1
|
Alarcon NO, Jaramillo M, Mansour HM, Sun B. Therapeutic Cancer Vaccines—Antigen Discovery and Adjuvant Delivery Platforms. Pharmaceutics 2022; 14:pharmaceutics14071448. [PMID: 35890342 PMCID: PMC9325128 DOI: 10.3390/pharmaceutics14071448] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 06/28/2022] [Accepted: 06/30/2022] [Indexed: 12/15/2022] Open
Abstract
For decades, vaccines have played a significant role in protecting public and personal health against infectious diseases and proved their great potential in battling cancers as well. This review focused on the current progress of therapeutic subunit vaccines for cancer immunotherapy. Antigens and adjuvants are key components of vaccine formulations. We summarized several classes of tumor antigens and bioinformatic approaches of identification of tumor neoantigens. Pattern recognition receptor (PRR)-targeting adjuvants and their targeted delivery platforms have been extensively discussed. In addition, we emphasized the interplay between multiple adjuvants and their combined delivery for cancer immunotherapy.
Collapse
Affiliation(s)
- Neftali Ortega Alarcon
- Skaggs Pharmaceutical Sciences Center, College of Pharmacy, The University of Arizona, Tucson, AZ 85721, USA; (N.O.A.); (M.J.); (H.M.M.)
| | - Maddy Jaramillo
- Skaggs Pharmaceutical Sciences Center, College of Pharmacy, The University of Arizona, Tucson, AZ 85721, USA; (N.O.A.); (M.J.); (H.M.M.)
| | - Heidi M. Mansour
- Skaggs Pharmaceutical Sciences Center, College of Pharmacy, The University of Arizona, Tucson, AZ 85721, USA; (N.O.A.); (M.J.); (H.M.M.)
- The University of Arizona Cancer Center, Tucson, AZ 85721, USA
- Department of Medicine, College of Medicine, The University of Arizona, Tucson, AZ 85724, USA
- BIO5 Institute, The University of Arizona, Tucson, AZ 85721, USA
| | - Bo Sun
- Skaggs Pharmaceutical Sciences Center, College of Pharmacy, The University of Arizona, Tucson, AZ 85721, USA; (N.O.A.); (M.J.); (H.M.M.)
- The University of Arizona Cancer Center, Tucson, AZ 85721, USA
- BIO5 Institute, The University of Arizona, Tucson, AZ 85721, USA
- Correspondence: ; Tel.: +1-520-621-6420
| |
Collapse
|
2
|
Luangwattananun P, Chiraphapphaiboon W, Thuwajit C, Junking M, Yenchitsomanus PT. Activation of cytotoxic T lymphocytes by self-differentiated myeloid-derived dendritic cells for killing breast cancer cells expressing folate receptor alpha protein. Bioengineered 2022; 13:14188-14203. [PMID: 35734827 PMCID: PMC9342379 DOI: 10.1080/21655979.2022.2084262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
Abstract
Adoptive cell transfer (ACT) is a promising approach for cancer treatment. Activation of T lymphocytes by self-differentiated myeloid-derived antigen-presenting-cells reactive against tumor (SmartDC) resulted in specific anti-cancer function. Folate receptor alpha (FRα) is highly expressed in breast cancer (BC) cells and thus potential to be a target antigen for ACT. To explore the SmartDC technology for treatment of BC, we create SmartDC expressing FRα antigen (SmartDC-FRα) for activation of FRα-specific T lymphocytes. Human primary monocytes were transduced with lentiviruses containing tri-cistronic complementary DNA sequences encoding granulocyte-macrophage colony-stimulating factor (GM-CSF), interleukin-4 (IL-4), and FRα to generate SmartDC-FRα. Autologous T lymphocytes were activated by SmartDC-FRα by coculture. The activated T lymphocytes exhibited enhanced cytotoxicity against FRα-expressing BC cell cultures. Up to 84.9 ± 6.2% of MDA-MB-231 and 89.7 ± 1.9% of MCF-7 BC cell lines were specifically lysed at an effector-to-target ratio of 20:1. The cytotoxicity of T lymphocytes activated by SmartDC-FRα was also demonstrated in three-dimensional (3D) spheroid culture of FRα-expressing BC cells marked by size reduction and spheroid disruption. This study thus portray the potential development of T lymphocytes activated by SmartDC-FRα as ACT in FRα-expressing BC treatment.
Collapse
Affiliation(s)
- Piriya Luangwattananun
- Siriraj Center of Research Excellence for Cancer Immunotherapy (SiCORE-CIT), Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand.,Division of Molecular Medicine, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol, University, Bangkok, Thailand
| | - Wannasiri Chiraphapphaiboon
- Siriraj Center of Research Excellence for Cancer Immunotherapy (SiCORE-CIT), Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand.,Division of Molecular Medicine, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol, University, Bangkok, Thailand
| | - Chanitra Thuwajit
- Siriraj Center of Research Excellence for Cancer Immunotherapy (SiCORE-CIT), Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand.,Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Mutita Junking
- Siriraj Center of Research Excellence for Cancer Immunotherapy (SiCORE-CIT), Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand.,Division of Molecular Medicine, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol, University, Bangkok, Thailand
| | - Pa-Thai Yenchitsomanus
- Siriraj Center of Research Excellence for Cancer Immunotherapy (SiCORE-CIT), Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand.,Division of Molecular Medicine, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol, University, Bangkok, Thailand
| |
Collapse
|
3
|
The generation and application of antigen-specific T cell therapies for cancer and viral-associated disease. Mol Ther 2022; 30:2130-2152. [PMID: 35149193 PMCID: PMC9171249 DOI: 10.1016/j.ymthe.2022.02.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 11/27/2021] [Accepted: 02/03/2022] [Indexed: 11/23/2022] Open
Abstract
Immunotherapy with antigen-specific T cells is a promising, targeted therapeutic option for patients with cancer as well as for immunocompromised patients with virus infections. In this review, we characterize and compare current manufacturing protocols for the generation of T cells specific to viral and non-viral tumor-associated antigens. Specifically, we discuss: (1) the different methodologies to expand virus-specific T cell and non-viral tumor-associated antigen-specific T cell products, (2) an overview of the immunological principles involved when developing such manufacturing protocols, and (3) proposed standardized methodologies for the generation of polyclonal, polyfunctional antigen-specific T cells irrespective of donor source. Ex vivo expanded cells have been safely administered to treat numerous patients with virus-associated malignancies, hematologic malignancies, and solid tumors. Hence, we have performed a comprehensive review of the clinical trial results evaluating the safety, feasibility, and efficacy of these products in the clinic. In summary, this review seeks to provide new insights regarding antigen-specific T cell technology to benefit a rapidly expanding T cell therapy field.
Collapse
|
4
|
García-Estevez L, González-Martínez S, Moreno-Bueno G. The Leptin Axis and Its Association With the Adaptive Immune System in Breast Cancer. Front Immunol 2021; 12:784823. [PMID: 34868066 PMCID: PMC8634160 DOI: 10.3389/fimmu.2021.784823] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 10/28/2021] [Indexed: 12/21/2022] Open
Abstract
Adipose tissue secretes various peptides, including leptin. This hormone acts through the leptin receptor (Ob-R), which is expressed ubiquitously on the surface of various cells, including breast cancer cells and immune cells. Increasing evidence points to an interaction between the tumor microenvironment, tumor cells, and the immune system. Leptin plays an important role in breast cancer tumorigenesis and may be implicated in activation of the immune system. While breast cancer cannot be considered an immunogenic cancer, the triple-negative subtype is an exception. Specific immune cells - tumor infiltrating lymphocytes - are involved in the immune response and act as predictive and prognostic factors in certain breast cancer subtypes. The aim of this article is to review the interaction between adipose tissue, through the expression of leptin and its receptor, and the adaptive immune system in breast cancer.
Collapse
Affiliation(s)
- Laura García-Estevez
- Breast Cancer Department, MD Anderson Cancer Center, Madrid, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.,MD Anderson International Foundation, Madrid, Spain
| | - Silvia González-Martínez
- Pathology Department, Hospital Ramón y Cajal, Madrid, Spain.,Fundación Contigo Contra el Cáncer de la Mujer, Madrid, Spain
| | - Gema Moreno-Bueno
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.,MD Anderson International Foundation, Madrid, Spain.,Biochemistry Department, Universidad Autónoma de Madrid (UAM), Instituto de Investigaciones Biomédicas 'Alberto Sols' (CSIC-UAM), IdiPaz, Madrid, Spain
| |
Collapse
|
5
|
Chen C, Yang L, Rivandi M, Franken A, Fehm T, Neubauer H. Bioinformatic Identification of a Breast-Specific Transcript Profile. Proteomics Clin Appl 2020; 14:e2000007. [PMID: 32558282 DOI: 10.1002/prca.202000007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 04/23/2020] [Indexed: 11/10/2022]
Abstract
PURPOSE To identify a breast-specific transcript profile for the first time, and present an updated bioinformatics strategy for searching tissue-specific transcripts and predicting their significance in cancer. EXPERIMENTAL DESIGN The RNA-seq data of 49 311 transcripts in 88 human tissues from the GTEx, the Illumina Body Map, and the RIKEN FANTOM5 project are integrated to screen breast-specific transcripts. Gene Expression Profiling Interactive Analysis, TGCA, and Kaplan-Meier Plotter are used to examine their expression in cancer tissues and values for prognosis prediction. RESULTS Only 96 transcripts in human genome are breast-specific for women. Among them, ankyrin repeat domain 30A (ANKRD30A) and long intergenic non-protein coding RNA 993 (LINC00993) are further analyzed. The two transcripts are also breast-specific in 33 types of common female cancer and are often dysregulated in breast cancer tissues. Their expression is higher in the luminal breast cancer while significantly downregulated in triple-negative breast cancer. Moreover, the high expression levels of ANKRD30A and LINC0993 in breast cancer tissues indicate a better prognosis of patients with breast cancer. CONCLUSIONS AND CLINICAL RELEVANCE Breast-specific transcripts in human genome are rare and poorly understood currently. The data indicate that these breast-specific biomarkers are promising candidates for screening early cancer, assessing treatment response, monitoring recurrence, identifying metastatic tumor origin, and serving as potential targets for immunotherapy.
Collapse
Affiliation(s)
- Chen Chen
- Department of Obstetrics and Gynecology, Heinrich Heine University of Duesseldorf, Duesseldorf, 40225, Germany.,Department of Breast and Thyroid Surgery, The Third Affiliated Hospital of Zunyi Medical University (The First People's Hospital of Zunyi), Zunyi, 563000, China
| | - Liwen Yang
- Department of Obstetrics and Gynecology, Heinrich Heine University of Duesseldorf, Duesseldorf, 40225, Germany
| | - Mahdi Rivandi
- Department of Obstetrics and Gynecology, Heinrich Heine University of Duesseldorf, Duesseldorf, 40225, Germany
| | - André Franken
- Department of Obstetrics and Gynecology, Heinrich Heine University of Duesseldorf, Duesseldorf, 40225, Germany
| | - Tanja Fehm
- Department of Obstetrics and Gynecology, Heinrich Heine University of Duesseldorf, Duesseldorf, 40225, Germany
| | - Hans Neubauer
- Department of Obstetrics and Gynecology, Heinrich Heine University of Duesseldorf, Duesseldorf, 40225, Germany
| |
Collapse
|
6
|
Das K, Eisel D, Vormehr M, Müller-Decker K, Hommertgen A, Jäger D, Zörnig I, Feuerer M, Kopp-Schneider A, Osen W, Eichmüller SB. A transplantable tumor model allowing investigation of NY-BR-1-specific T cell responses in HLA-DRB1*0401 transgenic mice. BMC Cancer 2019; 19:914. [PMID: 31519152 PMCID: PMC6743128 DOI: 10.1186/s12885-019-6102-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 08/28/2019] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND NY-BR-1 has been described as a breast cancer associated differentiation antigen with intrinsic immunogenicity giving rise to endogenous T and B cell responses. The current study presents the first murine tumor model allowing functional investigation of NY-BR-1-specific immune responses in vivo. METHODS A NY-BR-1 expressing tumor model was established in DR4tg mice based on heterotopic transplantation of stable transfectant clones derived from the murine H2 compatible breast cancer cell line EO771. Composition and phenotype of tumor infiltrating immune cells were analyzed by qPCR and FACS. MHC I binding affinity of candidate CTL epitopes predicted in silico was determined by FACS using the mutant cell line RMA-S. Frequencies of NY-BR-1 specific CTLs among splenocytes of immunized mice were quantified by FACS with an epitope loaded Db-dextramer. Functional CTL activity was determined by IFNγ catch or IFNγ ELISpot assays and statistical analysis was done applying the Mann Whitney test. Tumor protection experiments were performed by immunization of DR4tg mice with replication deficient recombinant adenovirus followed by s.c. challenge with NY-BR-1 expressing breast cancer cells. RESULTS Our results show spontaneous accumulation of CD8+ T cells and F4/80+ myeloid cells preferentially in NY-BR-1 expressing tumors. Upon NY-BR-1-specific immunization experiments combined with in silico prediction and in vitro binding assays, the first NY-BR-1-specific H2-Db-restricted T cell epitope could be identified. Consequently, flow cytometric analysis with fluorochrome conjugated multimers showed enhanced frequencies of CD8+ T cells specific for the newly identified epitope in spleens of immunized mice. Moreover, immunization with Ad.NY-BR-1 resulted in partial protection against outgrowth of NY-BR-1 expressing tumors and promoted intratumoral accumulation of macrophages. CONCLUSION This study introduces the first H2-Db-resctricted CD8+ T cell epitope-specific for the human breast cancer associated tumor antigen NY-BR-1. Our novel, partially humanized tumor model enables investigation of the interplay between HLA-DR4-restricted T cell responses and CTLs within their joint attack of NY-BR-1 expressing tumors.
Collapse
Affiliation(s)
- Krishna Das
- Research Group GMP & T Cell Therapy, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Division of Virology, Innsbruck Medical University, Innsbruck, Austria.,Faculty of Biosciences, University Heidelberg, Heidelberg, Germany
| | - David Eisel
- Research Group GMP & T Cell Therapy, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Faculty of Biosciences, University Heidelberg, Heidelberg, Germany.,Biopharmaceutical New Technologies (BioNTech) Corporation, Mainz, Germany
| | - Mathias Vormehr
- Biopharmaceutical New Technologies (BioNTech) Corporation, Mainz, Germany.,University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Karin Müller-Decker
- Core Facility Tumor Models, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Adriane Hommertgen
- Research Group GMP & T Cell Therapy, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Molecular & Radiation Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Dirk Jäger
- CCU Applied Tumor Immunity, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Medical Oncology, National Center for Tumor Diseases (NCT) and University Hospital Heidelberg, Heidelberg, Germany
| | - Inka Zörnig
- Department of Medical Oncology, National Center for Tumor Diseases (NCT) and University Hospital Heidelberg, Heidelberg, Germany
| | - Markus Feuerer
- Institute of Immunology, Regensburg Center for Interventional Immunology (RCI), University Regensburg and University Hospital Regensburg, Regensburg, Germany
| | | | - Wolfram Osen
- Research Group GMP & T Cell Therapy, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Stefan B Eichmüller
- Research Group GMP & T Cell Therapy, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
7
|
Baldin AV, Zamyatnin AA, Bazhin AV, Xu WH, Savvateeva LV. Advances in the Development of Anticancer HSP-based Vaccines. Curr Med Chem 2019; 26:427-445. [PMID: 29376489 DOI: 10.2174/0929867325666180129100015] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Revised: 10/11/2017] [Accepted: 01/01/2018] [Indexed: 01/01/2023]
Abstract
Current advances in cancer treatment are based on the recent discoveries of molecular mechanisms of tumour maintenance. It was shown that heat shock proteins (HSPs) play a crucial role in the development of immune response against tumours. Thus, HSPs represent multifunctional agents not only with chaperone functions, but also possessing immunomodulatory properties. These properties are exploited for the development of HSP-based anticancer vaccines aimed to induce cytotoxic responses against tumours. To date, a number of strategies have been suggested to facilitate HSP-based vaccine production and to increase its effectiveness. The present review focuses on the current trend for the development of HSPbased vaccines aimed at inducing strong immunological tumour-specific responses against cancer cells of distinct etiology and localization.
Collapse
Affiliation(s)
- Alexey V Baldin
- Sechenov First Moscow State Medical University, Institute of Molecular Medicine, 119991, Moscow, Russian Federation
| | - Andrey A Zamyatnin
- Sechenov First Moscow State Medical University, Institute of Molecular Medicine, 119991, Moscow, Russian Federation.,Lomonosov Moscow State University, Department of Cell Signaling, Belozersky Institute of Physico- Chemical Biology, 119991, Moscow, Russian Federation
| | - Alexandr V Bazhin
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich, Germany.,German Cancer Consortium (DKTK), Partner Site Munich, Germany
| | - Wan-Hai Xu
- Department of Urology, the Fourth Hospital of Harbin Medical University, Harbin, China
| | - Lyudmila V Savvateeva
- Sechenov First Moscow State Medical University, Institute of Molecular Medicine, 119991, Moscow, Russian Federation
| |
Collapse
|
8
|
The role of proteomics in the age of immunotherapies. Mamm Genome 2018; 29:757-769. [PMID: 30046851 DOI: 10.1007/s00335-018-9763-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Accepted: 07/20/2018] [Indexed: 12/12/2022]
Abstract
The antigenic landscape of the adaptive immune response is determined by the peptides presented by immune cells. In recent years, a number of immune-based cancer therapies have been shown to induce remarkable clinical responses through the activation of the patient's immune system. As a result, there is a need to identify immune biomarkers capable of predicting clinical response. Recent advances in proteomics have led to considerable developments in the more comprehensive profiling of the immune response. "Immunoproteomics" utilises a rapidly increasing collection of technologies in order to identify and quantify antigenic peptides or proteins. This includes gel-based, array-based, mass spectrometry (MS), DNA-based, or computer-based (in silico) approaches. Immunoproteomics is yielding an understanding of disease and disease progression, vaccine candidates, and biomarkers to a depth not before understood. This review gives an overview of the emerging role of proteomics in improving personalisation of immunotherapy treatment.
Collapse
|
9
|
Rozanov DV, Rozanov ND, Chiotti KE, Reddy A, Wilmarth PA, David LL, Cha SW, Woo S, Pevzner P, Bafna V, Burrows GG, Rantala JK, Levin T, Anur P, Johnson-Camacho K, Tabatabaei S, Munson DJ, Bruno TC, Slansky JE, Kappler JW, Hirano N, Boegel S, Fox BA, Egelston C, Simons DL, Jimenez G, Lee PP, Gray JW, Spellman PT. MHC class I loaded ligands from breast cancer cell lines: A potential HLA-I-typed antigen collection. J Proteomics 2018; 176:13-23. [PMID: 29331515 DOI: 10.1016/j.jprot.2018.01.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 12/01/2017] [Accepted: 01/04/2018] [Indexed: 12/30/2022]
Abstract
To build a catalog of peptides presented by breast cancer cells, we undertook systematic MHC class I immunoprecipitation followed by elution of MHC class I-loaded peptides in breast cancer cells. We determined the sequence of 3196 MHC class I ligands representing 1921 proteins from a panel of 20 breast cancer cell lines. After removing duplicate peptides, i.e., the same peptide eluted from more than one cell line, the total number of unique peptides was 2740. Of the unique peptides eluted, more than 1750 had been previously identified, and of these, sixteen have been shown to be immunogenic. Importantly, half of these immunogenic peptides were shared between different breast cancer cell lines. MHC class I binding probability was used to plot the distribution of the eluted peptides in accordance with the binding score for each breast cancer cell line. We also determined that the tested breast cancer cells presented 89 mutation-containing peptides and peptides derived from aberrantly translated genes, 7 of which were shared between four or two different cell lines. Overall, the high throughput identification of MHC class I-loaded peptides is an effective strategy for systematic characterization of cancer peptides, and could be employed for design of multi-peptide anticancer vaccines. SIGNIFICANCE By employing proteomic analyses of eluted peptides from breast cancer cells, the current study has built an initial HLA-I-typed antigen collection for breast cancer research. It was also determined that immunogenic epitopes can be identified using established cell lines and that shared immunogenic peptides can be found in different cancer types such as breast cancer and leukemia. Importantly, out of 3196 eluted peptides that included duplicate peptides in different cells 89 peptides either contained mutation in their sequence or were derived from aberrant translation suggesting that mutation-containing epitopes are on the order of 2-3% in breast cancer cells. Finally, our results suggest that interfering with MHC class I function is one of the mechanisms of how tumor cells escape immune system attack.
Collapse
Affiliation(s)
- Dmitri V Rozanov
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, OR, United States.
| | | | - Kami E Chiotti
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, OR, United States
| | - Ashok Reddy
- Proteomics Shared Resource, Oregon Health and Science University, Portland, OR, United States
| | - Phillip A Wilmarth
- Proteomics Shared Resource, Oregon Health and Science University, Portland, OR, United States
| | - Larry L David
- Proteomics Shared Resource, Oregon Health and Science University, Portland, OR, United States
| | - Seung W Cha
- Electrical and Computer Engineering, University of California, San Diego, CA, United States
| | - Sunghee Woo
- School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Pavel Pevzner
- The NIH Center for Computational Mass Spectrometry, University of California, San Diego, San Diego, CA, United States
| | - Vineet Bafna
- Computer Science & Engineering, University of California, San Diego, CA, United States
| | - Gregory G Burrows
- Neurology and Biochemistry & Molecular Biology, Oregon Health and Science University, Portland, OR, United States
| | | | - Trevor Levin
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, United States
| | - Pavana Anur
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, OR, United States
| | - Katie Johnson-Camacho
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, OR, United States
| | - Shaadi Tabatabaei
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, OR, United States
| | - Daniel J Munson
- Department of Immunology & Microbiology, University of Colorado, Denver, CO, United States
| | - Tullia C Bruno
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, United States
| | - Jill E Slansky
- Department of Immunology & Microbiology, University of Colorado, Denver, CO, United States
| | - John W Kappler
- National Jewish Medical and Research Center, Denver, CO, United States
| | - Naoto Hirano
- Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Sebastian Boegel
- University Medical Center, Johannes Gutenberg-University, Mainz, Germany
| | - Bernard A Fox
- Laboratory of Molecular and Tumor Immunology, Chiles Research Institute Providence PDX Medical Center, Portland, OR, United States
| | - Colt Egelston
- City of Hope National Medical Center, Duarte, CA, United States
| | - Diana L Simons
- City of Hope National Medical Center, Duarte, CA, United States
| | - Grecia Jimenez
- City of Hope National Medical Center, Duarte, CA, United States
| | - Peter P Lee
- City of Hope National Medical Center, Duarte, CA, United States
| | - Joe W Gray
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, United States; Center for Health & Healing, Oregon Health and Science University, Portland, OR, United States
| | - Paul T Spellman
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, OR, United States
| |
Collapse
|
10
|
Frascoli F, Flood E, Kim PS. A model of the effects of cancer cell motility and cellular adhesion properties on tumour-immune dynamics. MATHEMATICAL MEDICINE AND BIOLOGY-A JOURNAL OF THE IMA 2017; 34:215-240. [PMID: 27094601 DOI: 10.1093/imammb/dqw004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Accepted: 01/18/2016] [Indexed: 02/07/2023]
Abstract
We present a three-dimensional model simulating the dynamics of an anti-cancer T-cell response against a small, avascular, early-stage tumour. Interactions at the tumour site are accounted for using an agent-based model (ABM), while immune cell dynamics in the lymph node are modelled as a system of delay differential equations (DDEs). We combine these separate approaches into a two-compartment hybrid ABM-DDE system to capture the T-cell response against the tumour. In the ABM at the tumour site, movement of tumour cells is modelled using effective physical forces with a specific focus on cell-to-cell adhesion properties and varying levels of tumour cell motility, thus taking into account the ability of cancer cells to spread and form clusters. We consider the effectiveness of the immune response over a range of parameters pertaining to tumour cell motility, cell-to-cell adhesion strength and growth rate. We also investigate the dependence of outcomes on the distribution of tumour cells. Low tumour cell motility is generally a good indicator for successful tumour eradication before relapse, while high motility leads, almost invariably, to relapse and tumour escape. In general, the effect of cell-to-cell adhesion on prognosis is dependent on the level of tumour cell motility, with an often unpredictable cross influence between adhesion and motility, which can lead to counterintuitive effects. In terms of overall tumour shape and structure, the spatial distribution of cancer cells in clusters of various sizes has shown to be strongly related to the likelihood of extinction.
Collapse
Affiliation(s)
- Federico Frascoli
- Faculty of Science, Engineering and Technology, Department of Mathematics, Swinburne University of Technology, Melbourne, Victoria, Australia
| | - Emelie Flood
- School of Applied Sciences, RMIT University, Melbourne, Victoria, Australia
| | - Peter S Kim
- School of Mathematics and Statistics, University of Sydney, New South Wales, Australia
| |
Collapse
|
11
|
Zebularine Treatment Induces MAGE-A11 Expression and Improves CTL Cytotoxicity Using a Novel Identified HLA-A2-restricted MAGE-A11 Peptide. J Immunother 2017; 40:211-220. [DOI: 10.1097/cji.0000000000000170] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
|
12
|
Liu L, Sommermeyer D, Cabanov A, Kosasih P, Hill T, Riddell SR. Inclusion of Strep-tag II in design of antigen receptors for T-cell immunotherapy. Nat Biotechnol 2016; 34:430-4. [PMID: 26900664 PMCID: PMC4940167 DOI: 10.1038/nbt.3461] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Accepted: 12/15/2015] [Indexed: 02/06/2023]
Abstract
The tactical introduction of Strep-tag II into synthetic antigen
receptors provides engineered T cells with a marker for identification and rapid
purification, and a functional element for selective antibody coated
microbead-driven large-scale expansion. Such receptor designs can be applied to
chimeric antigen receptors of different ligand specificities and costimulatory
domains, and to T cell receptors to facilitate cGMP manufacturing of adoptive T
cell therapies to treat cancer and other diseases.
Collapse
Affiliation(s)
- Lingfeng Liu
- Program in Immunology, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Daniel Sommermeyer
- Program in Immunology, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Alexandra Cabanov
- Program in Immunology, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Paula Kosasih
- Program in Immunology, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Tyler Hill
- Program in Immunology, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Stanley R Riddell
- Program in Immunology, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA.,Department of Medicine, University of Washington, Seattle, Washington, USA.,Institute for Advanced Study, Technical University of Munich, Munich, Germany
| |
Collapse
|
13
|
T-cell libraries allow simple parallel generation of multiple peptide-specific human T-cell clones. J Immunol Methods 2016; 430:43-50. [PMID: 26826277 PMCID: PMC4783706 DOI: 10.1016/j.jim.2016.01.014] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Revised: 01/26/2016] [Accepted: 01/26/2016] [Indexed: 12/29/2022]
Abstract
Isolation of peptide-specific T-cell clones is highly desirable for determining the role of T-cells in human disease, as well as for the development of therapies and diagnostics. However, generation of monoclonal T-cells with the required specificity is challenging and time-consuming. Here we describe a library-based strategy for the simple parallel detection and isolation of multiple peptide-specific human T-cell clones from CD8(+) or CD4(+) polyclonal T-cell populations. T-cells were first amplified by CD3/CD28 microbeads in a 96U-well library format, prior to screening for desired peptide recognition. T-cells from peptide-reactive wells were then subjected to cytokine-mediated enrichment followed by single-cell cloning, with the entire process from sample to validated clone taking as little as 6 weeks. Overall, T-cell libraries represent an efficient and relatively rapid tool for the generation of peptide-specific T-cell clones, with applications shown here in infectious disease (Epstein-Barr virus, influenza A, and Ebola virus), autoimmunity (type 1 diabetes) and cancer.
Collapse
|
14
|
Zhang J, Zhu L, Du H, He X, Yin Y, Gu Y, Liu L, Lu K, Guo R, Liu P, Shu Y. Autologous cytokine-induced killer cell therapy in lung cancer patients: a retrospective study. Biomed Pharmacother 2014; 70:248-52. [PMID: 25776508 DOI: 10.1016/j.biopha.2014.12.025] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Accepted: 12/11/2014] [Indexed: 12/22/2022] Open
Abstract
Cytokine-induced killer (CIK) cells have the ability to kill tumor cells in vitro and in vivo. This study aimed to evaluate the clinical effect of adjuvant immunotherapy with CIK cells on the prognosis of lung cancer patients. In the present study, we investigated the clinical outcomes of autologous CIK cell immunotherapy for patients with lung cancer in a case-control study. Our study included 60 patients who received chemotherapy combined with autologous CIK cell adoptive immunotherapy in CIK treatment group and 60 patients who received chemotherapy alone in the control group. Progression-free survival (PFS) and overall survival (OS) of these two groups were evaluated. After 14 days of incubation in vitro, the percentages of CD3(+), CD3(+)CD8(+), CD3(+)CD56(+) and CD3(-)CD56(+) were significantly increased (P<0.05). The clinical symptoms of 60 patients were apparently improved. No severe toxicity and side effects were observed in the CIK treatment group. The 3-year, 5-year PFS rates were 44.7% and 26.8% and the 3-year, 5-year OS rates were 74% and 62% in the CIK group, respectively, which were significantly improved compared to that in the control group. The median PFS and OS in the CIK group were significantly improved than those in the control group (PFS, 24 months vs. 14 months, P=0.014; OS, 72 months vs. 44 months, P=0.006). Our results indicated that autologous CIK cells can efficiently improve the immunological status and prolong PFS and OS in patients with lung cancer.
Collapse
Affiliation(s)
- Jinying Zhang
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Lingjun Zhu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Haina Du
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xiang He
- The Billing Management Center of Nanjing Medical Insurance, Nanjing, China
| | - Yongmei Yin
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yanhong Gu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Lianke Liu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Kaihua Lu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Renhua Guo
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Ping Liu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yongqian Shu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
15
|
Peptide-Based Vaccination and Induction of CD8+ T-Cell Responses Against Tumor Antigens in Breast Cancer. BioDrugs 2014; 29:15-30. [DOI: 10.1007/s40259-014-0114-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
16
|
Gardyan A, Osen W, Zörnig I, Podola L, Agarwal M, Aulmann S, Ruggiero E, Schmidt M, Halama N, Leuchs B, von Kalle C, Beckhove P, Schneeweiss A, Jäger D, Eichmüller SB. Identification of NY-BR-1-specific CD4(+) T cell epitopes using HLA-transgenic mice. Int J Cancer 2014; 136:2588-97. [PMID: 25387692 DOI: 10.1002/ijc.29322] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Accepted: 10/21/2014] [Indexed: 12/11/2022]
Abstract
Breast cancer represents the second most common cancer type worldwide and has remained the leading cause of cancer-related deaths among women. The differentiation antigen NY-BR-1 appears overexpressed in invasive mammary carcinomas compared to healthy breast tissue, thus representing a promising target antigen for T cell based tumor immunotherapy approaches. Since efficient immune attack of tumors depends on the activity of tumor antigen-specific CD4(+) effector T cells, NY-BR-1 was screened for the presence of HLA-restricted CD4(+) T cell epitopes that could be included in immunological treatment approaches. Upon NY-BR-1-specific DNA immunization of HLA-transgenic mice and functional ex vivo analysis, a panel of NY-BR-1-derived library peptides was determined that specifically stimulated IFNγ secretion among splenocytes of immunized mice. Following in silico analyses, four candidate epitopes were determined which were successfully used for peptide immunization to establish NY-BR-1-specific, HLA-DRB1*0301- or HLA-DRB1*0401-restricted CD4(+) T cell lines from splenocytes of peptide immunized HLA-transgenic mice. Notably, all four CD4(+) T cell lines recognized human HLA-DR-matched dendritic cells (DC) pulsed with lysates of NY-BR-1 expressing human tumor cells, demonstrating natural processing of these epitopes also within the human system. Finally, CD4(+) T cells specific for all four CD4(+) T cell epitopes were detectable among PBMC of breast cancer patients, showing that CD4(+) T cell responses against the new epitopes are not deleted nor inactivated by self-tolerance mechanisms. Our results present the first NY-BR-1-specific HLA-DRB1*0301- and HLA-DRB1*0401-restricted T cell epitopes that could be exploited for therapeutic intervention against breast cancer.
Collapse
Affiliation(s)
- Adriane Gardyan
- Department of Translational Immunology, German Cancer Research Center Heidelberg (DKFZ), Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Stephen TL, Rutkowski MR, Allegrezza MJ, Perales-Puchalt A, Tesone AJ, Svoronos N, Nguyen JM, Sarmin F, Borowsky ME, Tchou J, Conejo-Garcia JR. Transforming growth factor β-mediated suppression of antitumor T cells requires FoxP1 transcription factor expression. Immunity 2014; 41:427-439. [PMID: 25238097 DOI: 10.1016/j.immuni.2014.08.012] [Citation(s) in RCA: 96] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2013] [Accepted: 08/27/2014] [Indexed: 12/15/2022]
Abstract
Tumor-reactive T cells become unresponsive in advanced tumors. Here we have characterized a common mechanism of T cell unresponsiveness in cancer driven by the upregulation of the transcription factor Forkhead box protein P1 (Foxp1), which prevents CD8⁺ T cells from proliferating and upregulating Granzyme-B and interferon-γ in response to tumor antigens. Accordingly, Foxp1-deficient lymphocytes induced rejection of incurable tumors and promoted protection against tumor rechallenge. Mechanistically, Foxp1 interacted with the transcription factors Smad2 and Smad3 in preactivated CD8⁺ T cells in response to microenvironmental transforming growth factor-β (TGF-β), and was essential for its suppressive activity. Therefore, Smad2 and Smad3-mediated c-Myc repression requires Foxp1 expression in T cells. Furthermore, Foxp1 directly mediated TGF-β-induced c-Jun transcriptional repression, which abrogated T cell activity. Our results unveil a fundamental mechanism of T cell unresponsiveness different from anergy or exhaustion, driven by TGF-β signaling on tumor-associated lymphocytes undergoing Foxp1-dependent transcriptional regulation.
Collapse
Affiliation(s)
- Tom L Stephen
- Tumor Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Melanie R Rutkowski
- Tumor Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Michael J Allegrezza
- Tumor Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Alfredo Perales-Puchalt
- Tumor Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Amelia J Tesone
- Tumor Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Nikolaos Svoronos
- Tumor Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Jenny M Nguyen
- Tumor Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Fahmida Sarmin
- Tumor Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Mark E Borowsky
- Helen F. Graham Cancer Center, Christiana Care Health System, 4701 Ogletown-Stanton Road, Newark, DE 19713, USA
| | - Julia Tchou
- Division of Endocrine and Oncologic Surgery, Department of Surgery, University of Pennsylvania, Philadelphia, PA 19104-1693, USA; Rena Rowan Breast Center, University of Pennsylvania, Philadelphia, PA 19104-1693, USA; Abramson Cancer Center Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104-1693, USA
| | - Jose R Conejo-Garcia
- Tumor Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, PA 19104, USA.
| |
Collapse
|
18
|
Zhang J, Zhu L, Zhang Q, He X, Yin Y, Gu Y, Guo R, Lu K, Liu L, Liu P, Shu Y. Effects of cytokine-induced killer cell treatment in colorectal cancer patients: a retrospective study. Biomed Pharmacother 2014; 68:715-20. [PMID: 25107840 DOI: 10.1016/j.biopha.2014.07.010] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2014] [Accepted: 07/08/2014] [Indexed: 01/26/2023] Open
Abstract
Cytokine-induced killer (CIK) cells are ex vivo generated heterogeneous NK-like T-lymphocytes, which have anti-tumor effects in vitro and in vivo. This present study was conducted to evaluate the effects of autologous CIK cell immunotherapy on the prognosis of colorectal cancer patients. Progression-free survival (PFS), overall survival (OS) and immune cells were assessed. We found that the percentages of CD8(+), CD3(+) CD56(+), CD3(-) CD56(+) cell subsets were significantly increased from 19.7±6.3%, 13.8±7.9%, 1.0±1.2% to 35.8±11.6% (P<0.001), 20.9±12.5 (P<0.001), 14.4±9.5% (P<0.001), respectively in the CIK group after 14 days of incubation. The median PFS and median OS in the CIK group were 25.8 months and 41.3 months respectively, while 12.0 months and 30.8 months in the control group. The PFS and OS curves of the CIK group and control group indicated that there were also statistically differences between two groups in PFS (log-rank, P=0.01) and OS (log-rank, P=0.037). Our results indicate that CIK cell immunotherapy in combination with chemotherapy can reduce the recurrence rate and promote the survival time of patients with colorectal cancer.
Collapse
Affiliation(s)
- Jinying Zhang
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, 300, GuangZhou Road, Nanjing 210029, China
| | - Lingjun Zhu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, 300, GuangZhou Road, Nanjing 210029, China
| | - Qian Zhang
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, 300, GuangZhou Road, Nanjing 210029, China
| | - Xiang He
- Nanjing Medical Insurance Billing Management Center, Nanjing, China
| | - Yongmei Yin
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, 300, GuangZhou Road, Nanjing 210029, China
| | - Yanhong Gu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, 300, GuangZhou Road, Nanjing 210029, China
| | - Renhua Guo
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, 300, GuangZhou Road, Nanjing 210029, China
| | - Kaihua Lu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, 300, GuangZhou Road, Nanjing 210029, China
| | - Lianke Liu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, 300, GuangZhou Road, Nanjing 210029, China
| | - Ping Liu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, 300, GuangZhou Road, Nanjing 210029, China
| | - Yongqian Shu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, 300, GuangZhou Road, Nanjing 210029, China.
| |
Collapse
|
19
|
Frascoli F, Kim PS, Hughes BD, Landman KA. A dynamical model of tumour immunotherapy. Math Biosci 2014; 253:50-62. [DOI: 10.1016/j.mbs.2014.04.003] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2013] [Revised: 04/08/2014] [Accepted: 04/10/2014] [Indexed: 12/26/2022]
|
20
|
Jiang X, Shapiro DJ. The immune system and inflammation in breast cancer. Mol Cell Endocrinol 2014; 382:673-682. [PMID: 23791814 PMCID: PMC4919022 DOI: 10.1016/j.mce.2013.06.003] [Citation(s) in RCA: 168] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2013] [Revised: 06/03/2013] [Accepted: 06/04/2013] [Indexed: 02/07/2023]
Abstract
During different stages of tumor development the immune system can either identify and destroy tumors, or promote their growth. Therapies targeting the immune system have emerged as a promising treatment modality for breast cancer, and immunotherapeutic strategies are being examined in preclinical and clinical models. However, our understanding of the complex interplay between cells of the immune system and breast cancer cells is incomplete. In this article, we review recent findings showing how the immune system plays dual host-protective and tumor-promoting roles in breast cancer initiation and progression. We then discuss estrogen receptor α (ERα)-dependent and ERα-independent mechanisms that shield breast cancers from immunosurveillance and enable breast cancer cells to evade immune cell induced apoptosis and produce an immunosuppressive tumor microenvironment. Finally, we discuss protumorigenic inflammation that is induced during tumor progression and therapy, and how inflammation promotes more aggressive phenotypes in ERα positive breast cancers.
Collapse
Affiliation(s)
- Xinguo Jiang
- Department of Medicine, VA Palo Alto Health Care System/Stanford University School of Medicine, Stanford, CA 94305, USA.
| | - David J Shapiro
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA.
| |
Collapse
|
21
|
Shi G, Zhou C, Wang D, Ma W, Zhang S. LAPTM4B-35 protein is a weak tumor-associated antigen candidate. Exp Ther Med 2014; 7:491-495. [PMID: 24396432 PMCID: PMC3881050 DOI: 10.3892/etm.2013.1427] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2013] [Accepted: 11/13/2013] [Indexed: 11/19/2022] Open
Abstract
Lysosome-associated protein transmembrane 4β (LAPTM4B) is a gene that has been indicated to be involved in cancer. It is located at chromosome 8q22 and is composed of seven exons and six introns. LAPTM4B encodes two protein isoforms: LAPTM4B-35 and LAPTM4B-24. LAPTM4B-35 is markedly upregulated and LAPTM4B-24 is downregulated in several types of cancer. LAPTM4B-35 is 91 amino acids (N91) longer than LAPTM4B-24 at the N-terminus. In the present study, western blotting, enzyme-linked immunosorbent spot analysis and the B16F10-N91 tumor bearing-mice experiments were used to evaluate whether the overexpression of N91 indicates its potential as a candidate tumor-associated antigen. The results revealed that N91 was expressed in a wide range of normal mouse tissues and human peripheral blood mononuclear cells, with varying expression levels. The weak immunogenicity of N91 protein suggested it was a weak candidate antigen; however, the N91 protein was associated with cell proliferation.
Collapse
Affiliation(s)
- Guilan Shi
- Department of Immunology, Zibo Vocational Institute, Zibo, Shandong 255314, P.R. China ; Department of Immunology, Cancer Institute, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100021, P.R. China
| | - Chunxia Zhou
- Department of Immunology, Cancer Institute, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100021, P.R. China
| | - Dongmei Wang
- Department of Immunology, Cancer Institute, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100021, P.R. China
| | - Wenbo Ma
- Department of Immunology, Cancer Institute, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100021, P.R. China
| | - Shuren Zhang
- Department of Immunology, Cancer Institute, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100021, P.R. China
| |
Collapse
|
22
|
Autologous CIK cell immunotherapy in patients with renal cell carcinoma after radical nephrectomy. Clin Dev Immunol 2013; 2013:195691. [PMID: 24382970 PMCID: PMC3872096 DOI: 10.1155/2013/195691] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2013] [Accepted: 11/13/2013] [Indexed: 11/17/2022]
Abstract
Objective. To evaluate the efficacy of autologous cytokine-induced killer (CIK) cells in patients with renal cell carcinoma (RCC). Methods. 20 patients diagnosed with TNM stage I or II RCC were randomly divided into two groups, a CIK cell treatment group and a control group. The endpoint was progression-free survival (PFS) evaluated by Kaplan-Meier analyses. Results. CD3+, CD3+/CD8+, CD3+/CD4+, and CD3+/CD56+ levels increased after CIK cell culture (P < 0.01). The median PFS in CIK cell treatment group was significantly longer than that in control group (PFS, 32.2 months versus 21.6 months; log-rank, P = 0.032), all patients were alive during the course of followup, and there are no statistically significant differences between two groups in OS (log-rank, P = 0.214). Grade III or greater adverse events were not observed. Conclusions. CIK cells treatment could prolong survival in patients with RCC after radical nephrectomy and showed acceptable curative effect with potential enhancement of cellular immune function. This trial is registered with Clinicaltrials.gov NCT01799083.
Collapse
|
23
|
Behnke NK, Alamanda VK, Song Y, Archer KR, Halpern JL, Schwartz HS, Holt GE. Does postoperative infection after soft tissue sarcoma resection affect oncologic outcomes? J Surg Oncol 2013; 109:415-20. [PMID: 24284805 DOI: 10.1002/jso.23518] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2013] [Accepted: 11/09/2013] [Indexed: 12/25/2022]
Abstract
BACKGROUND AND OBJECTIVES Prior studies have demonstrated postoperative infection may confer a survival benefit after osteosarcoma resection. Our aim was to determine whether infection after soft tissue sarcoma resection has similar effects on metastasis, recurrence and survival. METHODS A retrospective review was conducted; 396 patients treated surgically for a soft tissue sarcoma between 2000 and 2008 were identified. Relevant oncologic data were collected. Fifty-six patients with a postoperative infection were compared with 340 patients without infection. Hazard ratios and overall cumulative risk were evaluated. RESULTS There was no difference in survival, local recurrence or metastasis between patients with or without a postoperative infection. Patients were evenly matched for age at diagnosis, gender, smoking status, and diabetes status. Tumor characteristics did not differ between groups in tumor size, location, depth, grade, margin status, stage, and histologic subtype. There was no difference in utilization of chemotherapy or radiation therapy between groups. From our competing risk model, only positive margin status significantly impacted the risk of local recurrence. An increase in tumor size corresponded to an increased risk of metastasis and death. CONCLUSIONS Postoperative infection neither conferred a protective effect, nor increased the risk of adverse oncologic outcomes after soft tissue sarcoma resection.
Collapse
Affiliation(s)
- Nicole K Behnke
- Division of Orthopaedic Surgery, University of Alabama at Birmingham Medical Center, Birmingham, Alabama
| | | | | | | | | | | | | |
Collapse
|
24
|
Kim JS, Park YS, Kim JY, Kim YG, Kim YJ, Lee HK, Kim HS, Hong JT, Kim Y, Han SB. Inhibition of human pancreatic tumor growth by cytokine-induced killer cells in nude mouse xenograft model. Immune Netw 2012; 12:247-52. [PMID: 23396819 PMCID: PMC3566419 DOI: 10.4110/in.2012.12.6.247] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2012] [Revised: 10/29/2012] [Accepted: 11/02/2012] [Indexed: 01/01/2023] Open
Abstract
Pancreatic cancer is the fourth commonest cause of cancer-related deaths in the world. However, no adequate therapy for pancreatic cancer has yet been found. In this study, the antitumor activity of cytokine-induced killer (CIK) cells against the human pancreatic cancer was evaluated in vitro and in vivo. Human peripheral blood mononuclear cells were cultured with IL-2-containing medium in anti-CD3 for 14 days. The resulting populations of CIK cells comprised 94% CD3+, 4% CD3-CD56+, 41% CD3+CD56+, 11% CD4+, and 73% CD8+. This heterogeneous cell population was called cytokine-induced killer (CIK) cells. At an effector-target cell ratio of 100:1, CIK cells destroyed 51% of AsPC-1 human pancreatic cancer cells, as measured by the 51Cr-release assay. In addition, CIK cells at doses of 3 and 10 million cells per mouse inhibited 42% and 70% of AsPC-1 tumor growth in nude mouse xenograft assays, respectively. This study suggests that CIK cells may be used as an adoptive immunotherapy for pancreatic cancer patients.
Collapse
Affiliation(s)
- Ji Sung Kim
- College of Pharmacy, Chungbuk National University, Cheongju 361-763, Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Kim PS, Lee PP. Modeling protective anti-tumor immunity via preventative cancer vaccines using a hybrid agent-based and delay differential equation approach. PLoS Comput Biol 2012; 8:e1002742. [PMID: 23133347 PMCID: PMC3486888 DOI: 10.1371/journal.pcbi.1002742] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2012] [Accepted: 08/31/2012] [Indexed: 12/20/2022] Open
Abstract
A next generation approach to cancer envisions developing preventative vaccinations to stimulate a person's immune cells, particularly cytotoxic T lymphocytes (CTLs), to eliminate incipient tumors before clinical detection. The purpose of our study is to quantitatively assess whether such an approach would be feasible, and if so, how many anti-cancer CTLs would have to be primed against tumor antigen to provide significant protection. To understand the relevant dynamics, we develop a two-compartment model of tumor-immune interactions at the tumor site and the draining lymph node. We model interactions at the tumor site using an agent-based model (ABM) and dynamics in the lymph node using a system of delay differential equations (DDEs). We combine the models into a hybrid ABM-DDE system and investigate dynamics over a wide range of parameters, including cell proliferation rates, tumor antigenicity, CTL recruitment times, and initial memory CTL populations. Our results indicate that an anti-cancer memory CTL pool of 3% or less can successfully eradicate a tumor population over a wide range of model parameters, implying that a vaccination approach is feasible. In addition, sensitivity analysis of our model reveals conditions that will result in rapid tumor destruction, oscillation, and polynomial rather than exponential decline in the tumor population due to tumor geometry. An innovative approach to treating cancer envisions developing preventative anti-cancer vaccines to train a person's immune cells to eliminate early-stage tumors close to genesis. The design of such a treatment strategy requires an understanding of the tumor and immune interactions leading to a successful anti-cancer immune response. To engage this problem, we formulate a mathematical model of the immune response against incipient tumours consisting of as low as hundreds to thousands of cancer cells, which is far below the clinical detection threshold of over 100,000 cells. The model considers the initial stimulation of the immune response and the resulting immune attack on the tumor mass and is formulated as a hybrid agent-based and delay differential equation model. We apply the model to test dynamics over a wide range of dynamic parameters, including immune and tumor cell growth rates and the size of the initial anti-cancer immune population. Our results show that an anti-cancer memory immune cell population of 3% or less can successfully eradicate an incipient tumor population over a wide range of dynamic parameters, indicating that a vaccination approach is feasible.
Collapse
Affiliation(s)
- Peter S. Kim
- School of Mathematics and Statistics, University of Sydney, Sydney, New South Wales, Australia
| | - Peter P. Lee
- Cancer Immunotherapeutics and Tumor Immunology, City of Hope and Beckman Research Institute, Duarte, California, United States of America
- * E-mail:
| |
Collapse
|
26
|
Zhang J, Zhu L, Wei J, Liu L, Yin Y, Gu Y, Shu Y. The effects of cytokine-induced killer cells for the treatment of patients with solid tumors: a clinical retrospective study. J Cancer Res Clin Oncol 2012; 138:1057-62. [PMID: 22392076 DOI: 10.1007/s00432-012-1179-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2012] [Accepted: 02/14/2012] [Indexed: 12/18/2022]
Abstract
BACKGROUND Cytokine-induced killer (CIK) cells exert high impact on adoptive immunotherapeutic approaches for malignant tumors. This study aimed to evaluate the effect of adjuvant immunotherapy with CIK cells on the prognosis of solid tumor. METHODS Peripheral blood mononuclear cells were collected by a blood cell separator from 40 patients, then expanded by priming them with interferon-gamma followed by monoclonal antibody against CD3 and interleukin-2 the next day. The phenotypic patterns of CIK cells were characterized by flow cytometry on days 0, 7, 10, 14 and 21 of incubation, respectively. Then, 5 ml of venous blood was obtained from 40 patients before and after CIK cells were transfused into patients to assess the influence of CIK cells on the percentages of effector cells. RESULTS After 14 days of incubation in vitro, the percentages of CD3(+), CD8(+), CD3(+) and CD56(+) increased significantly (P < 0.05). The clinical symptoms of 40 patients were improved apparently. The 6-month, 1-year and 3-year overall survival rates were 70.0, 60.0 and 57.5%, respectively. CONCLUSIONS Our results indicated that CIKs immunotherapy can be an effective adjuvant instrument of the routine therapy of malignancy.
Collapse
Affiliation(s)
- Jinying Zhang
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Rd, Nanjing 210029, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
27
|
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: 2.1] [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
|
28
|
Su X, Zhang L, Jin L, Ye J, Guan Z, Chen R, Guo T. Immunotherapy with cytokine-induced killer cells in metastatic renal cell carcinoma. Cancer Biother Radiopharm 2011; 25:465-70. [PMID: 20701541 DOI: 10.1089/cbr.2010.0762] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Cytokine-induced killer (CIK) cells have shown antitumor activity against several tumor cells both in vitro and in vivo. This study reports on the large-scale expansion of CIK cells and also present preliminary results from a pilot clinical trial. Sixteen (16) patients with renal cell carcinoma (RCC), all of whom had metastases after radical nephrectomy and adjuvant therapy using interferon-alpha (IFN-alpha) and/or interleukin-2 (IL-2), were treated with CIK cells. CIK cells were generated from peripheral blood mononuclear cells (PBMCs) and incubated in the presence of IFN-gamma followed by OKT3 and IL-2. Treatment schedule consisted of two to three cycles of CIK cell infusions at an interval of 3 weeks. A total of 46 infusions were administered to 16 metastatic RCC (mRCC) patients. The median number of transferred cells per treatment was 6.7 x 10(9) (range, 2.5-12.3). At a 60:1 effector-target cell ratio, CIK cells killed 51.4% and 32.1% of two human kidney tumor cell lines (293 and SK-RC-42), respectively. After CIK cell infusion, the percentage of CD3(+), CD8(+), CD3(+)CD56(+), and NKG2D(+) cells and the intracellular products of two type 1 cytokines (IFN-gamma and tumor necrosis factor alpha) significantly increased in the patients' PBMCs. Toxicity was minimal, and there were no immediate adverse reactions to the infusions. Three (3) patients had complete response, 1 patient had partial response, and 6 patients had stable disease. These results showed that adoptive CIK cell immunotherapy is a safe and effective treatment, which may have essential benefits for the improvement of the immunologic function in mRCC patients and play an important role in the treatment of mRCC.
Collapse
Affiliation(s)
- Xiaosan Su
- Biomedical Research Center, The First Hospital of Kunming, People's Republic of China.
| | | | | | | | | | | | | |
Collapse
|
29
|
Kim YJ, Lim J, Kang JS, Kim HM, Lee HK, Ryu HS, Kim JY, Hong JT, Kim Y, Han SB. Adoptive immunotherapy of human gastric cancer with ex vivo expanded T cells. Arch Pharm Res 2010; 33:1789-95. [PMID: 21116782 DOI: 10.1007/s12272-010-1111-7] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2010] [Accepted: 07/09/2010] [Indexed: 01/09/2023]
Abstract
Surgical resection of gastric cancer has made significant progress, but majority of patients with advanced gastric cancer face relapse and die within five years. In this study, the antitumor activity of ex vivo expanded T cells against the human gastric cancer was evaluated in vitro and in vivo. Human peripheral blood mononuclear cells were cultured with IL-2-containing medium in anti-CD3 antibody-coated flasks for 5 days, followed by incubation in IL-2-containing medium for 9 days. The resulting populations were mostly CD3(+) T cells (97%) and comprised 1% CD3⁻CD56(+), 36% CD3(+)CD56(+), 11% CD4(+), and 80% CD8(+). This heterogeneous cell population was also called cytokine-induced killer (CIK) cells. CIK cells strongly produced IFN-γ, moderately TNF-α, but not IL-2 and IL-4. At an effector-target cell ratio of 30:1, CIK cells destroyed 58% of MKN74 human gastric cancer cells, as measured by the ⁵¹Cr-release assay. In addition, CIK cells at doses of 3 and 10 million cells per mouse inhibited 58% and 78% of MKN74 tumor growth in nude mouse xenograft assays, respectively. This study suggests that CIK cells may be used as an adoptive immunotherapy for gastric cancer patients.
Collapse
Affiliation(s)
- Yeon Jin Kim
- Medical Research Center, Chungbuk National University, Cheongju, Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Kim HM, Kang JS, Lim J, Kim JY, Kim YJ, Lee SJ, Song S, Hong JT, Kim Y, Han SB. Antitumor activity of cytokine-induced killer cells in nude mouse xenograft model. Arch Pharm Res 2009; 32:781-7. [PMID: 19471894 DOI: 10.1007/s12272-009-1518-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2009] [Revised: 05/11/2009] [Accepted: 05/11/2009] [Indexed: 12/31/2022]
Abstract
Malignant glioma is the most common primary brain tumor in adults and the median survival for patients is less than a year. Despite aggressive treatments including surgical resection, radiotherapy, and chemotherapy, only modest improvement has been achieved in the survival of patients with glioma. In this study, the antitumor activity of cytokine-induced killer (CIK) cells against human glioma cancer was evaluated in vitro and in vivo. Human peripheral blood mononuclear cells were cultured with IL-2-containing medium in anti-CD3 antibody-coated flasks for 5 days, followed by incubation in IL-2-containing medium for 9 days. The number of cells increased more than 200-fold and the viability was >90%. The resulting populations were consisted of 96% CD3(+), 2% CD3(-)CD56(+), 68% CD3(+)CD56(+), 2% CD4(+), <1% CD4(+)CD56(+), 80% CD8(+), and 49% CD8(+)CD56(+). This heterogeneous cell population was called as CIK cells. At an effector-target cell ratio of 30:1, CIK cells destroyed 43% of U-87 MG human glioma cells, as measured by the (51)Cr-release assay. In addition, CIK cells at doses of 0.3, 1, and 3 million cells per mouse inhibited 23%, 40%, and 50% of U-87 MG tumor growth in nude mouse xenograft assays, respectively. This study suggests that CIK cells may be used as an adoptive immunotherapy for glioma cancer patients.
Collapse
Affiliation(s)
- Hwan Mook Kim
- Korea Research Institute of Bioscience and Biotechnology, Ochang, Chungbuk, 363-883, Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Kim HM, Lim J, Kang JS, Park SK, Lee K, Kim JY, Kim YJ, Hong JT, Kim Y, Han SB. Inhibition of human cervical carcinoma growth by cytokine-induced killer cells in nude mouse xenograft model. Int Immunopharmacol 2009; 9:375-80. [DOI: 10.1016/j.intimp.2008.12.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2008] [Revised: 12/03/2008] [Accepted: 12/03/2008] [Indexed: 01/31/2023]
|
32
|
Immunity to Growth Factor Receptor–Bound Protein 10, a Signal Transduction Molecule, Inhibits the Growth of Breast Cancer in Mice. Cancer Res 2008; 68:2463-70. [DOI: 10.1158/0008-5472.can-07-5685] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
33
|
Antitumor activity of cytokine-induced killer cells against human lung cancer. Int Immunopharmacol 2007; 7:1802-7. [DOI: 10.1016/j.intimp.2007.08.016] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2007] [Revised: 08/16/2007] [Accepted: 08/16/2007] [Indexed: 11/22/2022]
|
34
|
Theurillat JP, Zürrer-Härdi U, Varga Z, Barghorn A, Saller E, Frei C, Storz M, Behnke S, Seifert B, Fehr M, Fink D, Rageth C, Linsenmeier C, Pestalozzi B, Chen YT, Knuth A, Jäger D, Moch H. Distinct expression patterns of the immunogenic differentiation antigen NY-BR-1 in normal breast, testis and their malignant counterparts. Int J Cancer 2007; 122:1585-91. [DOI: 10.1002/ijc.23241] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
35
|
Inhibition of human ovarian tumor growth by cytokine-induced killer cells. Arch Pharm Res 2007; 30:1464-70. [DOI: 10.1007/bf02977372] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
36
|
Theurillat JP, Zürrer-Härdi U, Varga Z, Storz M, Probst-Hensch NM, Seifert B, Fehr MK, Fink D, Ferrone S, Pestalozzi B, Jungbluth AA, Chen YT, Jäger D, Knuth A, Moch H. NY-BR-1 protein expression in breast carcinoma: a mammary gland differentiation antigen as target for cancer immunotherapy. Cancer Immunol Immunother 2007; 56:1723-31. [PMID: 17410359 PMCID: PMC11030863 DOI: 10.1007/s00262-007-0316-1] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2006] [Accepted: 03/10/2007] [Indexed: 10/23/2022]
Abstract
NY-BR-1 is a recently identified differentiation antigen of the mammary gland. To use NY-BR-1 for T-cell-based immunotherapy, analysis of its co-expression with HLA class I antigens is required. In the present tissue microarray study, primary breast cancers (n = 1,444), recurrences (n = 88), lymph node (n = 525) and distant metastases (n = 91) were studied for NY-BR-1 expression using a novel monoclonal antibody. NY-BR-1 expression was compared with prognosis, estrogen receptor, HER2-status, EGFR and HLA class I antigen expression. NY-BR-1 was more frequently expressed in grade 1 (82%) than in grade 2 (69%) and grade 3 (46%) carcinomas (P < 0.0001). Moreover, NY-BR-1 expression correlated directly with estrogen receptor expression (P < 0.0001) and inversely correlated with HER2-status and EGFR expression (P < 0.0001 for both). Considering high expression level of co-expression, 198/1,321 (15%) primary breast carcinomas and 4/65 (6%) distant metastases expressed NY-BR-1 and HLA class I, suggesting that active immunotherapy can be applied to about 10% of breast cancer patients. Survival analysis showed an association of NY-BR-1 expression with better patient outcome (P = 0.015). No difference between NY-BR-1 expression of primary tumors and metastases could be found, indicating that the presence of NY-BR-1 in metastases can be deduced from their corresponding primary. Forty-three paired biopsies taken from patients before and after chemotherapy suggest that NY-BR-1 expression is not influenced by preceding chemotherapy (kappa = 0.89, P < 0.0001). In summary, the co-expression of NY-BR-1 with HLA class I antigens and its expression in metastases without modification by chemotherapy suggest that NY-BR-1 targeted immunotherapy represents a viable strategy in addition to other targeted cancer drug therapies of breast cancer.
Collapse
Affiliation(s)
- Jean-Philippe Theurillat
- Institute for Surgical Pathology, Department of Pathology, University Hospital Zurich, Schmelzbergstrasse 12, 8091, Zurich, Switzerland.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Chu Y, Yang X, Xu W, Wang Y, Guo Q, Xiong S. In situ expression of IFN-gamma-inducible T cell alpha chemoattractant in breast cancer mounts an enhanced specific anti-tumor immunity which leads to tumor regression. Cancer Immunol Immunother 2007; 56:1539-49. [PMID: 17659370 PMCID: PMC11030116 DOI: 10.1007/s00262-007-0296-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2006] [Accepted: 01/23/2007] [Indexed: 10/23/2022]
Abstract
Increased evidence indicates that chemokines are involved in tumor growth. ITAC, a key member of chemokines, possesses the ability to recruit T cells and enhance immune responses. Therefore, ITAC might contribute to antitumor immunity. In this study, we evaluated the relationship between the expression of ITAC and human breast cancer advancement. We further investigated whether forced expression of ITAC in tumor sites could mediate enhanced antitumor immunity in a murine breast cancer model. Results showed that ITAC expression level was down-regulated in 31 breast cancer specimens compared to normal mammary tissues, and associated negatively with the stages of breast cancer. Contrarily, forced expression of ITAC in murine 4T1 tumor cells resulted in tumor regression after initial growth upon injection into naïve Balb/c mice. More lymphocytes were recruited to the site of tumor inoculated by 4T1-ITAC and more than 80% of these T cells expressed the ITAC receptor, CXCR3. ITAC-recruited TILs exhibited 4T1-specific proliferation and cytotoxicity, and an increased IFN-gamma but decreased IL-4 production. Importantly, forced expression of ITAC in 4T1 tumor nodules inhibited tumor growth. These findings demonstrated that the decreased expression of ITAC is associated with the advancement of breast cancer in patients. Forced expression of ITAC in tumor site not only induces increased T cell-recruitment and elicits a specific antitumor immunity, but also mediates regression of established 4T1 tumors, indicating the potential application of ITAC-expressing tumor cells in cancer immunotherapy and vaccine designing.
Collapse
Affiliation(s)
- Yiwei Chu
- Department of Immunology of Shanghai Medical College and Institute for Immunobiology, Fudan University, 138 Yixueyuan Road, Shanghai, 200032 Peoples Republic of China
- Key Laboratory of Molecular Medicine of Ministry of Education, Fudan University, Shanghai, 200032 Peoples Republic of China
| | - Xiuli Yang
- Department of Immunology of Shanghai Medical College and Institute for Immunobiology, Fudan University, 138 Yixueyuan Road, Shanghai, 200032 Peoples Republic of China
- Key Laboratory of Molecular Medicine of Ministry of Education, Fudan University, Shanghai, 200032 Peoples Republic of China
| | - Wei Xu
- Department of Immunology of Shanghai Medical College and Institute for Immunobiology, Fudan University, 138 Yixueyuan Road, Shanghai, 200032 Peoples Republic of China
- Key Laboratory of Molecular Medicine of Ministry of Education, Fudan University, Shanghai, 200032 Peoples Republic of China
| | - Ying Wang
- Department of Immunology of Shanghai Medical College and Institute for Immunobiology, Fudan University, 138 Yixueyuan Road, Shanghai, 200032 Peoples Republic of China
- Key Laboratory of Molecular Medicine of Ministry of Education, Fudan University, Shanghai, 200032 Peoples Republic of China
| | - Qiang Guo
- Department of Immunology of Shanghai Medical College and Institute for Immunobiology, Fudan University, 138 Yixueyuan Road, Shanghai, 200032 Peoples Republic of China
- Key Laboratory of Molecular Medicine of Ministry of Education, Fudan University, Shanghai, 200032 Peoples Republic of China
| | - Sidong Xiong
- Department of Immunology of Shanghai Medical College and Institute for Immunobiology, Fudan University, 138 Yixueyuan Road, Shanghai, 200032 Peoples Republic of China
- Key Laboratory of Molecular Medicine of Ministry of Education, Fudan University, Shanghai, 200032 Peoples Republic of China
- Immunology Division, E-Institutes of Shanghai Universities, Shanghai, 200025 Peoples Republic of China
| |
Collapse
|
38
|
Anti-tumor activity of ex vivo expanded cytokine-induced killer cells against human hepatocellular carcinoma. Int Immunopharmacol 2007; 7:1793-801. [PMID: 17996690 DOI: 10.1016/j.intimp.2007.08.007] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2007] [Revised: 07/13/2007] [Accepted: 08/09/2007] [Indexed: 11/22/2022]
Abstract
Cytokine-induced killer (CIK) cells are ex vivo expanded T cells with natural killer cell phenotypes and functions. In this study, the anti-tumor activity of CIK cells against hepatocellular carcinoma was evaluated in vitro and in vivo. In the presence of anti-CD3 antibody and IL-2 for 14 days, human peripheral blood mononuclear cell population changed to heterogeneous CIK cell population, which comprised 96% CD3(+), 3% CD3( inverted exclamation mark(c))CD56(+), 32% CD3(+)CD56(+), 11% CD4(+), 75% CD8(+), and 30% CD8(+)CD56(+). CIK cells produced significant amounts of IFN-gamma and TNF-alpha; however, produced only slight amounts of IL-2, IL-4, and IL-5. At an effector-target cell ratio of 30:1, CIK cells destroyed 33% of SNU-354 human hepatocellular carcinoma cells, which was determined by the (51)Cr-release assay. In addition, a dose of 1x10(6) CIK cells per mouse inhibited 60% of SNU-354 tumor growth in irradiated nude mice. This study suggests that CIK cells may be used as an adoptive immunotherapy for patients with hepatocellular carcinoma.
Collapse
|
39
|
McMahan RH, Slansky JE. Mobilizing the low-avidity T cell repertoire to kill tumors. Semin Cancer Biol 2007; 17:317-29. [PMID: 17651986 PMCID: PMC2040124 DOI: 10.1016/j.semcancer.2007.06.006] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2007] [Revised: 03/08/2007] [Accepted: 06/07/2007] [Indexed: 01/09/2023]
Abstract
Optimally, T cells destroy infected and transformed cells of the host. To be effective the T cell repertoire must have a sufficiently diverse number of T cell receptors (TCRs) to recognize the abundance of foreign and tumor antigens presented by MHC molecules. The T cell repertoire must also not be reactive toward self-antigens on healthy cells to prevent autoimmunity. Unlike antigens derived from pathogens, most tumor-associated antigens (TAA) are also self-antigens. Therefore, central and peripheral tolerance mechanisms delete or inhibit tumor-reactive T cells. Although there are T cells within the peripheral repertoire that recognize TAA, these T cells are not sufficient to prevent growth of clinically relevant tumors. We will discuss how this dysfunction results, in part, from the low functional avidity of T cells for tumor, or antigen presenting cells (APC) displaying TAA. We discuss the limitations of these low-avidity tumor-reactive T cells and review current immunotherapies aimed at enhancing the avidity and antitumor activity of the tumor-specific T cell repertoire.
Collapse
Affiliation(s)
- Rachel H. McMahan
- Integrated Department of Immunology, University of Colorado at Denver and Health Sciences Center, Denver, CO 80206, USA
| | - Jill E. Slansky
- Integrated Department of Immunology, University of Colorado at Denver and Health Sciences Center, Denver, CO 80206, USA
| |
Collapse
|
40
|
Chopra A, O-Sullivan I, Carr J, Kim TS, Cohen EP. T-regulatory cells are relatively deficient in squamous carcinomas undergoing regression in mice immunized with a squamous carcinoma vaccine enriched for immunotherapeutic cells. Cancer Gene Ther 2007; 14:573-82. [PMID: 17384578 DOI: 10.1038/sj.cgt.7701040] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
In a prior report (Int J Cancer 2006; 119: 339-348), we described a new vaccination strategy for squamous cell carcinoma (SCC). The vaccine was prepared by transfer of unfractionated DNA-fragments (25 kb) from KLN205 cells, a squamous carcinoma cell line (DBA/2 origin; H-2(d)) into LM cells, a highly immunogenic mouse fibroblast cell line (C3H/He origin; (H-2(k))). As only a small proportion of the transfected cell population was expected to have incorporated DNA segments that included genes specifying antigens associated with the squamous carcinoma cells, we devised a novel strategy to enrich the vaccine for immunotherapeutic cells. Enhanced immunity to squamous carcinoma was induced in tumor-bearing mice treated solely by immunization with the enriched vaccine, which translated into prolonged survival without toxicity. Here, we describe the characteristics of the cell populations infiltrating established squamous carcinomas undergoing regression in mice immunized with vaccines enriched for immunotherapeutic cells. The results indicated that CD8+ T cells were predominant and that T-regulatory cells (FoxP3+, CD4/CD25+, CD4/CD62L(high), CD4/CTLA-4e) were relatively deficient in the regressing tumors. Inflammatory infiltrates were not detected in various organs and tissues of mice immunized with the DNA-based vaccine.
Collapse
Affiliation(s)
- A Chopra
- Department of Microbiology and Immunology, University of Illinois College of Medicine, Chicago, IL 60612, USA
| | | | | | | | | |
Collapse
|