51
|
Tumor-expressed immune checkpoint B7x promotes cancer progression and antigen-specific CD8 T cell exhaustion and suppressive innate immune cells. Oncotarget 2017; 8:82740-82753. [PMID: 29137299 PMCID: PMC5669925 DOI: 10.18632/oncotarget.21098] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 08/29/2017] [Indexed: 02/04/2023] Open
Abstract
B7x (B7-H4 or B7S1) is a coinhibitory member of the B7 immune checkpoint ligand family that regulates immune function following ligation with its unknown cognate receptors. B7x has limited expression on normal tissues, but is up-regulated on solid human tumors to inhibit anti-tumor immunity and associates with poor clinical prognosis. We assessed the contribution of cytokine stimuli to induce surface B7x expression on cancer cells and the role of tumor-expressed B7x in a murine pulmonary metastasis model, and finally evaluated the potential interaction between B7x and Neuropilin-1, a suggested potential cognate receptor. We showed that pro-inflammatory and anti-inflammatory cytokines IFNγ, TNFα, and IL-10 did not induce expression of B7x on human or murine cancer cells. Following i.v. injection of CT26, a murine colon cancer cell line in the BALB/c background, we observed a significant increase in tumor burden in the lung of B7x-expressing CT26 mice compared to B7x-negative parental CT26 control mice. This was marked by a significant increase in M2 tumor associated macrophages and antigen-specific CD8 T cell exhaustion. Finally, we found through multiple systems that there was no evidence for B7x and Neuropilin-1 direct interaction. Thus, the B7x pathway has an essential role in modulating the innate and adaptive immune cell infiltrate in the tumor microenvironment with its currently unknown cognate receptor(s).
Collapse
|
52
|
Chen C, Qu QX, Xie F, Zhu WD, Zhu YH, Huang JA. Analysis of B7-H4 expression in metastatic pleural adenocarcinoma and therapeutic potential of its antagonists. BMC Cancer 2017; 17:652. [PMID: 28923053 PMCID: PMC5604341 DOI: 10.1186/s12885-017-3615-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Accepted: 08/28/2017] [Indexed: 12/03/2022] Open
Abstract
Background The increasing incidence and poor outcome associated with malignant pleural effusion (MPE) requires finding an effective treatment for this disease. Inhibitory B7-H4 is expressed in many different human cancers but its role in malignant pleural tissue has yet to be established. Methods Here, patients with metastatic pleural adenocarcinoma (MPA) or with early-stage lung adenocarcinoma were clinically and statistically analyzed. Immunohistochemistry and confocal microscopy were used to determinate the expression of B7-H4 in the cancer cells. By using MPE model, we sought to a potential immunotherapy for MPE with anti-B7-H4 mAb. Results When compared to early-stage lung adenocarcinoma, MPA possessed higher level of nuclei membranous B7-H4 and lower cytoplasmic B7-H4 expression. Also, nuclei membranous B7-H4 expression was found to be positively correlated to Ki-67 expression, and indicated a possible poor prognosis of MPA. In mouse MPE model, intra-pleurally injection of anti-B7-H4 mAb effectively suppressed MPE formation. Conclusions Taken together, our data was in support of the significance of B7-H4 expression in MPA, which also suggest it warrants further exploration for potential immunotherapy of MPE.
Collapse
Affiliation(s)
- Cheng Chen
- Respiratory Department, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, China
| | - Qiu-Xia Qu
- Clinical Immunology Laboratory, The First Affiliated Hospital of Soochow University, 788 Renmin Road, Suzhou, 215007, China
| | - Fang Xie
- Pathology Division, Soochow University, 1 Shizi Street, Suzhou, 215006, China
| | - Wei-Dong Zhu
- Pathology Department, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, China
| | - Ye-Han Zhu
- Respiratory Department, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, China.
| | - Jian-An Huang
- Respiratory Department, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, China.
| |
Collapse
|
53
|
Yu X, Qu L, Bigner DD, Chandramohan V. Selection of novel affinity-matured human chondroitin sulfate proteoglycan 4 antibody fragments by yeast display. Protein Eng Des Sel 2017; 30:639-647. [PMID: 28981720 PMCID: PMC5914443 DOI: 10.1093/protein/gzx038] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Revised: 06/15/2017] [Accepted: 07/13/2017] [Indexed: 12/22/2022] Open
Abstract
Chondroitin sulfate proteoglycan 4 (CSPG4) is a promising target for cancer immunotherapy due to its high level of expression in a number of malignant tumors, and its essential role in tumor growth and progression. Clinical application of CSPG4-targeting immunotherapies is hampered by the lack of fully human high-affinity CSPG4 antibodies or antibody fragments. To overcome this limitation, we performed affinity maturation on a novel human CSPG4 single-chain Fv fragment (scFv) using the random mutagenesis approach and screened for improved variants from a yeast display library using a modified whole-cell panning method followed by fluorescence-activated cell sorting. After six rounds of panning and sorting, the top seven mutant scFvs were isolated and their binding affinities were characterized by flow cytometry and surface plasmon resonance. These highly specific, affinity-matured variants displayed nanomolar to picomolar binding affinities to the CSPG4 antigen. While each of the mutants harbored only two to six amino acid substitutions, they represented ~270-3000-fold improvement in affinity compared to the parental clone. Our study has generated affinity-matured scFvs for the development of antibody-based clinical therapeutics targeting CSPG4-expressing tumors.
Collapse
Affiliation(s)
- Xin Yu
- Department of Pathology, Duke University Medical Center, Durham, NC 27710, USA
| | - Liang Qu
- Department of Pathology, Duke University Medical Center, Durham, NC 27710, USA
| | - Darell D Bigner
- Department of Pathology, Duke University Medical Center, Durham, NC 27710, USA
| | | |
Collapse
|
54
|
Adler AS, Mizrahi RA, Spindler MJ, Adams MS, Asensio MA, Edgar RC, Leong J, Leong R, Roalfe L, White R, Goldblatt D, Johnson DS. Rare, high-affinity anti-pathogen antibodies from human repertoires, discovered using microfluidics and molecular genomics. MAbs 2017; 9:1282-1296. [PMID: 28846502 PMCID: PMC5680809 DOI: 10.1080/19420862.2017.1371383] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Affinity-matured, functional anti-pathogen antibodies are present at low frequencies in natural human repertoires. These antibodies are often excellent candidates for therapeutic monoclonal antibodies. However, mining natural human antibody repertoires is a challenge. In this study, we demonstrate a new method that uses microfluidics, yeast display, and deep sequencing to identify 247 natively paired anti-pathogen single-chain variable fragments (scFvs), which were initially as rare as 1 in 100,000 in the human repertoires. Influenza A vaccination increased the frequency of influenza A antigen-binding scFv within the peripheral B cell repertoire from <0.1% in non-vaccinated donors to 0.3-0.4% in vaccinated donors, whereas pneumococcus vaccination did not increase the frequency of antigen-binding scFv. However, the pneumococcus scFv binders from the vaccinated library had higher heavy and light chain Replacement/Silent mutation (R/S) ratios, a measure of affinity maturation, than the pneumococcus binders from the corresponding non-vaccinated library. Thus, pneumococcus vaccination may increase the frequency of affinity-matured antibodies in human repertoires. We synthesized 10 anti-influenza A and nine anti-pneumococcus full-length antibodies that were highly abundant among antigen-binding scFv. All 10 anti-influenza A antibodies bound the appropriate antigen at KD<10 nM and neutralized virus in cellular assays. All nine anti-pneumococcus full-length antibodies bound at least one polysaccharide serotype, and 71% of the anti-pneumococcus antibodies that we tested were functional in cell killing assays. Our approach has future application in a variety of fields, including the development of therapeutic antibodies for emerging viral diseases, autoimmune disorders, and cancer.
Collapse
Affiliation(s)
- Adam S Adler
- a GigaGen Inc. , 407 Cabot Road, South San Francisco , CA , USA
| | - Rena A Mizrahi
- a GigaGen Inc. , 407 Cabot Road, South San Francisco , CA , USA
| | | | - Matthew S Adams
- a GigaGen Inc. , 407 Cabot Road, South San Francisco , CA , USA
| | | | - Robert C Edgar
- a GigaGen Inc. , 407 Cabot Road, South San Francisco , CA , USA
| | - Jackson Leong
- a GigaGen Inc. , 407 Cabot Road, South San Francisco , CA , USA
| | - Renee Leong
- a GigaGen Inc. , 407 Cabot Road, South San Francisco , CA , USA
| | - Lucy Roalfe
- b Immunobiology Section , Great Ormond Street Institute of Child Health, University College London , London , England, United Kingdom
| | - Rebecca White
- b Immunobiology Section , Great Ormond Street Institute of Child Health, University College London , London , England, United Kingdom
| | - David Goldblatt
- b Immunobiology Section , Great Ormond Street Institute of Child Health, University College London , London , England, United Kingdom
| | - David S Johnson
- a GigaGen Inc. , 407 Cabot Road, South San Francisco , CA , USA
| |
Collapse
|
55
|
Adler AS, Mizrahi RA, Spindler MJ, Adams MS, Asensio MA, Edgar RC, Leong J, Leong R, Johnson DS. Rare, high-affinity mouse anti-PD-1 antibodies that function in checkpoint blockade, discovered using microfluidics and molecular genomics. MAbs 2017; 9:1270-1281. [PMID: 28846506 PMCID: PMC5680806 DOI: 10.1080/19420862.2017.1371386] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Conventionally, mouse hybridomas or well-plate screening are used to identify therapeutic monoclonal antibody candidates. In this study, we present an alternative to hybridoma-based discovery that combines microfluidics, yeast single-chain variable fragment (scFv) display, and deep sequencing to rapidly interrogate and screen mouse antibody repertoires. We used our approach on six wild-type mice to identify 269 molecules that bind to programmed cell death protein 1 (PD-1), which were present at an average of 1 in 2,000 in the pre-sort scFv libraries. Two rounds of fluorescence-activated cell sorting (FACS) produced populations of PD-1-binding scFv with a mean enrichment of 800-fold, whereas most scFv present in the pre-sort mouse repertoires were de-enriched. Therefore, our work suggests that most of the antibodies present in the repertoires of immunized mice are not strong binders to PD-1. We observed clusters of related antibody sequences in each mouse following FACS, suggesting evolution of clonal lineages. In the pre-sort repertoires, these putative clonal lineages varied in both the complementary-determining region (CDR)3K and CDR3H, while the FACS-selected PD-1-binding subsets varied primarily in the CDR3H. PD-1 binders were generally not highly diverged from germline, showing 98% identity on average with germline V-genes. Some CDR3 sequences were discovered in more than one animal, even across different mouse strains, suggesting convergent evolution. We synthesized 17 of the anti-PD-1 binders as full-length monoclonal antibodies. All 17 full-length antibodies bound recombinant PD-1 with KD < 500 nM (average = 62 nM). Fifteen of the 17 full-length antibodies specifically bound surface-expressed PD-1 in a FACS assay, and nine of the antibodies functioned as checkpoint inhibitors in a cellular assay. We conclude that our method is a viable alternative to hybridomas, with key advantages in comprehensiveness and turnaround time.
Collapse
Affiliation(s)
- Adam S Adler
- a GigaGen Inc. , 407 Cabot Road, South San Francisco , CA , USA
| | - Rena A Mizrahi
- a GigaGen Inc. , 407 Cabot Road, South San Francisco , CA , USA
| | | | - Matthew S Adams
- a GigaGen Inc. , 407 Cabot Road, South San Francisco , CA , USA
| | | | - Robert C Edgar
- a GigaGen Inc. , 407 Cabot Road, South San Francisco , CA , USA
| | - Jackson Leong
- a GigaGen Inc. , 407 Cabot Road, South San Francisco , CA , USA
| | - Renee Leong
- a GigaGen Inc. , 407 Cabot Road, South San Francisco , CA , USA
| | - David S Johnson
- a GigaGen Inc. , 407 Cabot Road, South San Francisco , CA , USA
| |
Collapse
|
56
|
Clinical significance of the B7-H4 as a novel prognostic marker in breast cancer. Gene 2017; 623:24-28. [DOI: 10.1016/j.gene.2017.04.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2017] [Revised: 03/29/2017] [Accepted: 04/04/2017] [Indexed: 01/20/2023]
|
57
|
Abstract
Observations noting the presence of white blood cell infiltrates within tumors date back more than a century, however the cellular and molecular mechanisms regulating tumor immunity continue to be elucidated. The recent successful use of monoclonal antibodies to block immune regulatory pathways to enhance tumor-specific immune responses for the treatment of cancer has encouraged the identification of additional immune regulatory receptor/ligand pathways. Over the past several years, a growing body of data has identified B7-H4 (VTCN1/B7x/B7S1) as a potential therapeutic target for the treatment of cancer. The potential clinical significance of B7-H4 is supported by the high levels of B7-H4 expression found in numerous tumor tissues and correlation of the level of expression on tumor cells with adverse clinical and pathologic features, including tumor aggressiveness. The biological activity of B7-H4 has been associated with decreased inflammatory CD4+ T-cell responses and a correlation between B7-H4-expressing tumor-associated macrophages and FoxP3+ regulatory T cells (Tregs) within the tumor microenvironment. Since B7-H4 is expressed on tumor cells and tumor-associated macrophages in various cancer types, therapeutic blockade of B7-H4 could favorably alter the tumor microenvironment allowing for antigen-specific clearance tumor cells. The present review highlights the therapeutic potential of targeting B7-H4.
Collapse
Affiliation(s)
- Joseph R Podojil
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Stephen D Miller
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| |
Collapse
|
58
|
王 维, 王 丹, 秦 国, 陈 新, 张 毅. 免疫检查点抑制剂在结直肠癌中的应用以及未来发展方向. Shijie Huaren Xiaohua Zazhi 2017; 25:1714-1727. [DOI: 10.11569/wcjd.v25.i19.1714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
结直肠癌(colorectal cancer, CRC)是消化系最常见的恶性肿瘤之一, 在我国, 其发病率及死亡率处于逐年上升趋势, 且总体预后相对较差. 近年来, 免疫治疗的基础和临床研究都获得了快速发展, 已成为肿瘤研究的热点. 其中, 免疫检查点抑制剂已经被批准用于包括CRC在内的多种实体肿瘤的临床治疗. 本文将重点阐述免疫检查点的作用、机制和免疫检查点抑制剂在CRC中应用的最新进展, 以及影响其抗肿瘤疗效的因素. 已经完成和正在进行的临床试验肯定了免疫检查点抑制剂在CRC的治疗中的潜力, 尽管部分患者仍对免疫检查点治疗无应答. 因此, 探究免疫检查点抑制剂治疗CRC患者的敏感因素, 对实现个体化精准治疗至关重要. 未来, 免疫检查点抑制剂有望和其他多种治疗方法相联合, 提高患者反应率, 延长患者的生存期.
Collapse
|
59
|
Stern LA, Csizmar CM, Woldring DR, Wagner CR, Hackel BJ. Titratable Avidity Reduction Enhances Affinity Discrimination in Mammalian Cellular Selections of Yeast-Displayed Ligands. ACS COMBINATORIAL SCIENCE 2017; 19:315-323. [PMID: 28322543 DOI: 10.1021/acscombsci.6b00191] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Yeast surface display selections against mammalian cell monolayers have proven effective in isolating proteins with novel binding activity. Recent advances in this technique allow for the recovery of clones with even micromolar binding affinities. However, no efficient method has been shown for affinity-based selection in this context. This study demonstrates the effectiveness of titratable avidity reduction using dithiothreitol to achieve this goal. A series of epidermal growth factor receptor binding fibronectin domains with a range of affinities are used to quantitatively identify the number of ligands per yeast cell that yield the strongest selectivity between strong, moderate, and weak affinities. Notably, reduction of ligand display to 3,000-6,000 ligands per yeast cell of a 2 nM binder yields 16-fold better selectivity than that to a 17 nM binder. These lessons are applied to affinity maturation of an EpCAM-binding fibronectin population, yielding an enriched pool of ligands with significantly stronger affinity than that of an analogous pool sorted by standard cellular selection methods. Collectively, this study offers a facile approach for affinity selection of yeast-displayed ligands against full-length cellular targets and demonstrates the effectiveness of this method by generating EpCAM-binding ligands that are promising for further applications.
Collapse
Affiliation(s)
- Lawrence A. Stern
- Department
of Chemical Engineering and Materials Science and ‡Department of Medicinal Chemistry, University of Minnesota−Twin Cities, Minneapolis, Minnesota 55455, United States
| | - Clifford M. Csizmar
- Department
of Chemical Engineering and Materials Science and ‡Department of Medicinal Chemistry, University of Minnesota−Twin Cities, Minneapolis, Minnesota 55455, United States
| | - Daniel R. Woldring
- Department
of Chemical Engineering and Materials Science and ‡Department of Medicinal Chemistry, University of Minnesota−Twin Cities, Minneapolis, Minnesota 55455, United States
| | - Carston R. Wagner
- Department
of Chemical Engineering and Materials Science and ‡Department of Medicinal Chemistry, University of Minnesota−Twin Cities, Minneapolis, Minnesota 55455, United States
| | - Benjamin J. Hackel
- Department
of Chemical Engineering and Materials Science and ‡Department of Medicinal Chemistry, University of Minnesota−Twin Cities, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
60
|
Sankin A, Narasimhulu D, John P, Gartrell B, Schoenberg M, Zang X. The expanding repertoire of targets for immune checkpoint inhibition in bladder cancer: What lies beneath the tip of the iceberg, PD-L1. Urol Oncol 2017; 36:459-468. [PMID: 28495553 DOI: 10.1016/j.urolonc.2017.04.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 04/03/2017] [Accepted: 04/09/2017] [Indexed: 01/08/2023]
Abstract
Over the last decade, a new understanding of tumor-immune system interplay has been ushered in, lead in large part by the discovery of immune checkpoints mediated through B7-CD28 family interactions. Therapeutic blockade of the PD-L1 immune checkpoint pathway has already shown great success as a cancer immunotherapy for advanced urothelial carcinoma, leading to durable clinical remissions in an otherwise incurable disease. There are newly described members of the B7-CD28 family including B7-H3, B7x, and HHLA2. These ligands are thought to play an essential role in suppressing T-cell response, leading to immune tolerance of tumors. This feature makes them attractive targets for novel immunotherapy treatment paradigms. Here, we review the literature of current strategies and future directions of immune checkpoint blockade therapy for bladder cancer.
Collapse
Affiliation(s)
- Alexander Sankin
- Department of Urology, Montefiore Medical Center and Albert Einstein College of Medicine, Bronx, New York.
| | - Deepa Narasimhulu
- Department of Obstetrics and Gynecology, Montefiore Medical Center and Albert Einstein College of Medicine, Bronx, New York
| | - Peter John
- Department of Microbiology and Immunology, Montefiore Medical Center and Albert Einstein College of Medicine, Bronx, New York
| | - Benjamin Gartrell
- Department of Oncology, Montefiore Medical Center and Albert Einstein College of Medicine, Bronx, New York
| | - Mark Schoenberg
- Department of Urology, Montefiore Medical Center and Albert Einstein College of Medicine, Bronx, New York
| | - Xingxing Zang
- Department of Urology, Montefiore Medical Center and Albert Einstein College of Medicine, Bronx, New York; Department of Microbiology and Immunology, Montefiore Medical Center and Albert Einstein College of Medicine, Bronx, New York; Department of Oncology, Montefiore Medical Center and Albert Einstein College of Medicine, Bronx, New York
| |
Collapse
|
61
|
Leung J, St-Onge P, Stagg J, Suh WK. Synergistic effects of host B7-H4 deficiency and gemcitabine treatment on tumor regression and anti-tumor T cell immunity in a mouse model. Cancer Immunol Immunother 2017; 66:491-502. [PMID: 28074226 PMCID: PMC11028495 DOI: 10.1007/s00262-016-1950-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 12/20/2016] [Indexed: 02/08/2023]
Abstract
B7-H4 (B7x/B7S1), a B7 family inhibitor of T cell activity, is expressed in multiple human cancers and correlates with decreased infiltrating lymphocytes and poor prognosis. In murine models, tumor-expressed B7-H4 enhances tumor growth and reduces T cell immunity, and blockade of tumor-B7-H4 rescues T cell activity and lowers tumor burden. This implicates B7-H4 as a target for cancer immunotherapy, yet limits the efficacy of B7-H4 blockade exclusively to patients with B7-H4+ tumors. Given the expression of B7-H4 on host immune cells, we have previously shown that BALB/c mice lacking host B7-H4 have enhanced anti-tumor profiles, yet similar 4T1 tumor growth relative to control. Given that T cell-mediated immunotherapies work best for tumors presenting tumor-associated neoantigens, we further investigated the function of host B7-H4 in the growth of a more immunogenic derivative, 4T1-12B, which is known to elicit strong anti-tumor CD8 T cell responses due to expression of a surrogate tumor-specific antigen, firefly luciferase. Notably, B7-H4 knockout hosts not only mounted greater tumor-associated anti-tumor T cell responses, but also displayed reduced tumors. Additionally, B7-H4-deficiency synergized with gemcitabine to further inhibit tumor growth, often leading to tumor eradication and the generation of protective T cell immunity. These findings imply that inhibition of host B7-H4 can enhance anti-tumor T cell immunity in immunogenic cancers, and can be combined with other anti-cancer therapies to further reduce tumor burden regardless of tumor-B7-H4 positivity.
Collapse
Affiliation(s)
- Joanne Leung
- Institut de recherches cliniques de Montréal (IRCM), 110 avenue des Pins Ouest, Montréal, QC, H2W 1R7, Canada
- Department of Microbiology and Immunology, McGill University, Montréal, QC, Canada
| | - Philippe St-Onge
- Institut de recherches cliniques de Montréal (IRCM), 110 avenue des Pins Ouest, Montréal, QC, H2W 1R7, Canada
- Département de Médecine (Programmes de Biologie Moléculaire), Université de Montréal, Montréal, QC, Canada
| | - John Stagg
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Institut du Cancer de Montréal, Faculté de Pharmacie, Université de Montréal, Québec, Canada
| | - Woong-Kyung Suh
- Institut de recherches cliniques de Montréal (IRCM), 110 avenue des Pins Ouest, Montréal, QC, H2W 1R7, Canada.
- Department of Microbiology and Immunology, McGill University, Montréal, QC, Canada.
- Département de Médecine (Programmes de Biologie Moléculaire), Université de Montréal, Montréal, QC, Canada.
- Département de Médecine, Université de Montréal, Montréal, QC, Canada.
- Département de Microbiologie, Infectiologie, et Immunologie, Université de Montréal, Montréal, QC, Canada.
| |
Collapse
|
62
|
MacGregor HL, Ohashi PS. Molecular Pathways: Evaluating the Potential for B7-H4 as an Immunoregulatory Target. Clin Cancer Res 2017; 23:2934-2941. [PMID: 28325750 DOI: 10.1158/1078-0432.ccr-15-2440] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Revised: 12/05/2016] [Accepted: 03/15/2017] [Indexed: 11/16/2022]
Abstract
With the clinical success of CTLA-4 and PD-1 blockade in treating malignancies, there is tremendous interest in finding new ways to augment antitumor responses by targeting other inhibitory molecules. In this review, we describe one such molecule. B7-H4, a member of the B7 family of immunoregulatory proteins, inhibits T cell proliferation and cytokine production through ligation of an unknown receptor expressed by activated T cells. Notably, B7-H4 protein expression is observed in a high proportion of patients' tumors across a wide variety of malignancies. This high expression by tumors in combination with its low or absent protein expression in normal tissues makes B7-H4 an attractive immunotherapeutic target. Preclinical investigation into B7-H4-specific chimeric antigen receptor (CAR) T cells, antibody-mediated blockade of B7-H4, and anti-B7-H4 drug conjugates has shown antitumor efficacy in mouse models. The first clinical trials have been completed to assess the safety and efficacy of a B7-H4 fusion protein in ameliorating rheumatoid arthritis. Clin Cancer Res; 23(12); 2934-41. ©2017 AACR.
Collapse
Affiliation(s)
- Heather L MacGregor
- Campbell Family Institute for Breast Cancer Research, Princess Margaret Cancer Centre, Toronto, Ontario, Canada.,Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Pamela S Ohashi
- Campbell Family Institute for Breast Cancer Research, Princess Margaret Cancer Centre, Toronto, Ontario, Canada. .,Department of Immunology, University of Toronto, Toronto, Ontario, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
63
|
Affiliation(s)
- Ling Ni
- Institute for Immunology and School of Medicine; Tsinghua University; Beijing China
| | - Chen Dong
- Institute for Immunology and School of Medicine; Tsinghua University; Beijing China
| |
Collapse
|
64
|
Janakiram M, Shah UA, Liu W, Zhao A, Schoenberg MP, Zang X. The third group of the B7-CD28 immune checkpoint family: HHLA2, TMIGD2, B7x, and B7-H3. Immunol Rev 2017; 276:26-39. [PMID: 28258693 PMCID: PMC5338461 DOI: 10.1111/imr.12521] [Citation(s) in RCA: 166] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2016] [Revised: 11/02/2016] [Accepted: 11/16/2016] [Indexed: 12/29/2022]
Abstract
The B7-CD28 family of ligands and receptors play important roles in T-cell co-stimulation and co-inhibition. Phylogenetically they can be divided into three groups. The recent discovery of the new molecules (B7-H3 [CD276], B7x [B7-H4/B7S1], and HHLA2 [B7H7/B7-H5]/TMIGD2 [IGPR-1/CD28H]) of the group III has expanded therapeutic possibilities for the treatment of human diseases. In this review, we describe the discovery, structure, and function of B7-H3, B7x, HHLA2, and TMIGD2 in immune regulation. We also discuss their roles in important pathological states such as cancers, autoimmune diseases, transplantation, and infection. Various immunotherapeutical approaches are emerging including antagonistic monoclonal antibodies and agonistic fusion proteins to inhibit or potentiate these molecules and pathways in cancers and autoimmune diseases.
Collapse
Affiliation(s)
- Murali Janakiram
- Department of Medicine, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Oncology, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Urvi A Shah
- Department of Medicine, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Oncology, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Weifeng Liu
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Aimin Zhao
- Department of Obstetrics and Gynecology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Mark P Schoenberg
- Department of Urology, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Xingxing Zang
- Department of Medicine, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Oncology, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Urology, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| |
Collapse
|
65
|
Akiyama Y, Kondou R, Iizuka A, Ohshima K, Urakami K, Nagashima T, Shimoda Y, Tanabe T, Ohnami S, Ohnami S, Kusuhara M, Mochizuki T, Yamaguchi K. Immune response-associated gene analysis of 1,000 cancer patients using whole-exome sequencing and gene expression profiling-Project HOPE. Biomed Res 2017; 37:233-42. [PMID: 27544999 DOI: 10.2220/biomedres.37.233] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Project HOPE (High-tech Omics-based Patient Evaluation) has been progressing since its implementation in 2014 using whole-exome sequencing (WES) and gene expression profiling (GEP). With the aim of evaluating immune status in cancer patients, a gene panel consisting of 164 immune response-associated genes (56 antigen-presenting cell and T-cell-associated genes, 34 cytokine- and metabolism-associated genes, 47 TNF and TNF receptor superfamily genes, and 27 regulatory T-cell-associated genes) was established, and its expression and mutation status were investigated using 1,000 cancer patient-derived tumors. Regarding WES, sequencing and variant calling were performed using the Ion Proton system. The average number of single-nucleotide variants (SNVs) detected per sample was 183 ± 507, and the number of hypermutators with more than 500 total SNVs was 51 cases. Regarding GEP, seven immune response-associated genes (VTCN1, IL2RA, ULBP2, TREM1, MSR1, TNFSF9 and TNFRSF12A) were more than 2-fold overexpressed compared with normal tissues in more than 2 organs. Specifically, the positive rate of PD-L1 expression in all patients was 25.8%, and PD-L1 expression was significantly upregulated in hypermutators. The simultaneous analyses of WES and GEP based on immune response-associated genes are very intriguing tools to screen cancer patients suitable for immune checkpoint antibody therapy.
Collapse
Affiliation(s)
- Yasuto Akiyama
- Immunotherapy Division, Shizuoka Cancer Center Research Institute
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
66
|
|
67
|
Smith JB, Lanitis E, Dangaj D, Buza E, Poussin M, Stashwick C, Scholler N, Powell DJ. Tumor Regression and Delayed Onset Toxicity Following B7-H4 CAR T Cell Therapy. Mol Ther 2016; 24:1987-1999. [PMID: 27439899 PMCID: PMC5154474 DOI: 10.1038/mt.2016.149] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Accepted: 07/12/2016] [Indexed: 12/18/2022] Open
Abstract
B7-H4 protein is frequently overexpressed in ovarian cancer. Here, we engineered T cells with novel B7-H4-specific chimeric antigen receptors (CARs) that recognized both human and murine B7-H4 to test the hypothesis that B7-H4 CAR T cell therapy can be applied safely in preclinical models. B7-H4 CAR T cells specifically secreted IFN-γ and lysed B7-H4(+) targets. In vivo, B7-H4 CAR T cells displayed antitumor reactivity against B7-H4(+) human ovarian tumor xenografts. Unexpectedly, B7-H4 CAR T cell treatment reproducibly showed delayed, lethal toxicity 6-8 weeks after therapy. Comprehensive assessment of murine B7-H4 protein distribution uncovered expression in ductal and mucosal epithelial cells in normal tissues. Postmortem analysis revealed the presence of widespread histologic lesions that correlated with B7-H4(+) expression, and were inconsistent with graft versus host disease. Lastly, expression patterns of B7-H4 protein in normal human tissue were comparable to distribution in mice, advancing our understanding of B7-H4. We conclude that B7-H4 CAR therapy mediates control of cancer outgrowth. However, long-term engraftment of B7-H4 CAR T cells mediates lethal, off-tumor toxicity that is likely due to wide expression of B7-H4 in healthy mouse organs. This model system provides a unique opportunity for preclinical evaluation of safety approaches that limit CAR-mediated toxicity after tumor destruction in vivo.
Collapse
Affiliation(s)
- Jenessa B Smith
- Department of Pathology and Laboratory Medicine, Center for Cellular Immunotherapies, Philadelphia, Pennsylvania, USA; Department of Obstetrics and Gynecology, Ovarian Cancer Research Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Evripidis Lanitis
- Department of Pathology and Laboratory Medicine, Center for Cellular Immunotherapies, Philadelphia, Pennsylvania, USA; Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland
| | - Denarda Dangaj
- Department of Obstetrics and Gynecology, Ovarian Cancer Research Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA; Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland
| | - Elizabeth Buza
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, Pennsylvania, USA
| | - Mathilde Poussin
- Department of Pathology and Laboratory Medicine, Center for Cellular Immunotherapies, Philadelphia, Pennsylvania, USA; Department of Obstetrics and Gynecology, Ovarian Cancer Research Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Caitlin Stashwick
- Department of Obstetrics and Gynecology, Ovarian Cancer Research Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA; Lancaster General Health, Penn Medicine, Lancaster, Pennsylvania, USA
| | - Nathalie Scholler
- Department of Obstetrics and Gynecology, Ovarian Cancer Research Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA; SRI Biosciences, Menlo Park, California, USA
| | - Daniel J Powell
- Department of Pathology and Laboratory Medicine, Center for Cellular Immunotherapies, Philadelphia, Pennsylvania, USA; Department of Obstetrics and Gynecology, Ovarian Cancer Research Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.
| |
Collapse
|
68
|
Iizuka A, Kondou R, Nonomura C, Ashizawa T, Ohshima K, Kusuhara M, Isaka M, Ohde Y, Yamaguchi K, Akiyama Y. Unstable B7-H4 cell surface expression and T-cell redirection as a means of cancer therapy. Oncol Rep 2016; 36:2625-2632. [PMID: 27632942 DOI: 10.3892/or.2016.5084] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Accepted: 05/07/2016] [Indexed: 11/06/2022] Open
Abstract
Tumor immune regulation has been demonstrated in clinical studies using antibodies targeted to the B7/CD28 family. B7 homolog 4 (B7-H4) negatively regulates immune responses and is overexpressed in many types of human cancer, indicating that B7-H4 may be a potential target of cancer therapy. B7-H4 expression is affected by the microenvironment, and its presence has been reported in cancer tissues and immune cells. We found an upregulation of B7-H4 expression using comprehensive whole exome sequencing and gene expression profiling (project HOPE) launched by the Shizuoka Cancer Center based on tumor tissue samples from 1,058 cancer patients. We were successful in producing monoclonal antibodies for B7-H4 and demonstrated B7-H4 dimerization and rapid cell surface disappearance by antibody cross-linking in breast cancer cells, even under typical conditions. These observations may explain why antibody-dependent cellular cytotoxicity (ADCC) did not function in vivo on the B7-H4-expressing tumor cells. Unstable cell surface antigens are not suitable as targets for ADCC, and we therefore performed an indirect ADCC-redirecting T-cell cytotoxicity assay to study B7-H4 using polyclonal anti-mouse IgG antibody-mediated linking. Our results showed the possibility of targeting the B7-H4 molecule as a means of treating cancer.
Collapse
Affiliation(s)
- Akira Iizuka
- Division of Immunotherapy, Shizuoka Cancer Center Research Institute, Shizuoka 411-8777, Japan
| | - Ryota Kondou
- Division of Immunotherapy, Shizuoka Cancer Center Research Institute, Shizuoka 411-8777, Japan
| | - Chizu Nonomura
- Division of Immunotherapy, Shizuoka Cancer Center Research Institute, Shizuoka 411-8777, Japan
| | - Tadashi Ashizawa
- Division of Immunotherapy, Shizuoka Cancer Center Research Institute, Shizuoka 411-8777, Japan
| | - Keiichi Ohshima
- Division of Medical Genetics, Shizuoka Cancer Center Research Institute, Shizuoka 411-8777, Japan
| | - Masatoshi Kusuhara
- Division of Regional Resources, Shizuoka Cancer Center Research Institute, Shizuoka 411-8777, Japan
| | - Mitsuhiro Isaka
- Division of Thoracic Surgery, Shizuoka Cancer Center Hospital, Nagaizumi-cho, Sunto-gun, Shizuoka 411-8777, Japan
| | - Yasuhisa Ohde
- Division of Thoracic Surgery, Shizuoka Cancer Center Hospital, Nagaizumi-cho, Sunto-gun, Shizuoka 411-8777, Japan
| | - Ken Yamaguchi
- Office of the President, Shizuoka Cancer Center Hospital, Nagaizumi-cho, Sunto-gun, Shizuoka 411-8777, Japan
| | - Yasuto Akiyama
- Division of Immunotherapy, Shizuoka Cancer Center Research Institute, Shizuoka 411-8777, Japan
| |
Collapse
|
69
|
Singel KL, Segal BH. Neutrophils in the tumor microenvironment: trying to heal the wound that cannot heal. Immunol Rev 2016; 273:329-43. [PMID: 27558344 PMCID: PMC5477672 DOI: 10.1111/imr.12459] [Citation(s) in RCA: 117] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Neutrophils are the first responders to infection and injury and are critical for antimicrobial host defense. Through the generation of reactive oxidants, activation of granular constituents and neutrophil extracellular traps, neutrophils target microbes and prevent their dissemination. While these pathways are beneficial in the context of trauma and infection, their off-target effects in the context of tumor are variable. Tumor-derived factors have been shown to reprogram the marrow, skewing toward the expansion of myelopoiesis. This can result in stimulation of both neutrophilic leukocytosis and the release of immature granulocytic populations that accumulate in circulation and in the tumor microenvironment. While activated neutrophils have been shown to kill tumor cells, there is growing evidence for neutrophil activation driving tumor progression and metastasis through a number of pathways, including stimulation of thrombosis and angiogenesis, stromal remodeling, and impairment of T cell-dependent anti-tumor immunity. There is also growing appreciation of neutrophil heterogeneity in cancer, with distinct neutrophil populations promoting cancer control or progression. In addition to the effects of tumor on neutrophil responses, anti-neoplastic treatment, including surgery, chemotherapy, and growth factors, can influence neutrophil responses. Future directions for research are expected to result in more mechanistic knowledge of neutrophil biology in the tumor microenvironment that may be exploited as prognostic biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Kelly L. Singel
- Department of Immunology, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Brahm H. Segal
- Department of Immunology, Roswell Park Cancer Institute, Buffalo, NY, USA
- Department of Medicine, Roswell Park Cancer Institute, Buffalo, NY, USA
- Department of Medicine, University at Buffalo Jacobs School of Medicine and Biomedical Sciences, Buffalo, NY, USA
| |
Collapse
|
70
|
Schalper KA, Carvajal-Hausdorf D, McLaughlin J, Altan M, Velcheti V, Gaule P, Sanmamed MF, Chen L, Herbst RS, Rimm DL. Differential Expression and Significance of PD-L1, IDO-1, and B7-H4 in Human Lung Cancer. Clin Cancer Res 2016; 23:370-378. [PMID: 27440266 DOI: 10.1158/1078-0432.ccr-16-0150] [Citation(s) in RCA: 145] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Revised: 07/11/2016] [Accepted: 07/13/2016] [Indexed: 11/16/2022]
Abstract
PURPOSE To determine the expression level, associations, and biological role of PD-L1, IDO-1, and B7-H4 in non-small cell lung cancer (NSCLC). EXPERIMENTAL DESIGN Using multiplexed quantitative immunofluorescence (QIF), we measured the levels of PD-L1, IDO-1, B7-H4, and different tumor-infiltrating lymphoycte (TIL) subsets in 552 stages I-IV lung carcinomas from two independent populations. Associations between the marker levels, TILs, and major clinicopathologic variables were determined. Validation of findings was performed using mRNA expression data from The Cancer Genome Atlas (TCGA) and in vitro stimulation of lung adenocarcinoma A549 cells with IFNγ and IL10. RESULTS PD-L1 was detected in 16.9% and 21.8% of cases in each population. IDO-1 was expressed in 42.6% and 49.8%; and B7-H4 in 12.8% and 22.6% of cases, respectively. Elevated PD-L1 and IDO-1 were consistently associated with prominent B- and T-cell infiltrates, but B7-H4 was not. Coexpression of the three protein markers was infrequent, and comparable results were seen in the lung cancer TCGA dataset. Levels of PD-L1 and IDO-1 (but not B7-H4) were increased by IFNγ stimulation in A549 cells. Treatment with IL10 upregulated B7-H4 but did not affect PD-L1 and IDO-1 levels. CONCLUSIONS PD-L1, IDO-1, and B7-H4 are differentially expressed in human lung carcinomas and show limited co-expression. While PD-L1 and IDO-1 are associated with increased TILs and IFNγ stimulation, B7-H4 is not. The preferential expression of discrete immune evasion pathways in lung cancer could participate in therapeutic resistance and support design of optimal clinical trials. Clin Cancer Res; 23(2); 370-8. ©2016 AACR.
Collapse
MESH Headings
- A549 Cells
- Aged
- B7-H1 Antigen/genetics
- Biomarkers, Tumor/genetics
- Carcinoma, Non-Small-Cell Lung/drug therapy
- Carcinoma, Non-Small-Cell Lung/genetics
- Carcinoma, Non-Small-Cell Lung/pathology
- Disease-Free Survival
- Drug Resistance, Neoplasm/genetics
- Gene Expression Regulation, Neoplastic/drug effects
- Humans
- Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics
- Interferon-gamma/pharmacology
- Interleukin-10/pharmacology
- Lymphocytes, Tumor-Infiltrating/pathology
- Middle Aged
- Neoplasm Staging
- RNA, Messenger/drug effects
- RNA, Messenger/genetics
- V-Set Domain-Containing T-Cell Activation Inhibitor 1/genetics
Collapse
Affiliation(s)
- Kurt A Schalper
- Department of Pathology, Yale School of Medicine, New Haven, Connecticut.
- Medical Oncology Section, Yale School of Medicine, New Haven, Connecticut
- Translational Immuno-oncology Laboratory, Yale Cancer Center, New Haven, Connecticut
| | | | - Joseph McLaughlin
- Medical Oncology Section, Yale School of Medicine, New Haven, Connecticut
| | - Mehmet Altan
- Medical Oncology Section, Yale School of Medicine, New Haven, Connecticut
| | | | - Patricia Gaule
- Department of Pathology, Yale School of Medicine, New Haven, Connecticut
| | | | - Lieping Chen
- Immunobiology, Yale School of Medicine, New Haven, Connecticut
| | - Roy S Herbst
- Medical Oncology Section, Yale School of Medicine, New Haven, Connecticut
| | - David L Rimm
- Department of Pathology, Yale School of Medicine, New Haven, Connecticut
- Medical Oncology Section, Yale School of Medicine, New Haven, Connecticut
| |
Collapse
|
71
|
Stern LA, Schrack IA, Johnson SM, Deshpande A, Bennett NR, Harasymiw LA, Gardner MK, Hackel BJ. Geometry and expression enhance enrichment of functional yeast-displayed ligands via cell panning. Biotechnol Bioeng 2016; 113:2328-41. [PMID: 27144954 DOI: 10.1002/bit.26001] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2015] [Revised: 03/23/2016] [Accepted: 04/25/2016] [Indexed: 01/10/2023]
Abstract
Yeast surface display has proven to be an effective tool in the discovery and evolution of ligands with new or improved binding activity. Selections for binding activity are generally carried out using immobilized or fluorescently labeled soluble domains of target molecules such as recombinant ectodomain fragments. While this method typically provides ligands with high affinity and specificity for the soluble molecular target, translation to binding true membrane-bound cellular target is commonly problematic. Direct selections against mammalian cell surfaces can be carried out either exclusively or in combination with soluble target-based selections to further direct towards ligands for genuine cellular target. Using a series of fibronectin domain, affibody, and Gp2 ligands and human cell lines expressing a range of their targets, epidermal growth factor receptor and carcinoembryonic antigen, this study quantitatively identifies the elements that dictate ligand enrichment and yield. Most notably, extended flexible linkers between ligand and yeast enhance enrichment ratios from 1.4 ± 0.8 to 62 ± 57 for a low-affinity (>600 nM) binder on cells with high target expression and from 14 ± 13 to 74 ± 25 for a high-affinity binder (2 nM) on cells with medium valency. Inversion of the yeast display fusion from C-terminal display to N-terminal display still enables enrichment albeit with 40-97% reduced efficacy. Collectively, this study further enlightens the conditions-while highlighting new approaches-that yield successful enrichment of yeast-displayed binding ligands via panning on mammalian cells. Biotechnol. Bioeng. 2016;113: 2328-2341. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Lawrence A Stern
- Department of Chemical Engineering and Materials Science, University of Minnesota-Twin Cities, Minneapolis, Minnesota, 55455
| | - Ian A Schrack
- Department of Chemical Engineering and Materials Science, University of Minnesota-Twin Cities, Minneapolis, Minnesota, 55455
| | - Sadie M Johnson
- Department of Chemical Engineering and Materials Science, University of Minnesota-Twin Cities, Minneapolis, Minnesota, 55455
| | - Aakash Deshpande
- Department of Chemical Engineering and Materials Science, University of Minnesota-Twin Cities, Minneapolis, Minnesota, 55455
| | - Nathaniel R Bennett
- Department of Chemical Engineering and Materials Science, University of Minnesota-Twin Cities, Minneapolis, Minnesota, 55455
| | - Lauren A Harasymiw
- Department of Genetics, Cell Biology, and Development, University of Minnesota-Twin Cities, Minneapolis, Minnesota
| | - Melissa K Gardner
- Department of Genetics, Cell Biology, and Development, University of Minnesota-Twin Cities, Minneapolis, Minnesota
| | - Benjamin J Hackel
- Department of Chemical Engineering and Materials Science, University of Minnesota-Twin Cities, Minneapolis, Minnesota, 55455.
| |
Collapse
|
72
|
Liang L, Jiang Y, Chen JS, Niu N, Piao J, Ning J, Zu Y, Zhang J, Liu J. B7-H4 expression in ovarian serous carcinoma: a study of 306 cases. Hum Pathol 2016; 57:1-6. [PMID: 27349304 DOI: 10.1016/j.humpath.2016.06.011] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Revised: 05/11/2016] [Accepted: 06/11/2016] [Indexed: 01/07/2023]
Abstract
The B7 family of immune costimulatory ligands is a group of cell surface proteins that bind to the surface receptors of lymphocytes to fine-tune immune responses. The aberrant expression of these proteins plays a key role in tumor immune evasion. Immunotherapy targeting certain B7 family members, including programmed death ligand 1, has proven quite effective in suppressing tumor growth. However, why such therapy works in only a subgroup of tumors is unclear. We hypothesized that other B7 family members, either alone or in concert with programmed death ligand 1, play a crucial role in tumor pathogenesis and progression. We therefore examined the expression of a newly discovered B7 family member, B7-H4, in 306 cases of ovarian serous carcinoma by immunohistochemistry. We found that 91% (267/293) of the high-grade ovarian serous carcinomas and 69% (9/13) of the low-grade ovarian serous carcinomas expressed B7-H4. The difference between B7-H4 expression in high-grade and low-grade ovarian serous carcinoma was statistically significant (P=.002). Moreover, B7-H4 protein expression in high-grade serous carcinoma was associated with tumor stage (P<.01) but not overall survival or disease-free survival. In conclusion, B7-H4 is frequently expressed in ovarian serous carcinomas, especially high-grade serous carcinomas, and may represent a novel immunotherapeutic target in this cancer.
Collapse
Affiliation(s)
- Li Liang
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA 77030
| | - Yi Jiang
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA 77030; Department of Obstetrics and Gynecology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, People's Republic of China 210029
| | - Jun-Song Chen
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA 77030
| | - Na Niu
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA 77030
| | - Jin Piao
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA 77030
| | - Jing Ning
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA 77030
| | - Youli Zu
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, TX, USA 77030.
| | - Jing Zhang
- Department of Pathology, Xijing Hospital, Fourth Military Medical University, Xi'an, People's Republic of China 710032.
| | - Jinsong Liu
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA 77030.
| |
Collapse
|
73
|
Alme AKB, Karir BS, Faltas BM, Drake CG. Blocking immune checkpoints in prostate, kidney, and urothelial cancer: An overview. Urol Oncol 2016; 34:171-81. [PMID: 26923782 PMCID: PMC4834698 DOI: 10.1016/j.urolonc.2016.01.006] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Accepted: 01/25/2016] [Indexed: 12/31/2022]
Abstract
Despite a long history of immunotherapeutic approaches to treatment, most genitourinary malignancies are not cured by existing immunotherapy regimens. More recently, cell surface molecules known as immune checkpoints have become the focus of efforts to develop more effective immunotherapies. Interactions between these molecules and their ligands inhibit the proliferation and function of tumor-specific lymphocytes. A monoclonal antibody blocking 1 of these checkpoints was approved for the treatment of metastatic melanoma and is now being tested in other malignancies. The objective responses seen in these early trials of checkpoint blockade are driving renewed enthusiasm for cancer immunotherapy. There are several ongoing and planned trials in genitourinary malignancies of single-agent inhibitors, as well as combinations targeting multiple checkpoints or adding other types of therapies to checkpoint blockade.
Collapse
Affiliation(s)
- Angela K B Alme
- Department of Oncology, James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD
| | - Beerinder S Karir
- Department of Medicine, Division of Hematology and Medical Oncology, Weill Cornell Medicine, New York, NY
| | - Bishoy M Faltas
- Department of Medicine, Division of Hematology and Medical Oncology, Weill Cornell Medicine, New York, NY
| | - Charles G Drake
- Department of Oncology, James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD; Department of Urology, James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD.
| |
Collapse
|
74
|
Chen C, Huang JA. New insights into the anti-PD-L1 and anti-PD-1 reagents in cancer therapy. EUR J INFLAMM 2016. [DOI: 10.1177/1721727x15626423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Immunotherapy that inhibits the interaction between programmed death ligand 1 (PD-L1), present on the surface of tumor or antigen-presenting cells, and programmed death 1 (PD-1), present on the surface of activated lymphocytes, is generating much excitement and enthusiasm. Although considerable knowledge has been accumulated on anti-PD-L1 and anti-PD-1 reagents, discovering immunotherapy-associated issues still remains a pressing task for the researchers and clinicians.
Collapse
Affiliation(s)
- Cheng Chen
- Respiratory Department, The First Affiliated Hospital of Soochow University, Suzhou, PR China
- Institute of Respiratory Diseases, Soochow University, Suzhou, PR China
| | - Jian-An Huang
- Respiratory Department, The First Affiliated Hospital of Soochow University, Suzhou, PR China
- Institute of Respiratory Diseases, Soochow University, Suzhou, PR China
| |
Collapse
|
75
|
Khan ANH, Kolomeyevskaya N, Singel KL, Grimm MJ, Moysich KB, Daudi S, Grzankowski KS, Lele S, Ylagan L, Webster GA, Abrams SI, Odunsi K, Segal BH. Targeting myeloid cells in the tumor microenvironment enhances vaccine efficacy in murine epithelial ovarian cancer. Oncotarget 2016; 6:11310-26. [PMID: 25888637 PMCID: PMC4484458 DOI: 10.18632/oncotarget.3597] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Accepted: 02/20/2015] [Indexed: 12/16/2022] Open
Abstract
Epithelial ovarian cancer (EOC) is typically diagnosed at advanced stages, and is associated with a high relapse rate. Patients in remission are ideal candidates for immunotherapy aimed at cure or prolonging disease-free periods. However, immunosuppressive pathways in the tumor microenvironment are obstacles to durable anti-tumor immunity. In a metastatic syngeneic mouse model of EOC, immunosuppressive macrophages and myeloid-derived suppressor cells (MDSCs) accumulate in the local tumor environment. In addition, resident peritoneal macrophages from non-tumor-bearing mice were highly immunosuppressive, abrogating stimulated T cell proliferation in a cell contact-dependent manner. Immunization with microparticles containing TLR9 and NOD-2 ligands (MIS416) significantly prolonged survival in tumor-bearing mice. The strategy of MIS416 immunization followed by anti-CD11b administration further delayed tumor progression, thereby establishing the proof of principle that myeloid depletion can enhance vaccine efficacy. In patients with advanced EOC, ascites analysis showed substantial heterogeneity in the relative proportions of myeloid subsets and their immunosuppressive properties. Together, these findings point to immunosuppressive myeloid cells in the EOC microenvironment as targets to enhance vaccination. Further studies of myeloid cell accumulation and functional phenotypes in the EOC microenvironment may identify patients who are likely to benefit from vaccination combined with approaches that deplete tumor-associated myeloid cells.
Collapse
Affiliation(s)
- Anm Nazmul H Khan
- Department of Medicine, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Nonna Kolomeyevskaya
- Department of Gynecologic Oncology, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Kelly L Singel
- Department of Immunology, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Melissa J Grimm
- Department of Medicine, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Kirsten B Moysich
- Department of Cancer Prevention and Control, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Sayeema Daudi
- Department of Gynecologic Oncology, Roswell Park Cancer Institute, Buffalo, NY, USA
| | | | - Sashikant Lele
- Department of Gynecologic Oncology, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Lourdes Ylagan
- Department of Pathology, Roswell Park Cancer Institute, Buffalo, NY, USA
| | | | - Scott I Abrams
- Department of Immunology, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Kunle Odunsi
- Department of Gynecologic Oncology, Roswell Park Cancer Institute, Buffalo, NY, USA.,Department of Immunology, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Brahm H Segal
- Department of Medicine, Roswell Park Cancer Institute, Buffalo, NY, USA.,Department of Immunology, Roswell Park Cancer Institute, Buffalo, NY, USA.,Department of Medicine, University at Buffalo School of Medicine, Buffalo, NY, USA
| |
Collapse
|
76
|
Beavis PA, Slaney CY, Kershaw MH, Gyorki D, Neeson PJ, Darcy PK. Reprogramming the tumor microenvironment to enhance adoptive cellular therapy. Semin Immunol 2016; 28:64-72. [DOI: 10.1016/j.smim.2015.11.003] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 10/29/2015] [Accepted: 11/04/2015] [Indexed: 12/22/2022]
|
77
|
Abstract
Since the development of therapeutic antibodies the demand of recombinant human antibodies is steadily increasing. Traditionally, therapeutic antibodies were generated by immunization of rat or mice, the generation of hybridoma clones, cloning of the antibody genes and subsequent humanization and engineering of the lead candidates. In the last few years, techniques were developed that use transgenic animals with a human antibody gene repertoire. Here, modern recombinant DNA technologies can be combined with well established immunization and hybridoma technologies to generate already affinity maturated human antibodies. An alternative are in vitro technologies which enabled the generation of fully human antibodies from antibody gene libraries that even exceed the human antibody repertoire. Specific antibodies can be isolated from these libraries in a very short time and therefore reduce the development time of an antibody drug at a very early stage.In this review, we describe different technologies that are currently used for the in vitro and in vivo generation of human antibodies.
Collapse
|
78
|
Zhang C, Li Y, Wang Y. Diagnostic value of serum B7-H4 for hepatocellular carcinoma. J Surg Res 2015; 197:301-6. [DOI: 10.1016/j.jss.2015.04.034] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2015] [Revised: 03/15/2015] [Accepted: 04/09/2015] [Indexed: 10/23/2022]
|
79
|
Turnis ME, Andrews LP, Vignali DAA. Inhibitory receptors as targets for cancer immunotherapy. Eur J Immunol 2015; 45:1892-905. [PMID: 26018646 PMCID: PMC4549156 DOI: 10.1002/eji.201344413] [Citation(s) in RCA: 107] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Revised: 04/25/2015] [Accepted: 05/26/2015] [Indexed: 12/11/2022]
Abstract
Inhibitory receptors expressed on T cells control immune responses while limiting autoimmunity. However, tumors can hijack these "checkpoints" for protection from immune attack. Tumor-specific T cells that exhibit an exhausted, unresponsive phenotype express high levels of inhibitory receptors including CTLA4, PD1, and LAG3, among others. Intratumoral regulatory T cells promote immunosuppression and also express multiple inhibitory receptors. Overcoming this inhibitory receptor-mediated immune tolerance has thus been a major focus of recent cancer immunotherapeutic developments. Here, we review how boosting the host's immune system by blocking inhibitory receptor signaling with antagonistic mAbs restores the capacity of T cells to drive durable antitumor immune responses. Clinical trials targeting the CTLA4 and PD1 pathways have shown durable effects in multiple tumor types. Many combinatorial therapies are currently being investigated with encouraging results that highlight enhanced antitumor immunogenicity and improved patient survival. Finally, we will discuss the ongoing identification and dissection of novel T-cell inhibitory receptor pathways, which could lead to the development of new combinatorial therapeutic approaches.
Collapse
Affiliation(s)
- Meghan E Turnis
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | | | - Dario A A Vignali
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
80
|
Winkler I, Wilczynska B, Bojarska-Junak A, Gogacz M, Adamiak A, Postawski K, Darmochwal-Kolarz D, Rechberger T, Tabarkiewicz J. Regulatory T lymphocytes and transforming growth factor beta in epithelial ovarian tumors-prognostic significance. J Ovarian Res 2015; 8:39. [PMID: 26077607 PMCID: PMC4513978 DOI: 10.1186/s13048-015-0164-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2014] [Accepted: 06/01/2015] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Regulatory T lymphocytes (Treg) are characterized by the presence of CD4+ surface antigen. Today the transcription factor FOXP3 is considered to be the most specific marker of Treg cells. The aim of the study was to estimate the percentage of Treg in peripheral blood and the tissue of the epithelial ovarian tumor and blood serum TGF-beta concentrations and relationships between them. Moreover, the aim of the study was to answer the question whether the percentage of Treg lymphocytes affects the time of survival in patients with ovarian cancer. METHODS The patients were divided into four groups, depending on the histopathological examination result: I--a group without any pathology within the ovaries (C; n = 20), II--a group with benign tumors (B; n = 25), III - with borderline tumors (BR; n = 11), IV--a group with cancer of the ovary (M; n = 24). The percentage of Treg lymphocytes in peripheral blood and the tissue was assessed using the flow cytometry method. TGF-beta cytokine concentration was estimated with the ELISA immunoenzymatic test. Statistical analysis of the results was conducted using the computer program Statistica 10.0PL (StatSoft, Inc). RESULTS No significant differences were found in percentages of Treg lymphocytes in peripheral blood between individual groups of patients (p = 0.11). However, we observed marked differences in the tissue of malignant and non-malignant tumors between individual groups of patients (p = 0.003). The analysis with the post hoc test revealed significantly higher TGF-beta concentration in the group of women with malignant tumors. Moreover, no relationship was found between TGF-beta concentration and the percentage of Treg cells in peripheral blood and tumors of the ovary. No correlation was found between the percentage of Treg lymphocytes in peripheral blood (p = 0.4) and the tissue of ovarian tumors (p = 0.3) and the time of survival of patients with ovarian cancer. CONCLUSIONS The recruitment of Treg lymphocytes toward the tumor is one of the mechanisms of escape of neoplasm from the response of the immune system. The percentage of Treg lymphocytes in peripheral blood and the neoplastic tissue does not influence the time of survival of patients with ovarian cancer.
Collapse
Affiliation(s)
- Izabela Winkler
- II Department of Surgical Gynaecology, Medical University in Lublin, Jaczewski Street, 20-954, Lublin, Poland.
| | - Barbara Wilczynska
- Department of Clinical Immunology, Medical University in Lublin, Chodźki 4a Street, 20-093, Lublin, Poland. .,Department of Paediatric Endocrinology and Diabetology with Endocrine-Metabolic Laboratory, Chodźki 2 Street, 20-093, Lublin, Poland.
| | - Agnieszka Bojarska-Junak
- Department of Clinical Immunology, Medical University in Lublin, Chodźki 4a Street, 20-093, Lublin, Poland.
| | - Marek Gogacz
- II Department of Surgical Gynaecology, Medical University in Lublin, Jaczewski Street, 20-954, Lublin, Poland.
| | - Aneta Adamiak
- II Department of Surgical Gynaecology, Medical University in Lublin, Jaczewski Street, 20-954, Lublin, Poland.
| | - Krzysztof Postawski
- II Department of Surgical Gynaecology, Medical University in Lublin, Jaczewski Street, 20-954, Lublin, Poland.
| | - Dorota Darmochwal-Kolarz
- Department of Obstetrics and Perinatology, Medical University of Lublin, Jaczewski Street, 20-954, Lublin, Poland. .,Centre for Innovative Research in Medical and Natural Sciences, Medical Faculty of University of Rzeszów, Warzywna Street, 35-959, Rzeszów, Poland.
| | - Tomasz Rechberger
- II Department of Surgical Gynaecology, Medical University in Lublin, Jaczewski Street, 20-954, Lublin, Poland.
| | - Jacek Tabarkiewicz
- Centre for Innovative Research in Medical and Natural Sciences, Medical Faculty of University of Rzeszów, Warzywna Street, 35-959, Rzeszów, Poland.
| |
Collapse
|
81
|
De Wolf K, Vermaelen K, De Meerleer G, Lambrecht BN, Ost P. The potential of radiotherapy to enhance the efficacy of renal cell carcinoma therapy. Oncoimmunology 2015; 4:e1042198. [PMID: 26464810 PMCID: PMC4590014 DOI: 10.1080/2162402x.2015.1042198] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Revised: 04/11/2015] [Accepted: 04/14/2015] [Indexed: 12/13/2022] Open
Abstract
Renal cell carcinoma (RCC) is an immunogenic tumor, but uses several immune-suppressive mechanisms to shift the balance from tumor immune response toward tumor growth. Although RCC has traditionally been considered to be radiation resistant, recent evidence suggests that hypofractionated radiotherapy contributes to systemic antitumor immunity. Because the efficacy of antitumor immune responses depends on the complex balance between diverse immune cells and progressing tumor cells, radiotherapy alone is unlikely to induce persistent antitumor immunity. Therefore, the combination of radiotherapy with drugs having synergistic immunomodulatory properties holds great promise with the optimal timing and sequence of modalities depending on the agent used. We highlight the immunomodulatory properties of targeted therapies, such as tyrosine kinase inhibitors, mammalian target of rapamycin (mTOR) inhibitors and vascular endothelial growth factor (VEGF) neutralizing antibodies, and will suggest a combination schedule with radiotherapy based on the available literature. We also address the combination of radiotherapy with innovative treatments in the field of immunotherapy.
Collapse
Affiliation(s)
- Katrien De Wolf
- Department of Radiation Oncology and Experimental Cancer Research; Ghent University Hospital ; Ghent, Belgium
| | - Karim Vermaelen
- Tumor Immunology Laboratory; Department of Pulmonary Medicine; Ghent University Hospital ; Ghent, Belgium
| | - Gert De Meerleer
- Department of Radiation Oncology and Experimental Cancer Research; Ghent University Hospital ; Ghent, Belgium
| | - Bart N Lambrecht
- Unit Immunoregulation and Mucosal Immunology; VIB Inflammation Research Center ; Ghent, Belgium ; GROUP-ID Consortium; Ghent University and University Hospital ; Ghent, Belgium ; Department of Respiratory Medicine; Ghent University ; Ghent, Belgium ; Department of Pulmonary Medicine; Erasmus MC ; Rotterdam, The Netherlands
| | - Piet Ost
- Department of Radiation Oncology and Experimental Cancer Research; Ghent University Hospital ; Ghent, Belgium
| |
Collapse
|
82
|
Radaelli E, Hermans E, Omodho L, Francis A, Vander Borght S, Marine JC, van den Oord J, Amant F. Spontaneous Post-Transplant Disorders in NOD.Cg- Prkdcscid Il2rgtm1Sug/JicTac (NOG) Mice Engrafted with Patient-Derived Metastatic Melanomas. PLoS One 2015; 10:e0124974. [PMID: 25996609 PMCID: PMC4440639 DOI: 10.1371/journal.pone.0124974] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Accepted: 03/20/2015] [Indexed: 12/18/2022] Open
Abstract
Patient-derived tumor xenograft (PDTX) approach is nowadays considered a reliable preclinical model to study in vivo cancer biology and therapeutic response. NOD scid and Il2rg-deficient mice represent the "gold standard" host for the generation of PDTXs. Compared to other immunocompromised murine lines, these mice offers several advantages including higher engraftment rate, longer lifespan and improved morphological and molecular preservation of patient-derived neoplasms. Here we describe a spectrum of previously uncharacterized post-transplant disorders affecting 14/116 (12%) NOD.Cg- Prkdcscid Il2rgtm1Sug/JicTac (NOG) mice subcutaneously engrafted with patient-derived metastatic melanomas. Affected mice exhibited extensive scaling/crusting dermatitis (13/14) associated with emaciation (13/14) and poor/unsuccessful tumor engraftment (14/14). In this context, the following pathological conditions have been recognized and characterized in details: (i) immunoinflammatory disorders with features of graft versus host disease (14/14); (ii) reactive lymphoid infiltrates effacing xenografted tumors (8/14); (iii) post-transplant B cell lymphomas associated with Epstein-Barr virus reactivation (2/14). We demonstrate that all these entities are driven by co-transplanted human immune cells populating patient-derived tumor samples. Since the exploding interest in the utilization of NOD scid and Il2rg-deficient mice for the establishment of PDTX platforms, it is of uppermost importance to raise the awareness of the limitations associated with this model. The disorders here described adversely impact tumor engraftment rate and animal lifespan, potentially representing a major confounding factor in the context of efficacy and personalized therapy studies. The occurrence of these conditions in the NOG model reflects the ability of this mouse line to promote efficient engraftment of human immune cells. Co-transplanted human lymphoid cells have indeed the potential to colonize the recipient mouse initiating the post-transplant conditions here reported. On the other hand, the evidence of an immune response of human origin against the xenotransplanted melanoma opens intriguing perspectives for the establishment of suitable preclinical models of anti-melanoma immunotherapy.
Collapse
Affiliation(s)
- Enrico Radaelli
- VIB11 Center for the Biology of Disease, KU Leuven Center for Human Genetics, Leuven, Belgium
- InfraMouse, KU Leuven-VIB, Leuven, Belgium
| | - Els Hermans
- Gynaecological Oncology, UZ Leuven—Department of Oncology, KU Leuven, Leuven, Belgium
- * E-mail:
| | - Lorna Omodho
- VIB11 Center for the Biology of Disease, KU Leuven Center for Human Genetics, Leuven, Belgium
| | - Annick Francis
- VIB11 Center for the Biology of Disease, KU Leuven Center for Human Genetics, Leuven, Belgium
- InfraMouse, KU Leuven-VIB, Leuven, Belgium
| | - Sara Vander Borght
- Department of Pathology, Laboratory of Morphology and Molecular Pathology, University Hospitals of Leuven, Leuven, Belgium
| | - Jean-Christophe Marine
- Laboratory for Molecular Cancer Biology, VIB11 Center for the Biology of Disease, KU Leuven Center for Human Genetics, Leuven, Belgium
| | - Joost van den Oord
- Translational Cell and Tissue Research, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Frédéric Amant
- Gynaecological Oncology, UZ Leuven—Department of Oncology, KU Leuven, Leuven, Belgium
| |
Collapse
|
83
|
Abstract
The accumulation of tumor infiltrating lymphocytes (TILs) in ovarian cancer is prognostic for increased survival while increases in immunosuppressive regulatory T-cells (Tregs) are associated with poor outcomes. Approaches that bolster tumor-reactive TILs may limit tumor progression. However, identifying tumor-reactive TILs in ovarian cancer has been challenging, though adoptive TIL therapy in patients has been encouraging. Other forms of TIL immunomodulation remain under investigation including Treg depletion, antibody-based checkpoint modification, activation and amplification using dendritic cells, antigen presenting cells or IL-2 cytokine culture, adjuvant cytokine injections, and gene-engineered T-cells. Many approaches to TIL manipulation inhibit ovarian cancer progression in preclinical or clinical studies as monotherapy. Here, we review the impact of TILs in ovarian cancer and attempts to mobilize TILs to halt tumor progression. We conclude that effective TIL therapy for ovarian cancer is at the brink of translation and optimal TIL activity may require combined methodologies to deliver clinically-relevant treatment.
Collapse
Affiliation(s)
- Phillip P Santoiemma
- a Ovarian Cancer Research Center ; Department of Obstetrics and Gynecology ; Perelman School of Medicine; University of Pennsylvania ; Philadelphia , PA USA
| | | |
Collapse
|
84
|
Santoiemma PP, Powell DJ. Tumor infiltrating lymphocytes in ovarian cancer. Cancer Biol Ther 2015. [PMID: 25894333 DOI: 10.1080/15384047.2015.1040960]+[] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/30/2022] Open
Abstract
The accumulation of tumor infiltrating lymphocytes (TILs) in ovarian cancer is prognostic for increased survival while increases in immunosuppressive regulatory T-cells (Tregs) are associated with poor outcomes. Approaches that bolster tumor-reactive TILs may limit tumor progression. However, identifying tumor-reactive TILs in ovarian cancer has been challenging, though adoptive TIL therapy in patients has been encouraging. Other forms of TIL immunomodulation remain under investigation including Treg depletion, antibody-based checkpoint modification, activation and amplification using dendritic cells, antigen presenting cells or IL-2 cytokine culture, adjuvant cytokine injections, and gene-engineered T-cells. Many approaches to TIL manipulation inhibit ovarian cancer progression in preclinical or clinical studies as monotherapy. Here, we review the impact of TILs in ovarian cancer and attempts to mobilize TILs to halt tumor progression. We conclude that effective TIL therapy for ovarian cancer is at the brink of translation and optimal TIL activity may require combined methodologies to deliver clinically-relevant treatment.
Collapse
Affiliation(s)
- Phillip P Santoiemma
- a Ovarian Cancer Research Center ; Department of Obstetrics and Gynecology ; Perelman School of Medicine; University of Pennsylvania ; Philadelphia , PA USA
| | | |
Collapse
|
85
|
Abstract
The accumulation of tumor infiltrating lymphocytes (TILs) in ovarian cancer is prognostic for increased survival while increases in immunosuppressive regulatory T-cells (Tregs) are associated with poor outcomes. Approaches that bolster tumor-reactive TILs may limit tumor progression. However, identifying tumor-reactive TILs in ovarian cancer has been challenging, though adoptive TIL therapy in patients has been encouraging. Other forms of TIL immunomodulation remain under investigation including Treg depletion, antibody-based checkpoint modification, activation and amplification using dendritic cells, antigen presenting cells or IL-2 cytokine culture, adjuvant cytokine injections, and gene-engineered T-cells. Many approaches to TIL manipulation inhibit ovarian cancer progression in preclinical or clinical studies as monotherapy. Here, we review the impact of TILs in ovarian cancer and attempts to mobilize TILs to halt tumor progression. We conclude that effective TIL therapy for ovarian cancer is at the brink of translation and optimal TIL activity may require combined methodologies to deliver clinically-relevant treatment.
Collapse
Affiliation(s)
- Phillip P Santoiemma
- a Ovarian Cancer Research Center ; Department of Obstetrics and Gynecology ; Perelman School of Medicine; University of Pennsylvania ; Philadelphia , PA USA
| | | |
Collapse
|
86
|
Immunotherapeutic approaches to ovarian cancer treatment. J Immunother Cancer 2015; 3:7. [PMID: 25806106 PMCID: PMC4372273 DOI: 10.1186/s40425-015-0051-7] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Accepted: 02/16/2015] [Indexed: 01/03/2023] Open
Abstract
Despite advances in combinatorial chemotherapy regimens and the advent of intraperitoneal chemotherapy administration, current therapeutic options for ovarian cancer patients are inadequate. Immunotherapy offers a novel and promising therapeutic strategy for treating ovarian tumors. Following the demonstration of the immunogenicity of ovarian tumors, multiple immunotherapeutic modalities have been developed. Antibody-based therapies, immune checkpoint blockade, cancer vaccines, and chimeric antigen receptor-modified T cells have demonstrated preclinical success and entered clinical testing. In this review, we discuss these promising immunotherapeutic approaches and emphasize the importance of combinatorial treatment strategies and biomarker discovery.
Collapse
|
87
|
Isolation and Validation of Anti-B7-H4 scFvs from an Ovarian Cancer scFv Yeast-Display Library. Methods Mol Biol 2015; 1319:37-49. [PMID: 26060068 DOI: 10.1007/978-1-4939-2748-7_2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
B7-H4 (VTCN1, B7x, B7s) is an inhibitory modulator of T-cell response implicated in antigen tolerization. As such, B7-H4 is an immune checkpoint of potential therapeutic interest. To generate anti-B7-H4 targeting reagents, we isolated antibodies by differential cell screening of a yeast-display library of recombinant antibodies (scFvs) derived from ovarian cancer patients and we screened for functional scFvs capable to interfere with B7-H4-mediated inhibition of antitumor responses. We found one antibody binding to B7-H4 that could restore antitumor T cell responses. This chapter gives an overview of the methods we developed to isolate a functional anti-B7-H4 antibody fragment.
Collapse
|
88
|
Leung J, Suh WK. The CD28-B7 Family in Anti-Tumor Immunity: Emerging Concepts in Cancer Immunotherapy. Immune Netw 2014; 14:265-76. [PMID: 25550693 PMCID: PMC4275384 DOI: 10.4110/in.2014.14.6.265] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2014] [Revised: 11/20/2014] [Accepted: 11/28/2014] [Indexed: 12/15/2022] Open
Abstract
The interactions between B7 molecules and CD28-family receptors are crucial in the regulation of adaptive cellular immunity. In cancer, the aberrant expression of co-inhibitory B7 molecules has been attributed to reduced anti-tumor immunity and cancer immune evasion, prompting the development of cancer therapeutics that can restore T cell function. Murine tumor models have provided significant support for the targeting of multiple immune checkpoints involving CTLA-4, PD-1, ICOS, B7-H3 and B7-H4 during tumor growth, and clinical studies investigating the therapeutic effects of CTLA-4 and PD-1 blockade have shown exceptionally promising results in patients with advanced melanoma and other cancers. The expression pattern of co-inhibitory B7 ligands in the tumor microenvironment has also been largely correlated with poor patient prognosis, and recent evidence suggests that the presence of several B7 molecules may predict the responsiveness of immunotherapies that rely on pre-existing tumor-associated immune responses. While monotherapies blocking T cell co-inhibition have beneficial effects in reducing tumor burden, combinatorial immunotherapy targeting multiple immune checkpoints involved in various stages of the anti-tumor response has led to the most substantial impact on tumor reduction. In this review, we will examine the contributions of B7- and CD28-family members in the context of cancer development, and discuss the implications of current human findings in cancer immunotherapy.
Collapse
Affiliation(s)
- Joanne Leung
- Institut de Recherches Cliniques de Montréal (IRCM), Montreal, QC H2W 1R7, Canada. ; Department of Microbiology and Immunology, McGill University, Montreal, QC H3A 0G4, Canada
| | - Woong-Kyung Suh
- Institut de Recherches Cliniques de Montréal (IRCM), Montreal, QC H2W 1R7, Canada. ; Department of Microbiology and Immunology, McGill University, Montreal, QC H3A 0G4, Canada. ; Department of Medicine; Department of Microbiology, Infectiology, and Immunology, University of Montreal, Montreal, QC H3T 1J4, Canada
| |
Collapse
|
89
|
Byers JT, Paniccia A, Kaplan J, Koenig M, Kahn N, Wilson L, Chen L, Schulick RD, Edil BH, Zhu Y. Expression of the Novel Costimulatory Molecule B7-H5 in Pancreatic Cancer. Ann Surg Oncol 2014; 22 Suppl 3:S1574-9. [DOI: 10.1245/s10434-014-4293-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Indexed: 11/18/2022]
|
90
|
Jeon H, Vigdorovich V, Garrett-Thomson SC, Janakiram M, Ramagopal UA, Abadi YM, Lee JS, Scandiuzzi L, Ohaegbulam KC, Chinai JM, Zhao R, Yao Y, Mao Y, Sparano JA, Almo SC, Zang X. Structure and cancer immunotherapy of the B7 family member B7x. Cell Rep 2014; 9:1089-98. [PMID: 25437562 PMCID: PMC4250833 DOI: 10.1016/j.celrep.2014.09.053] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2014] [Revised: 08/29/2014] [Accepted: 09/26/2014] [Indexed: 11/24/2022] Open
Abstract
B7x (B7-H4 or B7S1) is a member of the B7 family that can inhibit T cell function. B7x protein is absent in most normal human tissues and immune cells, but it is overexpressed in human cancers and often correlates with negative clinical outcome. The expression pattern and function of B7x suggest that it may be a potent immunosuppressive pathway in human cancers. Here, we determined the crystal structure of the human B7x immunoglobulin variable (IgV) domain at 1.59 Å resolution and mapped the epitopes recognized by monoclonal antibodies. We developed an in vivo system to screen therapeutic monoclonal antibodies against B7x and found that the clone 1H3 significantly inhibited growth of B7x-expressing tumors in vivo via multiple mechanisms. Furthermore, the surviving mice given 1H3 treatment were resistant to tumor rechallenge. Our data suggest that targeting B7x on tumors is a promising cancer immunotherapy and humanized 1H3 may be efficacious for immunotherapy of human cancers.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/metabolism
- Antibodies, Monoclonal/pharmacology
- Antibody-Dependent Cell Cytotoxicity/drug effects
- Cell Proliferation/drug effects
- Crystallography, X-Ray
- Disease Models, Animal
- Epitope Mapping
- Epitopes/chemistry
- Epitopes/immunology
- Humans
- Immunosuppression Therapy
- Immunotherapy
- Killer Cells, Natural/drug effects
- Killer Cells, Natural/immunology
- Lymphocytes, Tumor-Infiltrating/drug effects
- Lymphocytes, Tumor-Infiltrating/immunology
- Mice, Inbred BALB C
- Models, Molecular
- Neoplasms/immunology
- Neoplasms/pathology
- Neoplasms/therapy
- Protein Binding/drug effects
- Protein Structure, Tertiary
- Surface Plasmon Resonance
- Survival Analysis
- T-Lymphocytes/immunology
- Tumor Microenvironment/drug effects
- V-Set Domain-Containing T-Cell Activation Inhibitor 1/chemistry
- V-Set Domain-Containing T-Cell Activation Inhibitor 1/immunology
- V-Set Domain-Containing T-Cell Activation Inhibitor 1/metabolism
Collapse
Affiliation(s)
- Hyungjun Jeon
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Vladimir Vigdorovich
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Sarah C Garrett-Thomson
- Department of Biochemistry, Physiology and Biophysics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Murali Janakiram
- Department of Oncology, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY 10467, USA
| | - Udupi A Ramagopal
- Department of Biochemistry, Physiology and Biophysics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Yael M Abadi
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Jun Sik Lee
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Lisa Scandiuzzi
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Kim C Ohaegbulam
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Jordan M Chinai
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Ruihua Zhao
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Yu Yao
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Ying Mao
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Joseph A Sparano
- Department of Oncology, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY 10467, USA
| | - Steven C Almo
- Department of Biochemistry, Physiology and Biophysics, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| | - Xingxing Zang
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Department of Oncology, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY 10467, USA.
| |
Collapse
|
91
|
Abstract
Optimal T cell response is dependent not only on T cell receptor activation, but also on additional signaling from coreceptors. The main coreceptors include B7 and tumor necrosis factor family members. They exert costimulatory or coinhibitory effects, and their balance determines the fate of T cell response. In normal conditions, costimulators facilitate the development of protective immune response, whereas coinhibitors dampen inflammation to avoid organ/tissue damage from excessive immune reaction. In the tumor microenvironment, the balance is garbled: inhibitory pathways predominate, and T cell response is impaired. The importance of cosignaling in the tumor immune response has been experimentally and clinically demonstrated. New therapeutic strategies targeting T cell cosignaling, especially coinhibitory molecules, are under active experimental and clinical investigation. This review summarizes the functions of main T cell cosignaling axes and discusses their clinical application.
Collapse
|
92
|
Lanitis E, Smith JB, Dangaj D, Flingai S, Poussin M, Xu S, Czerniecki BJ, Li YF, Robbins PF, Powell DJ. A human ErbB2-specific T-cell receptor confers potent antitumor effector functions in genetically engineered primary cytotoxic lymphocytes. Hum Gene Ther 2014; 25:730-9. [PMID: 25003657 PMCID: PMC4137348 DOI: 10.1089/hum.2014.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2014] [Accepted: 06/20/2014] [Indexed: 01/10/2023] Open
Abstract
The ErbB2 protein is a member of the tyrosine kinase family of growth factor receptors that is overexpressed in cancers of the breast, ovary, stomach, kidney, colon, and lung, and therefore represents an attractive candidate antigen for targeted cancer immunotherapy. Cytotoxic T lymphocytes specific for various immunogenic ErbB2 peptides have been described, but they often exhibit both poor functional avidity and tumor reactivity. In order to generate potent CD8(+) T cells with specificity for the ErbB2(369-377) peptide, we performed one round of in vitro peptide stimulation of CD8(+) T cells isolated from an HLA-A2(+) patient who was previously vaccinated with autologous dendritic cells pulsed with HLA class I ErbB2 peptides. Using this approach, we enriched highly avid ErbB2-reactive T cells with strong ErbB2-specific, antitumor effector functions. We then stimulated these ErbB2-reactive T cells with ErbB2(+) HLA-A2(+) tumor cells in vitro and sorted tumor-activated ErbB2(369-377) peptide T cells, which allowed for the isolation of a novel T-cell receptor (TCR) with ErbB2(369-377) peptide specificity. Primary human CD8(+) T cells genetically modified to express this ErbB2-specific TCR specifically bound ErbB2(369-377) peptide containing HLA-A2 tetramers, and efficiently recognized target cells pulsed with low nanomolar concentrations of ErbB2(369-377) peptide as well as nonpulsed ErbB2(+) HLA-A2(+) tumor cell lines in vitro. In a novel xenograft model, ErbB2-redirected T cells also significantly delayed progression of ErbB2(+) HLA-A2(+) human tumor in vivo. Together, these results support the notion that redirection of normal T-cell specificity by TCR gene transfer can have potential applications in the adoptive immunotherapy of ErbB2-expressing malignancies.
Collapse
Affiliation(s)
- Evripidis Lanitis
- Ovarian Cancer Research Center, Department of Obstetrics and Gynecology, University of Pennsylvania, Philadelphia, PA 19104
| | - Jenessa B. Smith
- Ovarian Cancer Research Center, Department of Obstetrics and Gynecology, University of Pennsylvania, Philadelphia, PA 19104
| | - Denarda Dangaj
- Ovarian Cancer Research Center, Department of Obstetrics and Gynecology, University of Pennsylvania, Philadelphia, PA 19104
| | - Seleeke Flingai
- Ovarian Cancer Research Center, Department of Obstetrics and Gynecology, University of Pennsylvania, Philadelphia, PA 19104
| | - Mathilde Poussin
- Ovarian Cancer Research Center, Department of Obstetrics and Gynecology, University of Pennsylvania, Philadelphia, PA 19104
| | - Shuwen Xu
- Department of Surgery, University of Pennsylvania Medical Center, Philadelphia, PA 19104
| | - Brian J. Czerniecki
- Department of Surgery, University of Pennsylvania Medical Center, Philadelphia, PA 19104
| | - Yong F. Li
- Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Paul F. Robbins
- Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Daniel J. Powell
- Ovarian Cancer Research Center, Department of Obstetrics and Gynecology, University of Pennsylvania, Philadelphia, PA 19104
- Abramson Cancer Center, Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104
| |
Collapse
|
93
|
B7-H4 as a potential target for immunotherapy for gynecologic cancers: a closer look. Gynecol Oncol 2014; 134:181-189. [PMID: 24657487 DOI: 10.1016/j.ygyno.2014.03.553] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2013] [Revised: 03/14/2014] [Accepted: 03/16/2014] [Indexed: 12/16/2022]
Abstract
B7-H4 is a transmembrane protein that binds an unknown receptor on activated T cells resulting in inhibition of T-cell effector function via cell cycle arrest, decreased proliferation, and reduced IL-2 production. B7-H4 is up-regulated on the surface of cancer cells and immunosuppressive tumor-associated macrophages (TAMs) in a variety of human cancers. Notably, B7-H4 expression levels inversely correlate with patient survival in ovarian cancer, making B7-H4 an attractive candidate for therapeutic intervention. Here, we summarize the experimental data and methodologies that have revealed B7-H4's mRNA and protein expression and function in both mice and humans since its discovery in 2003, with a specific focus on B7-H4's role in ovarian cancer. We also underscore the discrepancies in published data due to high variability in methodology and use of different antibodies, most of which are not commercially available. Finally, since B7-H4 is expressed on tumor cells and TAMs in various cancer types, directing therapeutics against B7-H4 could have tremendous synergistic outcomes in favorably altering the tumor micro-environment and eliminating cancer cells. We highlight the therapeutic potential of targeting B7-H4, both by comparing other negative immune modulators such as PD-1 and CTLA-4 and by identifying novel methods to target B7-H4 directly or indirectly to overcome B7-H4-mediated T-cell inhibition.
Collapse
|
94
|
Xu Y, Zhu S, Song M, Liu W, Liu C, Li Y, Wang M. B7-H4 expression and its role in interleukin-2/interferon treatment of clear cell renal cell carcinoma. Oncol Lett 2014; 7:1474-1478. [PMID: 24765159 PMCID: PMC3997729 DOI: 10.3892/ol.2014.1961] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2013] [Accepted: 12/16/2013] [Indexed: 12/20/2022] Open
Abstract
The immunological mechanism mediated by T cells is the main therapeutic target in the treatment of renal cell carcinoma (RCC) with interleukin (IL)-2 and interferon (IFN)-α. The aim of the present study was to evaluate the role of B7-H4 in the IL-2, IFN-α and IFN-γ treatment of clear cell RCC (ccRCC). A total of 154 paraffin-embedded ccRCC tissues were studied using immunohistochemistry, which subsequently indicated that positive B7-H4 expression is associated with adverse clinical features in ccRCC. The effects of IL-2, IFN-α and IFN-γ on B7-H4 expression in a ccRCC cell line were evaluated at the mRNA and protein levels. In addition, the effect of B7-H4 on the killing activity of T cells was detected. B7-H4 expression was identified to be upregulated by IL-2, IFN-α and IFN-γ, of which, IFN-γ was the most capable. Additionally, blocking of B7-H4/B7-H4 ligand interactions may rescue the killing activity of T cells. Altogether, the observations of the current study showed that the immune escape pathway induced by B7-H4 may be one of the most important reasons for the low efficacy of IL-2 and IFN-α and the inability to observe the efficacy of IFN-γ in mRCC. This indicates that B7-H4 may be used as a new molecular biology marker to select treatment options for patients with ccRCC.
Collapse
Affiliation(s)
- Yipeng Xu
- Department of Urology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, P.R. China
| | - Shaoxing Zhu
- Department of Urology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, P.R. China
| | - Mei Song
- Department of Ultrasonography, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, P.R. China
| | - Weihui Liu
- Department of Urology, Fujian Medical University Second Affiliated Hospital, Quanzhou, Fujian 362000, P.R. China
| | - Chengyi Liu
- Department of Urology, Lu'an Affiliated Hospital of Anhui Medical University, Lu'an, Anhui 237005, P.R. China
| | - Yongsheng Li
- Department of Urology, The Union Hospital of Fujian Medical University, Fuzhou, Fujian 350001, P.R. China
| | - Min Wang
- Institute of Ophthalmology, Shandong Academy of Medical Sciences, Qingdao, Shandong 266071, P.R. China
| |
Collapse
|
95
|
Bhatia A, Kumar Y. Cellular and molecular mechanisms in cancer immune escape: a comprehensive review. Expert Rev Clin Immunol 2013; 10:41-62. [PMID: 24325346 DOI: 10.1586/1744666x.2014.865519] [Citation(s) in RCA: 95] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Immune escape is the final phase of cancer immunoediting process wherein cancer modulates our immune system to escape from being destroyed by it. Many cellular and molecular events govern the cancer's evasion of host immune response. The tumor undergoes continuous remodeling at the genetic, epigenetic and metabolic level to acquire resistance to apoptosis. At the same time, it effectively modifies all the components of the host's immunome so as to escape from its antitumor effects. Moreover, it induces accumulation of suppressive cells like Treg and myeloid derived suppressor cells and factors which also enable it to elude the immune system. Recent research in this area helps in defining the role of newer players like miRNAs and exosomes in immune escape. The immunotherapeutic approaches developed to target the escape phase appear quite promising; however, the quest for a perfect therapeutic agent that can achieve maximum cure with minimal toxicity continues.
Collapse
Affiliation(s)
- Alka Bhatia
- Department of Experimental Medicine & Biotechnology, PGIMER, Chandigarh-160012, India
| | | |
Collapse
|
96
|
Vasaturo A, Di Blasio S, Peeters DGA, de Koning CCH, de Vries JM, Figdor CG, Hato SV. Clinical Implications of Co-Inhibitory Molecule Expression in the Tumor Microenvironment for DC Vaccination: A Game of Stop and Go. Front Immunol 2013; 4:417. [PMID: 24348481 PMCID: PMC3847559 DOI: 10.3389/fimmu.2013.00417] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2013] [Accepted: 11/15/2013] [Indexed: 12/13/2022] Open
Abstract
The aim of therapeutic dendritic cell (DC) vaccines in cancer immunotherapy is to activate cytotoxic T cells to recognize and attack the tumor. T cell activation requires the interaction of the T cell receptor with a cognate major-histocompatibility complex-peptide complex. Although initiated by antigen engagement, it is the complex balance between co-stimulatory and co-inhibitory signals on DCs that results in T cell activation or tolerance. Even when already activated, tumor-specific T cells can be neutralized by the expression of co-inhibitory molecules on tumor cells. These and other immunosuppressive cues in the tumor microenvironment are major factors currently hampering the application of DC vaccination. In this review, we discuss recent data regarding the essential and complex role of co-inhibitory molecules in regulating the immune response within the tumor microenvironment. In particular, possible therapeutic intervention strategies aimed at reversing or neutralizing suppressive networks within the tumor microenvironment will be emphasized. Importantly, blocking co-inhibitory molecule signaling, often referred to as immune checkpoint blockade, does not necessarily lead to an effective activation of tumor-specific T cells. Therefore, combination of checkpoint blockade with other immune potentiating therapeutic strategies, such as DC vaccination, might serve as a synergistic combination, capable of reversing effector T cells immunosuppression while at the same time increasing the efficacy of T cell-mediated immunotherapies. This will ultimately result in long-term anti-tumor immunity.
Collapse
Affiliation(s)
- Angela Vasaturo
- Department of Tumor Immunology, Nijmegen Centre for Molecular Life Sciences, Radboud University Nijmegen Medical Centre , Nijmegen , Netherlands
| | - Stefania Di Blasio
- Department of Tumor Immunology, Nijmegen Centre for Molecular Life Sciences, Radboud University Nijmegen Medical Centre , Nijmegen , Netherlands
| | - Deborah G A Peeters
- Department of Tumor Immunology, Nijmegen Centre for Molecular Life Sciences, Radboud University Nijmegen Medical Centre , Nijmegen , Netherlands
| | - Coco C H de Koning
- Department of Tumor Immunology, Nijmegen Centre for Molecular Life Sciences, Radboud University Nijmegen Medical Centre , Nijmegen , Netherlands
| | - Jolanda M de Vries
- Department of Tumor Immunology, Nijmegen Centre for Molecular Life Sciences, Radboud University Nijmegen Medical Centre , Nijmegen , Netherlands ; Department of Medical Oncology, Nijmegen Centre for Molecular Life Sciences, Radboud University Nijmegen Medical Centre , Nijmegen , Netherlands
| | - Carl G Figdor
- Department of Tumor Immunology, Nijmegen Centre for Molecular Life Sciences, Radboud University Nijmegen Medical Centre , Nijmegen , Netherlands
| | - Stanleyson V Hato
- Department of Tumor Immunology, Nijmegen Centre for Molecular Life Sciences, Radboud University Nijmegen Medical Centre , Nijmegen , Netherlands
| |
Collapse
|
97
|
Butt AQ, Mills KHG. Immunosuppressive networks and checkpoints controlling antitumor immunity and their blockade in the development of cancer immunotherapeutics and vaccines. Oncogene 2013; 33:4623-31. [PMID: 24141774 DOI: 10.1038/onc.2013.432] [Citation(s) in RCA: 107] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2013] [Revised: 08/21/2013] [Accepted: 08/22/2013] [Indexed: 01/04/2023]
Abstract
Vaccines that promote protective adaptive immune responses have been successfully developed against a range of infectious diseases, and these are normally administered prior to exposure with the relevant virus or bacteria. Adaptive immunity also plays a critical role in the control of tumors. Immunotherapeutics and vaccines that promote effector T cell responses have the potential to eliminate tumors when used in a therapeutic setting. However, the induction of protective antitumor immunity is compromised by innate immunosuppressive mechanisms and regulatory cells that often dominate the tumor microenvironment. Recent studies have shown that blocking these suppressor cells and immune checkpoints to allow induction of antitumor immunity is a successful immunotherapeutic modality for the treatment of cancer. Furthermore, stimulation of innate and consequently adaptive immune responses with concomitant inhibition of immune suppression, especially that mediated by regulatory T (Treg) cells, is emerging as a promising approach to enhance the efficacy of therapeutic vaccines against cancer. This review describes the immunosuppressive mechanisms controlling antitumor immunity and the novel strategies being employed to design effective immunotherapeutics against tumors based on inhibition of suppressor cells or blockade of immune checkpoints to allow induction of more potent effector T cell responses. This review also discusses the potential of using a combination of adjuvants with inhibition of immune checkpoint or suppressor cells for therapeutic vaccines and the translation of pre-clinical studies to the next-generation vaccines against cancer in humans.
Collapse
Affiliation(s)
- A Q Butt
- Immune Regulation Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - K H G Mills
- Immune Regulation Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
98
|
Dangaj D, Scholler N. Blocking the B7-H4 pathway with novel recombinant antibodies enhances T cell-mediated antitumor responses. Oncoimmunology 2013; 2:e25913. [PMID: 24083083 PMCID: PMC3782523 DOI: 10.4161/onci.25913] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2013] [Accepted: 07/26/2013] [Indexed: 12/22/2022] Open
Abstract
B7-H4 inhibits T-cell activation and is widely expressed by solid neoplasms. We have recently demonstrated that the expression of B7-H4 on the surface of malignant cells in vivo is inducible, and that novel anti-B7-H4 recombinant antibodies can reverse the inhibition of tumor-specific T cells. Thus, antibodies targeting the B7-H4 pathways may extend the survival of cancer patients by restoring T cell-mediated antitumor responses.
Collapse
Affiliation(s)
- Denarda Dangaj
- Department of Oncology; Ludwig Cancer Research Center; University of Lausanne; Lausanne, Switzerland
| | | |
Collapse
|