/ijms17091525] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Revised: 08/24/2016] [Accepted: 08/30/2016] [Indexed: 11/25/2022] Open
Abstract
Background: While immunotherapy moved to the forefront of treatment of various cancers, it remains underexplored for uterine cancer. This might be due to the small patient population with advanced endometrial carcinoma and uterine sarcoma. Data about immunotherapeutic targets are scarce in endometrial carcinoma and lacking in uterine sarcoma. Methods: Expression of five tumor-associated antigens (TAA) (BORIS, MUC1, hTERT, MAGE-A3 and Sp17) was validated in uterine tumor samples by immunohistochemistry (IHC) and/or quantitative reverse-transcriptase polymerase chain reaction (qRT-PCR). TAA immunogenicity was analyzed by determining spontaneous T cell responses towards overlapping peptide pools covering the whole TAA in patient blood. Results: At mRNA level, MAGE-A3 and Sp17 were overexpressed in a minority of patients and BORIS was moderately overexpressed (26% in endometrial carcinoma and 62% in uterine sarcoma). hTERT was overexpressed in the vast majority of tumors. On protein level, MUC1 was upregulated in primary, recurrent and metastatic EMCAR and in metastatic US tumors. hTERT protein was highly expressed in both normal and malignant tissue. Spontaneous TAA-specific T cell responses were detected in a minority of patients, except for hTERT to which T cell responses occurred more frequently. Conclusions: These data point to MUC1 and hTERT as most suitable targets based on expression levels and T cell immunogenicity for use in immunotherapeutic regimens.
Collapse
|
6
|
ATZIN-MÉNDEZ J, LÓPEZ-GONZÁLEZ J, BÁEZ R, ARENAS-DEL ANGEL M, MONTAÑO L, SILVA-ADAYA D, LASCURAIN R, GOROCICA P. Expansion of quiescent lung adenocarcinoma CD8+ T cells by MUC1-8-mer peptide-T2 cell-β2 microglobulin complexes. Oncol Rep 2016; 35:33-42. [PMID: 26498650 PMCID: PMC4699617 DOI: 10.3892/or.2015.4328] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Accepted: 08/21/2015] [Indexed: 12/20/2022] Open
Abstract
Adoptive immunotherapy requires the isolation of CD8+ T cells specific for tumor-associated antigens, their expansion in vitro and their transfusion to the patient to mediate a therapeutic effect. MUC1 is an important adenocarcinoma antigen immunogenic for T cells. The MUC1-derived SAPDTRPA (MUC1-8-mer) peptide is a potent epitope recognized by CD8+ T cells in murine models. Likewise, the T2 cell line has been used as an antigen-presenting cell to activate CD8+ T cells, but so far MUC1 has not been assessed in this context. We evaluated whether the MUC1-8-mer peptide can be presented by T2 cells to expand CD25+CD8+ T cells isolated from HLA-A2+ lung adenocarcinoma patients with stage III or IV tumors. The results showed that MUC1-8-mer peptide-loaded T2 cells activated CD8+ T cells from cancer HLA-A2+ patients when anti-CD2, anti-CD28 antibodies and IL-2 were added. The percentage of CD25+CD8+ T cells was 3-fold higher than those in the non-stimulated cells (P=0.018). HLA-A2+ patient cells showed a significant difference (2.3-fold higher) in activation status than HLA-A2+ healthy control cells (P=0.04). Moreover, 77.6% of MUC1-8-mer peptide-specific CD8+ T cells proliferated following a second stimulation with MUC1-8-mer peptide-loaded T2 cells after 10 days of cell culture. There were significant differences in the percentage of basal CD25+CD8+ T cells in relation to the cancer stage; this difference disappeared after MUC1-8-mer peptide stimulation. In conclusion, expansion of CD25+CD8+ T cells by MUC1-8 peptide-loaded T2 cells plus costimulatory signals via CD2, CD28 and IL-2 can be useful in adoptive immunotherapy.
Collapse
Affiliation(s)
- J.A. ATZIN-MÉNDEZ
- Department of Research in Biochemistry, National Institute of Respiratory Diseases 'Ismael Cosio Villegas', Mexico, DF 14080, Mexico
| | - J.S. LÓPEZ-GONZÁLEZ
- Lung Cancer Laboratory, National Institute of Respiratory Diseases 'Ismael Cosio Villegas', Mexico, DF 14080, Mexico
| | - R. BÁEZ
- Clinical Oncology and Pneumology, National Institute of Respiratory Diseases 'Ismael Cosio Villegas', Mexico, DF 14080, Mexico
| | - M.C. ARENAS-DEL ANGEL
- Department of Biochemistry, National Autonomous University of Mexico, Mexico, DF 04510, Mexico
| | - L.F. MONTAÑO
- Immunobiology Laboratory, Department of Cell and Tissue Biology, Faculty of Medicine, National Autonomous University of Mexico, Mexico, DF 04510, Mexico
| | - D. SILVA-ADAYA
- Experimental Laboratory for Neurodegenerative Diseases, National Institute of Neurology and Neurosurgery, Mexico, DF 14269, Mexico
| | - R. LASCURAIN
- Department of Research in Biochemistry, National Institute of Respiratory Diseases 'Ismael Cosio Villegas', Mexico, DF 14080, Mexico
- Department of Biochemistry, National Autonomous University of Mexico, Mexico, DF 04510, Mexico
| | - P. GOROCICA
- Department of Research in Biochemistry, National Institute of Respiratory Diseases 'Ismael Cosio Villegas', Mexico, DF 14080, Mexico
| |
Collapse
|
7
|
Kulikova EV, Kurilin VV, Shevchenko JA, Obleukhova IA, Khrapov EA, Boyarskikh UA, Filipenko ML, Shorokhov RV, Yakushenko VK, Sokolov AV, Sennikov SV. Dendritic Cells Transfected with a DNA Construct Encoding Tumour-associated Antigen Epitopes Induce a Cytotoxic Immune Response Against Autologous Tumour Cells in a Culture of Mononuclear Cells from Colorectal Cancer Patients. Scand J Immunol 2015; 82:110-7. [PMID: 25966778 DOI: 10.1111/sji.12311] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Accepted: 05/02/2015] [Indexed: 12/30/2022]
Abstract
Significant effort has been devoted to developing effective cancer vaccines based on dendritic cells (DCs) loaded with various tumour antigens, including DNA constructs that carry sequences of tumour-associated antigens (TAAs). Such vaccines efficiently and selectively activate the T cell immune response. In this study, we describe a method to induce an antitumour immune response in mononuclear cell (MNC) cultures from colorectal cancer patients using DNA-transfected DCs encoding TAA epitopes of carcinoembryonic antigen, epithelial cell adhesion molecule and mucin 4. DCs were obtained from peripheral blood monocytes of colorectal cancer patients. Magnetic-assisted transfection was used to deliver the genetic constructs to DCs. To assess the potency of the immune response, the antitumour cytotoxic response was assessed by lymphocyte intracellular perforin and the MNC cytotoxic activity against autologous tumour cells. We showed that polyepitope DNA-transfected DCs enhanced MNC antitumour activity, increasing tumour cell death and the percentage of perforin-positive lymphocytes. In addition, DNA-transfected DCs elicited a cytotoxic response that was as efficient as that of tumour lysate-loaded DCs. Taken together, the data suggest that it is feasible to induce an antitumour immune response in colorectal MNCs using transfected DCs. Thus, the DNA construct reported in this study may potentially be used in therapeutic and prophylactic DC-based vaccines.
Collapse
Affiliation(s)
- E V Kulikova
- Federal State Budgetary Scientific Institution "Research Institute of Fundamental and Clinical Immunology", Novosibirsk, Russian Federation
| | - V V Kurilin
- Federal State Budgetary Scientific Institution "Research Institute of Fundamental and Clinical Immunology", Novosibirsk, Russian Federation
| | - J A Shevchenko
- Federal State Budgetary Scientific Institution "Research Institute of Fundamental and Clinical Immunology", Novosibirsk, Russian Federation
| | - I A Obleukhova
- Federal State Budgetary Scientific Institution "Research Institute of Fundamental and Clinical Immunology", Novosibirsk, Russian Federation
| | - E A Khrapov
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russian Federation
| | - U A Boyarskikh
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russian Federation
| | - M L Filipenko
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russian Federation
| | - R V Shorokhov
- City Clinical Hospital No. 1, Novosibirsk, Russian Federation
| | - V K Yakushenko
- City Clinical Hospital No. 11, Novosibirsk, Russian Federation
| | - A V Sokolov
- City Clinical Hospital No. 1, Novosibirsk, Russian Federation
| | - S V Sennikov
- Federal State Budgetary Scientific Institution "Research Institute of Fundamental and Clinical Immunology", Novosibirsk, Russian Federation
| |
Collapse
|
8
|
Blessing or curse? Proteomics in granzyme research. Proteomics Clin Appl 2014; 8:351-81. [DOI: 10.1002/prca.201300096] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2013] [Revised: 11/29/2013] [Accepted: 12/21/2013] [Indexed: 01/08/2023]
|
9
|
Peripheral blood mononuclear cells of patients with breast cancer can be reprogrammed to enhance anti-HER-2/neu reactivity and overcome myeloid-derived suppressor cells. Breast Cancer Res Treat 2013; 142:45-57. [PMID: 24197563 DOI: 10.1007/s10549-013-2733-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2013] [Accepted: 10/10/2013] [Indexed: 01/04/2023]
Abstract
Two major barriers in the immunotherapy of breast cancer include tumor-induced immune suppression and the establishment of long-lasting immune responses against the tumor. Recently, we demonstrated in an animal model of breast carcinoma that expanding and reprogramming tumor-sensitized lymphocytes, ex vivo, yielded T memory (Tm) cells as well as activated CD25+ NKT cells and NK cells. The presence of activated CD25+ NKT and NK cells rendered reprogrammed T cells resistant to MDSC-mediated suppression, and adoptive cellular therapy (ACT) of reprogrammed lymphocytes protected the host from tumor development and relapse. Here, we performed a pilot study to determine the clinical applicability of our protocol using peripheral blood mononuclear cells (PBMCs) of breast cancer patients, ex vivo. We show that bryostatin 1 and ionomycin combined with IL-2, IL-7, and IL-15 can expand and reprogram tumor-sensitized PBMCs. Reprogrammed lymphocytes contained activated CD25+ NKT and NK cells as well as Tm cells and displayed enhanced reactivity against HER-2/neu in the presence of MDSCs. The presence of activated NKT cells was highly correlated with the rescue of anti-HER-2/neu immune responses from MDSC suppression. Ex vivo blockade experiments suggest that the NKG2D pathway may play an important role in overcoming MDSC suppression. Our results show the feasibility of reprogramming tumor-sensitized immune cells, ex vivo, and provide rationale for ACT of breast cancer patients.
Collapse
|
10
|
Abstract
Various strategies have been used to generate cellular cancer vaccines with the expectation that they will become an effective part of the overall management of cancer patients. However, with few notable exceptions, immunization has not resulted in significant long-term therapeutic benefits. Tumor growth has continued and patient survival has been at best only modestly prolonged. One possible explanation is that as only a small proportion of the constituents of malignant cells are "tumor specific" and the vast majority are the products of nonantigenic, normal "housekeeping" genes, the immune response in patients immunized with cellular cancer vaccines is not sufficient to result in tumor rejection. Here, we review and characterize various types of cellular cancer vaccines. In addition, in a mouse breast cancer model system, we describe a unique strategy designed to enrich cellular vaccines for cells that induce tumor immunity. Numerous advantages and disadvantages of cancer immunotherapy with cellular vaccines are also presented.
Collapse
Affiliation(s)
- Edward P Cohen
- Department of Microbiology & Immunology, University of Illinois College of Medicine, 835 South Wolcott Ave., Chicago, IL 60612, USA.
| | | | | | | |
Collapse
|
11
|
Gnjatic S, Cao Y, Reichelt U, Yekebas EF, Nölker C, Marx AH, Erbersdobler A, Nishikawa H, Hildebrandt Y, Bartels K, Horn C, Stahl T, Gout I, Filonenko V, Ling KL, Cerundolo V, Luetkens T, Ritter G, Friedrichs K, Leuwer R, Hegewisch-Becker S, Izbicki JR, Bokemeyer C, Old LJ, Atanackovic D. NY-CO-58/KIF2C is overexpressed in a variety of solid tumors and induces frequent T cell responses in patients with colorectal cancer. Int J Cancer 2010; 127:381-93. [PMID: 19937794 DOI: 10.1002/ijc.25058] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
NY-CO-58/KIF2C has been identified as a tumor antigen by screening antibody responses in patients with colorectal cancer. However, expression had not consequently been examined, and nothing was known about its ability to induce spontaneous T cell responses, which have been suggested to play a role in the development of colorectal cancer. We analyzed 5 colorectal cancer cell lines, and tumor samples and adjacent healthy tissues from 176 patients with epithelial cancers for the expression of NY-CO-58/KIF2C by RT-PCR and Western Blot. T cell responses of 43 colorectal cancer patients and 35 healthy donors were evaluated by ELISpot following stimulation with 30mer peptides or full-length protein. All cell lines and tumor samples from colorectal cancer patients expressed NY-CO-58/KIF2C on the protein and RNA level, and expression levels correlated strongly with Ki-67 expression (r = 0.69; p = 0.0003). Investigating NY-CO-58/KIF2C-specific T cell responses, CD8(+) T cells directed against 1 or more peptides were found in less than 10% of patients, whereas specific CD4(+) T cells were detected in close to 50% of patients. These T cells were of high avidity, recognized the naturally processed antigen and secreted IFN-gamma and TNF-alpha. Depletion of CD4(+)CD25(+) T cells before stimulation significantly increased the intensity of the preexisting response. NY-CO-58/KIF2C is significantly overexpressed in colorectal and other epithelial cancers and expression levels correlate with the proliferative activity of the tumor. Importantly, NY-CO-58/KIF2C was able to induce spontaneous CD4(+) T cell responses of the Th1-type, which were tightly controlled by peripheral T regulatory cells.
Collapse
Affiliation(s)
- Sacha Gnjatic
- Ludwig Institute for Cancer Research, Memorial Sloan-Kettering Cancer Center, New York, NY, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
CD8+ T-cell responses to tumor-associated antigens correlate with superior relapse-free survival after allo-SCT. Bone Marrow Transplant 2009; 43:399-410. [PMID: 19139738 DOI: 10.1038/bmt.2008.426] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The GVL effect following allo-SCT is one of the most prominent examples showing the ability of the immune system to eliminate malignant hematological diseases. Tumor-associated Ags (TAA), for instance WT1 and proteinase-3, have been proposed as targets for T cells to establish a GVL effect. Here, we examined an additional TAA (MUC1) as a possible T-cell target of GVL-related immune responses. We have defined new peptide epitopes from the MUC1 Ag to broaden patients' screening and to expand the repertoire of immunologic monitoring as well as for therapeutic approaches in the future. Twenty-eight patients after allo-SCT have been screened for T-cell responses toward TAA (proteinase-3, WT1, MUC1). We could detect a significant relationship between relapse and the absence of a TAA-specific T-cell response, whereby only 2/13 (15%) patients with TAA-specific CTL relapsed, in contrast to 9/15 (60%) patients without TAA-specific CTL responses (P<0.05). In conclusion, CD8(+) T-cell responses directed to TAA might contribute to the GVL effect. These observations highlight both the importance and the potential of immunotherapeutic approaches after allo-SCT.
Collapse
|
13
|
Kokowski K, Harnack U, Dorn DC, Pecher G. Quantification of the CD8+ T cell response against a mucin epitope in patients with breast cancer. Arch Immunol Ther Exp (Warsz) 2008; 56:141-5. [PMID: 18373244 PMCID: PMC2766492 DOI: 10.1007/s00005-008-0011-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2007] [Accepted: 01/25/2008] [Indexed: 01/28/2023]
Abstract
Introduction: Mucin 1, encoded by the MUC1 gene, is a tumor-associated antigen expressed on the surface of breast cancer cells. It would be of interest to see whether there is a naturally existing T cell immune response against mucin epitopes in cancer patients. Materials and Methods: Using tetramer and interferon-γ assays, the immune response to one MUC1 peptide epitope in the peripheral blood of breast cancer patients was quantified. The data were compared with the clinical course of the patients. Results: CD8+ T cells capable of recognizing the HLA-A*0201-restricted STAPPVHNV epitope were detected in 9 of 19 patients with a frequency ranging 0.01–0.082%. No significant difference was found between the occurrence of epitope-specific CD8+ T cells of patients with progressive disease and disease-free patients. However, all patients with stable disease showed a specific immune response, including both patients with the highest frequency. Conclusions: The results of this study provide further evidence that a natural specific cellular immune response against this mucin epitope exists in breast cancer patients.
Collapse
Affiliation(s)
- Konrad Kokowski
- Medical Clinic of Oncology and Hematology, Charité-Universitätsmedizin Berlin, Charité Campus Mitte, Charitéplatz 1, 10117 Berlin, Germany
| | - Ulf Harnack
- Medical Clinic of Oncology and Hematology, Charité-Universitätsmedizin Berlin, Charité Campus Mitte, Charitéplatz 1, 10117 Berlin, Germany
| | - David C. Dorn
- Medical Clinic of Oncology and Hematology, Charité-Universitätsmedizin Berlin, Charité Campus Mitte, Charitéplatz 1, 10117 Berlin, Germany
- Laboratory of Developmental Hematopoiesis, Cell Biology Program, Memorial Sloan-Kettering Cancer Center, New York, 10021 NY USA
| | - Gabriele Pecher
- Medical Clinic of Oncology and Hematology, Charité-Universitätsmedizin Berlin, Charité Campus Mitte, Charitéplatz 1, 10117 Berlin, Germany
| |
Collapse
|
14
|
Knutson KL, Disis ML, Salazar LG. CD4 regulatory T cells in human cancer pathogenesis. Cancer Immunol Immunother 2007; 56:271-85. [PMID: 16819631 PMCID: PMC11030088 DOI: 10.1007/s00262-006-0194-y] [Citation(s) in RCA: 88] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2006] [Accepted: 06/09/2006] [Indexed: 12/18/2022]
Abstract
Over the past decade, there has been an accelerated understanding of immune regulatory mechanisms. Peripheral immune regulation is linked to a collection of specialized regulatory cells of the CD4(+) T cell lineage (i.e., CD4(+) Tregs). This collection consists of Tregs that are either thymically derived (i.e., natural) or peripherally induced. Tregs are important for controlling potentially autoreactive immune effectors and immunity to foreign organisms and molecules. Their importance in maintaining immune homeostasis and the overall health of an organism is clear. However, Tregs may also be involved in the pathogenesis of malignancies as now compelling evidence shows that tumors induce or recruit CD4(+) Tregs to block immune priming and antitumor effectors. Efforts are underway to develop approaches that specifically inhibit the function of tumor-associated Tregs which could lead to an increased capability of the body's immune system to respond to tumors but without off-target immune-related pathologies (i.e., autoimmune disease). In this review, the biology of human CD4(+) Tregs is discussed along with their involvement in malignancies and emerging strategies to block their function.
Collapse
Affiliation(s)
- Keith L Knutson
- Department of Immunology, Mayo Clinic College of Medicine, Mayo Clinic, 342C Guggenheim, 200 First Street SW, Rochester, MN 55905, USA.
| | | | | |
Collapse
|
15
|
Knutson KL. Strong-arming immune regulation: suppressing regulatory T-cell function to treat cancers. Future Oncol 2006; 2:379-89. [PMID: 16787118 DOI: 10.2217/14796694.2.3.379] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
In recent years there has been an accelerated understanding of immune regulatory mechanisms. Much of this immune regulation is linked to a collection of specialized regulatory cells of the T-cell lineage (Tregs). This collection consists of Tregs that are either thymically derived or peripherally induced. Tregs are important for controlling potentially autoreactive immune effectors and immune responses to foreign organisms and molecules. Their importance in maintaining immune homeostasis and the overall health of an organism cannot be overstated. However, there is a dark side, and Tregs may also be involved in the pathogenesis of malignancies. Evidence shows that tumors induce or recruit Tregs to block antitumor effectors. Thus, there are efforts underway to identify approaches that specifically inhibit the function of intratumoral Tregs, which could lead to increased immunity to tumors without off-target immune-related pathologies (i.e., autoimmune disease). In this review, the biology of Tregs is discussed along with their involvement in malignancies and emerging strategies to block their function.
Collapse
Affiliation(s)
- Keith L Knutson
- Department of Immunology, Mayo Clinic College of Medicine, 342C Guggenheim, 200 First St. SW, Mayo Clinic, Rochester, MN 55905, USA.
| |
Collapse
|
16
|
Knutson KL, Krco CJ, Erskine CL, Goodman K, Kelemen LE, Wettstein PJ, Low PS, Hartmann LC, Kalli KR. T-cell immunity to the folate receptor alpha is prevalent in women with breast or ovarian cancer. J Clin Oncol 2006; 24:4254-61. [PMID: 16908932 DOI: 10.1200/jco.2006.05.9311] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE Studies have demonstrated that the generation of immunity to tumor antigens is associated with improved prognosis for many cancers. A candidate antigen is the folate receptor alpha (FRalpha), which is overexpressed in breast and ovarian cancers. Our goal in this study was to attain a better understanding of the extent of endogenous FRalpha immunity. METHODS Using a CD4+ T cell epitope prediction algorithm, we predicted promiscuous epitopes of FRalpha, and tested for immunity in 30 breast (n = 17) or ovarian (n = 13) cancer patients and 18 healthy donors using enzyme-linked immunospot analysis. RESULTS Fourteen peptides were predicted, seven each from the carboxy- and amino-terminus halves of the protein. More than 70% of patients demonstrated immunity to at least one FRalpha peptide. Patients responded to an average of 3 +/- 0.5 peptides, whereas healthy donors responded to 1 +/- 0.4 peptides (P = .004). Five peptides were recognized by more than 25% of patients. Responses to three peptides were higher (P < .05) in patients than in healthy donors, suggesting augmented immunity. Compared with healthy individuals, patients developed higher immunity to the amino-terminus half of the receptor (P = .03). There was no difference between each group in the responses to nonspecific (P = .2) and viral stimuli (P = .5). Lastly, patients demonstrated elevated levels of FRalpha antibodies consistent with a coordinated immune response. CONCLUSION These findings demonstrate that the FRalpha is a target of the immune system in breast and ovarian cancer patients. Understanding which antigens are targeted by the immune system may be important for prognosis or immune-based therapies.
Collapse
Affiliation(s)
- Keith L Knutson
- Department of Immunology, Mayo Clinic College of Medicine, Rochester, MN 55905, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|