1
|
Ohmine K, Uchibori R. Novel immunotherapies in multiple myeloma. Int J Hematol 2022; 115:799-810. [PMID: 35583724 DOI: 10.1007/s12185-022-03365-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 04/18/2022] [Accepted: 04/19/2022] [Indexed: 11/24/2022]
Abstract
For a substantial period, options for the treatment of multiple myeloma (MM) were limited; however, the advent of novel therapies into clinical practice in the 1990s resulted in dramatic changes in the prognosis of the disease. Subsequently, new proteasome inhibitors and immunomodulators with innovations in efficacy and toxicity were introduced; yet there remains a spectrum of patients with poor outcomes with current treatment strategies. One of the causes of disease progression in MM is the loss of the ability of the dysfunctional immune environment to control virulent cell clones. In recent years, therapies to overcome the immunosuppressive tumor microenvironment and activate the host immune system have shown promise in MM, especially in relapsed and refractory disease. Clinical use of this approach has been approved for several immunotherapies, and a number of studies are currently underway in clinical trials. This review outlines three of the newest and most promising approaches being investigated to enhance the immune system against MM: (1) overcoming immunosuppression with checkpoint inhibitors, (2) boosting immunity against tumors with vaccines, and (3) enhancing immune effectors with adoptive cell therapy. Information on the latest clinical trials in each class will be provided, and further developments will be discussed.
Collapse
Affiliation(s)
- Ken Ohmine
- Division of Hematology, Department of Medicine, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi, 329-0498, Japan.
- Division of Immuno-Gene and Cell Therapy (Takara Bio), Jichi Medical University, Shimotsuke, Tochigi, Japan.
| | - Ryosuke Uchibori
- Division of Immuno-Gene and Cell Therapy (Takara Bio), Jichi Medical University, Shimotsuke, Tochigi, Japan
| |
Collapse
|
2
|
Verheye E, Bravo Melgar J, Deschoemaeker S, Raes G, Maes A, De Bruyne E, Menu E, Vanderkerken K, Laoui D, De Veirman K. Dendritic Cell-Based Immunotherapy in Multiple Myeloma: Challenges, Opportunities, and Future Directions. Int J Mol Sci 2022; 23:904. [PMID: 35055096 PMCID: PMC8778019 DOI: 10.3390/ijms23020904] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 01/11/2022] [Indexed: 12/12/2022] Open
Abstract
Immunotherapeutic approaches, including adoptive cell therapy, revolutionized treatment in multiple myeloma (MM). As dendritic cells (DCs) are professional antigen-presenting cells and key initiators of tumor-specific immune responses, DC-based immunotherapy represents an attractive therapeutic approach in cancer. The past years, various DC-based approaches, using particularly ex-vivo-generated monocyte-derived DCs, have been tested in preclinical and clinical MM studies. However, long-term and durable responses in MM patients were limited, potentially attributed to the source of monocyte-derived DCs and the immunosuppressive bone marrow microenvironment. In this review, we briefly summarize the DC development in the bone marrow niche and the phenotypical and functional characteristics of the major DC subsets. We address the known DC deficiencies in MM and give an overview of the DC-based vaccination protocols that were tested in MM patients. Lastly, we also provide strategies to improve the efficacy of DC vaccines using new, improved DC-based approaches and combination therapies for MM patients.
Collapse
Affiliation(s)
- Emma Verheye
- Laboratory of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel, 1090 Brussel, Belgium; (E.V.); (A.M.); (E.D.B.); (E.M.); (K.V.)
- Laboratory of Myeloid Cell Immunology, VIB Center for Inflammation Research, 1050 Brussels, Belgium; (J.B.M.); (S.D.); (G.R.)
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, 1050 Brussels, Belgium
| | - Jesús Bravo Melgar
- Laboratory of Myeloid Cell Immunology, VIB Center for Inflammation Research, 1050 Brussels, Belgium; (J.B.M.); (S.D.); (G.R.)
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, 1050 Brussels, Belgium
| | - Sofie Deschoemaeker
- Laboratory of Myeloid Cell Immunology, VIB Center for Inflammation Research, 1050 Brussels, Belgium; (J.B.M.); (S.D.); (G.R.)
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, 1050 Brussels, Belgium
| | - Geert Raes
- Laboratory of Myeloid Cell Immunology, VIB Center for Inflammation Research, 1050 Brussels, Belgium; (J.B.M.); (S.D.); (G.R.)
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, 1050 Brussels, Belgium
| | - Anke Maes
- Laboratory of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel, 1090 Brussel, Belgium; (E.V.); (A.M.); (E.D.B.); (E.M.); (K.V.)
| | - Elke De Bruyne
- Laboratory of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel, 1090 Brussel, Belgium; (E.V.); (A.M.); (E.D.B.); (E.M.); (K.V.)
| | - Eline Menu
- Laboratory of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel, 1090 Brussel, Belgium; (E.V.); (A.M.); (E.D.B.); (E.M.); (K.V.)
| | - Karin Vanderkerken
- Laboratory of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel, 1090 Brussel, Belgium; (E.V.); (A.M.); (E.D.B.); (E.M.); (K.V.)
| | - Damya Laoui
- Laboratory of Myeloid Cell Immunology, VIB Center for Inflammation Research, 1050 Brussels, Belgium; (J.B.M.); (S.D.); (G.R.)
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, 1050 Brussels, Belgium
| | - Kim De Veirman
- Laboratory of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel, 1090 Brussel, Belgium; (E.V.); (A.M.); (E.D.B.); (E.M.); (K.V.)
| |
Collapse
|
3
|
Poplawska M, Dutta D, Lee Y, Lim SH. Sperm protein 17 targeting for epithelial ovarian cancer treatment in the era of modern immunoengineering. Mol Ther Oncolytics 2021; 23:378-386. [PMID: 34853809 PMCID: PMC8604669 DOI: 10.1016/j.omto.2021.10.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
|
4
|
Cancer-Testis Antigens in Triple-Negative Breast Cancer: Role and Potential Utility in Clinical Practice. Cancers (Basel) 2021; 13:cancers13153875. [PMID: 34359776 PMCID: PMC8345750 DOI: 10.3390/cancers13153875] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 07/22/2021] [Accepted: 07/27/2021] [Indexed: 12/15/2022] Open
Abstract
Breast cancer cells commonly express tumour-associated antigens that can induce immune responses to eradicate the tumour. Triple-negative breast cancer (TNBC) is a form of breast cancer lacking the expression of hormone receptors and cerbB2 (HER2) and tends to be more aggressive and associated with poorer prognoses due to the limited treatment options. Characterisation of biomarkers or treatment targets is thus of great significance in revealing additional therapeutic options. Cancer-testis antigens (CTAs) are tumour-associated antigens that have garnered strong attention as potential clinical biomarkers in targeted immunotherapy due to their cancer-restricted expressions and robust immunogenicity. Previous clinical studies reported that CTAs correlated with negative hormonal status, advanced tumour behaviour and a poor prognosis in a variety of cancers. Various studies also demonstrated the oncogenic potential of CTAs in cell proliferation by inhibiting cell death and inducing metastasis. Multiple clinical trials are in progress to evaluate the role of CTAs as treatment targets in various cancers. CTAs hold great promise as potential treatment targets and biomarkers in cancer, and further research could be conducted on elucidating the mechanism of actions of CTAs in breast cancer or combination therapy with other immune modulators. In the current review, we summarise the current understandings of CTAs in TNBC, addressing the role and utility of CTAs in TNBC, as well as discussing the potential applications and advantage of incorporating CTAs in clinical practise.
Collapse
|
5
|
Song Y, Qi X, Kang J, Wang X, Ou N, Zhu J, Wang S, Liu X. Identification of new biomarkers in immune microenvironment of testicular germ cell tumour. Andrologia 2021; 53:e13986. [PMID: 33544925 DOI: 10.1111/and.13986] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Revised: 12/24/2020] [Accepted: 01/06/2021] [Indexed: 12/24/2022] Open
Abstract
To seek novel prognostic biomarkers for testicular germ cell tumour (TGCT) and investigate the tumour immune microenvironment, we identified critical differentially expressed genes (DEGs) by overlapping GSE1818 dataset from Gene Expression Omnibus (GEO). Protein-protein interaction (PPI) network was used to investigate key modules and hub genes. Functional enrichment analysis was performed to investigate the underlying molecular functions of the DEGs in TGCT development and progression. The following survival analysis based on The Cancer Genome Atlas (TCGA) TGCT dataset indicated that AKAP4, SPA17 and TNP1 are correlated with TGCT prognosis. Immunohistochemistry and quantitative real-time polymerase chain reaction verified the down-regulation of the 3 hub genes in TGCT. Gene set enrichment analysis was conducted to further explore the role of the 3 hub genes in TGCT respectively. In addition, TGCT samples had high infiltration of CD8+ T cells, M0 and M1 macrophage cells, and resting myeloid dendritic cells in immune microenvironment. We also constructed the microRNA-gene regulatory networks to identify the key upstream microRNAs in TGCT. In conclusion, our findings indicated that AKAP4, SPA17 and TNP1 are promising biomarkers of TGCT. AKAP4 and TNP1 might regulate immune cells infiltration in immune microenvironment.
Collapse
Affiliation(s)
- Yuxuan Song
- Department of Urology, Tianjin Medical University General Hospital, Tianjin, China
| | - Xiangjie Qi
- Department of Urology, Linzi District People's Hospital, Zibo, China
| | - Jiaqi Kang
- Department of Urology, Tianjin Medical University General Hospital, Tianjin, China
| | - Xiao Wang
- Department of Urology, Tianjin Medical University General Hospital, Tianjin, China
| | - Ningjing Ou
- Department of Urology, Tianjin Medical University General Hospital, Tianjin, China
| | - Jun Zhu
- Department of Urology, Tianjin Medical University General Hospital, Tianjin, China
| | - Shangren Wang
- Department of Urology, Tianjin Medical University General Hospital, Tianjin, China
| | - Xiaoqiang Liu
- Department of Urology, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
6
|
Jakobsen MK, Gjerstorff MF. CAR T-Cell Cancer Therapy Targeting Surface Cancer/Testis Antigens. Front Immunol 2020; 11:1568. [PMID: 32983080 PMCID: PMC7492268 DOI: 10.3389/fimmu.2020.01568] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 06/15/2020] [Indexed: 12/11/2022] Open
Affiliation(s)
- Mie K Jakobsen
- Department of Cancer and Inflammation Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Morten F Gjerstorff
- Department of Cancer and Inflammation Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark.,Department of Oncology, Odense University Hospital, Odense, Denmark.,Academy of Geriatric Cancer Research (AgeCare), Odense University Hospital, Odense, Denmark
| |
Collapse
|
7
|
Mirandola L, Chiriva-Internati M, Bresalier R, Piccotti L, Grizzi F, Marincola FM. A novel method for efficient generation of antigen-specific effector T-cells using dendritic cells transduced with recombinant adeno-associated virus and p38 kinase blockade. J Transl Med 2019; 17:424. [PMID: 31878933 PMCID: PMC6931250 DOI: 10.1186/s12967-019-02163-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 11/30/2019] [Indexed: 12/11/2022] Open
Abstract
Background The inefficacy of standard therapeutic strategies for ovarian cancer is reflected by the enduring poor prognosis of this malignancy. Due to the potential for exquisite specificity, sensitivity and long-term memory, immunotherapy offers an alternative modality for durable control of the disease, provided appropriate antigens can be identified and
presented in the right context. Methods We tested a novel dendritic cell vaccine formulation to reprogram autologous antigen-specific T-cells in vitro, in vivo in a murine model of ovarian cancer, and ex vivo using human cells from patients. Results We show that dendritic cells (DCs) treated with a p38 MAPK inhibitor and transduced with a recombinant adenovirus associated vector (AAV) expressing Sperm protein (Sp) 17 are highly effective in generating antigen-specific T-cell cytotoxic response against ovarian cancer cells. Additionally, these DCs enhanced the differentiation of effector T-cells while reducing the frequency of Foxp3+ T-reg cells in vitro. Conclusions This work provides a rationale for translation of pharmacologically reprogrammed DCs into clinical trials for prevention of tumor recurrence and progression in high-risk ovarian cancer patients.
Collapse
Affiliation(s)
| | - Maurizio Chiriva-Internati
- Kiromic, Inc, 7707 Fannin St., Suite 140, Houston, TX, 77054, USA. .,Division of Internal Medicine, Department of Gastroenterology Hepatology and Nutrition, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA.
| | - Robert Bresalier
- Division of Internal Medicine, Department of Gastroenterology Hepatology and Nutrition, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Lucia Piccotti
- Kiromic, Inc, 7707 Fannin St., Suite 140, Houston, TX, 77054, USA
| | - Fabio Grizzi
- Department of Immunology and Inflammation, Humanitas Clinical and Research Center, Via Manzoni 56, 20089, Rozzano, Milan, Italy
| | | |
Collapse
|
8
|
Hu J, Chen X, Zhang X, Yuan X, Yang M, Dai H, Yang W, Zhou Q, Wen W, Wang Q, Qin W, Zhao A. A fusion-protein approach enabling mammalian cell production of tumor targeting protein domains for therapeutic development. Protein Sci 2018; 27:933-944. [PMID: 29500915 PMCID: PMC5916118 DOI: 10.1002/pro.3399] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 02/11/2018] [Accepted: 03/01/2018] [Indexed: 02/05/2023]
Abstract
A single chain Fv fragment (scFv) is a fusion of the variable regions of heavy (VH ) and light (VL ) chains of immunoglobulins. They are important elements of chimeric antigen receptors for cancer therapy. We sought to produce a panel of 16 extracellular protein domains of tumor markers for use in scFv yeast library screenings. A series of vectors comprising various combinations of expression elements was made, but expression was unpredictable and more than half of the protein domains could not be produced using any of the constructs. Here we describe a novel fusion expression system based on mouse TEM7 (tumor endothelial marker 7), which could facilitate protein expression. With this approach we could produce all but one of the tumor marker domains that could not otherwise be expressed. In addition, we demonstrated that the tumor associated antigen hFZD10 produced as a fusion protein with mTEM7 could be used to enrich scFv antibodies from a yeast display library. Collectively our study demonstrates the potential of specific fusion proteins based on mTEM7 in enabling mammalian cell production of tumor targeting protein domains for therapeutic development.
Collapse
Affiliation(s)
- Jia Hu
- Lung Cancer Research CenterWest China Hospital, Sichuan UniversityChengduChina
- Abramson Cancer Center, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvania
| | - Xiang Chen
- Abramson Cancer Center, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvania
| | - Xuhua Zhang
- Abramson Cancer Center, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvania
- School of Life SciencesZhengzhou UniversityZhengzhouChina
| | - Xiaopeng Yuan
- Abramson Cancer Center, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvania
- Zhujiang Hospital, SouthernMedical UniversityGuangzhouChina
| | - Mingjuan Yang
- Abramson Cancer Center, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvania
| | - Hui Dai
- Xinjiang Karamay Central HospitalKaramay CityXinjiangChina
| | - Wei Yang
- Abramson Cancer Center, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvania
| | - Qinghua Zhou
- Lung Cancer Research CenterWest China Hospital, Sichuan UniversityChengduChina
| | - Weihong Wen
- State Key Laboratory of Cancer Biology, Department of ImmunologyXijing Hospital, Fourth Military Medical UniversityChinaXi'an
| | - Qirui Wang
- Abramson Cancer Center, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvania
- College of Traditional Chinese MedicineSouthernMedical UniversityGuang DongChina
| | - Weijun Qin
- Department of UrologyXijing Hospital, Fourth Military Medical UniversityChinaXi'an
| | - Aizhi Zhao
- Abramson Cancer Center, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvania
| |
Collapse
|
9
|
Ma J, Li Q, Yu Z, Cao Z, Liu S, Chen L, Li H, Gao S, Yan T, Wang Y, Liu Q. Immunotherapy Strategies Against Multiple Myeloma. Technol Cancer Res Treat 2017. [PMCID: PMC5762093 DOI: 10.1177/1533034617743155] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
Multiple myeloma is a monoclonal B-cell malignancy characterized by an accumulation of malignant plasma cells in the bone marrow, the presence of a monoclonal protein in the serum and/or urine, decreased normal immunoglobulin levels, and lytic bone disease. Patients with multiple myeloma benefit from combination therapy including novel therapeutic agents followed by autologous stem cell transplantation prolonged maintenance therapy. However, multiple myeloma remains incurable; most patients with multiple myeloma will eventually become resistant to chemotherapy, and progression or relapse of the disease is inevitable. Immunotherapy represents a novel therapeutic approach with few adverse effects and good targeting capability that might be a powerful pool to allow long-term control of minimal residual disease. This article reviews the literature evaluating 4 major immunotherapeutic approaches for multiple myeloma including cellular immunotherapy, humoral immunotherapy, radio immunotherapy, and immunomodulation.
Collapse
Affiliation(s)
- Jing Ma
- Tianjin Key Laboratory of Cancer Prevention and Therapy, Department of Hematology and Blood and Marrow Transplantation, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Hexi District, Tianjin, People’s Republic of China
| | - Qian Li
- Tianjin Key Laboratory of Cancer Prevention and Therapy, Department of Hematology and Blood and Marrow Transplantation, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Hexi District, Tianjin, People’s Republic of China
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin, People’s Republic of China
| | - Zhen Yu
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Disease Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, People’s Republic of China
| | - Zeng Cao
- Tianjin Key Laboratory of Cancer Prevention and Therapy, Department of Hematology and Blood and Marrow Transplantation, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Hexi District, Tianjin, People’s Republic of China
| | - Su Liu
- Tianjin Key Laboratory of Cancer Prevention and Therapy, Department of Hematology and Blood and Marrow Transplantation, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Hexi District, Tianjin, People’s Republic of China
| | - Lin Chen
- Tianjin Key Laboratory of Cancer Prevention and Therapy, Department of Hematology and Blood and Marrow Transplantation, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Hexi District, Tianjin, People’s Republic of China
| | - Han Li
- Tianjin Key Laboratory of Cancer Prevention and Therapy, Department of Hematology and Blood and Marrow Transplantation, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Hexi District, Tianjin, People’s Republic of China
| | - Shuang Gao
- Tianjin Key Laboratory of Cancer Prevention and Therapy, Department of Hematology and Blood and Marrow Transplantation, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Hexi District, Tianjin, People’s Republic of China
| | - Tinghui Yan
- Tianjin Key Laboratory of Cancer Prevention and Therapy, Department of Hematology and Blood and Marrow Transplantation, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Hexi District, Tianjin, People’s Republic of China
| | - Yafei Wang
- Tianjin Key Laboratory of Cancer Prevention and Therapy, Department of Hematology and Blood and Marrow Transplantation, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Hexi District, Tianjin, People’s Republic of China
| | - Qiang Liu
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin, People’s Republic of China
| |
Collapse
|
10
|
Sp17 Protein Expression and Major Histocompatibility Class I and II Epitope Presentation in Diffuse Large B Cell Lymphoma Patients. Adv Hematol 2017; 2017:6527306. [PMID: 29204156 PMCID: PMC5674480 DOI: 10.1155/2017/6527306] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2017] [Accepted: 09/12/2017] [Indexed: 12/29/2022] Open
Abstract
Improved therapies are urgently needed for patients with diffuse large B cell lymphoma (DLBCL). Success using immune checkpoint inhibitors and chimeric antigen receptor T cell technology has fuelled demand for validated cancer epitopes. Immunogenic cancer testis antigens (CTAs), with their widespread expression in many tumours but highly restricted normal tissue distribution, represent attractive immunotherapeutic targets that may improve treatment options for DLBCL and other malignancies. Sperm protein 17 (Sp17), a CTA reported to be immunogenic in ovarian cancer and myeloma patients, is expressed in DLBCL. The aim of the present study was to investigate Sp17 epitope presentation via the presence of a cytotoxic T cell (CTL) and a CD4 T-helper (Th) response in DLBCL patients. A significant γ-interferon CTL response was detected in peripheral blood mononuclear cells of 13/31 DLBCL patients following short-term cell stimulation with two novel HLA-A⁎0201 peptides and one previously reported HLA-A⁎0101-restricted nine-mer Sp17 peptide. No significant responses were detected in the HLA-A⁎0201-negative DLBCL patients or four healthy subjects. A novel immunogenic 20-mer CD4 Th Sp17 peptide was detected in 8/17 DLBCL patients. This is the first report of a CTL and a CD4 Th response to Sp17 in DLBCL and supports Sp17 as a potential immunotherapeutic target for DLBCL.
Collapse
|
11
|
Mirandola L, Pedretti E, Figueroa JA, Chiaramonte R, Colombo M, Chapman C, Grizzi F, Patrinicola F, Kast WM, Nguyen DD, Rahman RL, Daver N, Ruvolo P, Post SM, Bresalier RS, Chiriva-Internati M. Cancer testis antigen Sperm Protein 17 as a new target for triple negative breast cancer immunotherapy. Oncotarget 2017; 8:74378-74390. [PMID: 29088794 PMCID: PMC5650349 DOI: 10.18632/oncotarget.20102] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 05/31/2017] [Indexed: 01/09/2023] Open
Abstract
Breast carcinoma is a major health issue for millions of women. Current therapies have serious side effects, and are only partially effective in patients with metastatic tumors. Thus, the need for novel and less toxic therapies is urgent. Moreover, hormonal and antibody therapies effective in other subtypes are not effective in Triple Negative Breast Cancer (TNBC). Immunotherapeutic strategies directed against specific tumor-associated antigens (TAAs) and mediated by specific cytotoxic T lymphocytes (CTL) have been largely underexplored in this disease. Cancer-testis antigens (CTA) are a group of TAAs displaying the ideal characteristics of promising vaccine targets, i.e. strong immunogenicity and cancer specificity. The CTA, Sperm Protein 17 (SP17), has been found to be aberrantly expressed in different neoplasms, including ovarian and esophageal cancers, nervous system tumors and multiple myeloma, and has been suggested as a candidate target for immunotherapy. Here, we evaluated SP17 expression levels in breast cancer cell lines, invasive ductal breast carcinoma, including patients with TNBC, and adjacent non-neoplastic breast tissue, and determined whether SP17 was capable of generating SP17-specific cytotoxic T lymphocytes in vitro. We showed that SP17 is expressed in breast cancer cell lines and primary breast tumors and importantly in TNBC subtype, but not in adjacent non-tumoral breast tissue or unaffected tissues, except in male germinal cells. Furthermore, we detected specific anti-SP17 antibodies in patients’ sera and we generated SP17-specific, HLA class I-restricted, cytotoxic T lymphocytes capable of efficiently killing breast cancer cells.
Collapse
Affiliation(s)
| | | | | | | | - Michela Colombo
- Department of Health Sciences, Universita' degli Studi di Milano, Milano, Italy
| | - Caroline Chapman
- Bowel Cancer Screening Programme, Eastern Hub Queens Medical Centre, Nottingham University Hospitals, Nottingham, UK
| | - Fabio Grizzi
- Department of Immunology & Inflammation, Humanitas Clinical & Research Center, Milan, Italy
| | - Federica Patrinicola
- Department of Immunology & Inflammation, Humanitas Clinical & Research Center, Milan, Italy
| | - W Martin Kast
- Departments of Obstetrics & Gynecology and Molecular Microbiology & Immunology, University of Southern California, Los Angeles, CA, USA
| | | | | | - Naval Daver
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Peter Ruvolo
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sean M Post
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Robert S Bresalier
- Department of Gastroenterology, Hepatology and Nutrition, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Maurizio Chiriva-Internati
- Kiromic Inc., Houston, TX, USA.,Department of Lymphoma & Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
12
|
Shallis RM, Terry CM, Lim SH. The multi-faceted potential of CD38 antibody targeting in multiple myeloma. Cancer Immunol Immunother 2017; 66:697-703. [PMID: 28341874 PMCID: PMC11029060 DOI: 10.1007/s00262-017-1990-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2016] [Accepted: 03/16/2017] [Indexed: 10/19/2022]
Abstract
CD38, an adenine dinucleotide phosphate (ADP) ribose cyclase and a cyclic ADP ribose hydrolase, is widely expressed on the surface of multiple myeloma (MM) cells. It is known to play a pivotal role in the downstream pathways that mediate MM cell growth, signal transduction, and adhesion. The clinical use of CD38 monoclonal antibodies (MoAbs), such as daratumumab, either as monotherapy or in combination with other anti-MM agents, has produced impressive results in patients who have failed standard MM therapy. CD38 MoAbs exhibit several cytotoxic mechanisms on MM cells. In addition to the classical effector mechanisms associated with antibody therapy, CD38 MoAbs induce MM apoptosis and clonal T-cell expansion. Here, we summarize the results of some pivotal clinical studies using a human CD38 MoAb, daratumumab, in patients with MM, discuss the anti-MM effector mechanisms induced by CD38 MoAbs, and review the potential tumor antigens that may be suitable targets for immunotherapy of MM. Finally, we present a paradigm of immunotherapy for MM patients using CD38 MoAbs followed by GM-CSF and an immune checkpoint inhibitor in patients who have undergone high dose chemotherapy and autologous stem cell transplant. CD38 MoAbs have emerged as a novel and ultimately very promising immunotherapeutic agent for MM because of its ability to induce MM cytotoxicity through both arms of the adaptive immune responses.
Collapse
Affiliation(s)
- Rory M Shallis
- Division of Hematology and Oncology, Rhode Island Hospital/Brown University Warren Alpert Medical School, Room 140, APC Building, 593 Eddy Street, Providence, RI, 02903, USA
| | - Christopher M Terry
- Division of Hematology and Oncology, Rhode Island Hospital/Brown University Warren Alpert Medical School, Room 140, APC Building, 593 Eddy Street, Providence, RI, 02903, USA
| | - Seah H Lim
- Division of Hematology and Oncology, Rhode Island Hospital/Brown University Warren Alpert Medical School, Room 140, APC Building, 593 Eddy Street, Providence, RI, 02903, USA.
| |
Collapse
|
13
|
Yin K, Liu Y, Chu M, Wang Y. TFDP3 Regulates Epithelial-Mesenchymal Transition in Breast Cancer. PLoS One 2017; 12:e0170573. [PMID: 28114432 PMCID: PMC5256886 DOI: 10.1371/journal.pone.0170573] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Accepted: 01/06/2017] [Indexed: 11/19/2022] Open
Abstract
Breast cancer remains a lethal disease to women due to lymph node metastasis, the tumor microenvironment, secondary resistance and other unknown factors. Several important transcription factors involved in this disease, such as PTEN, p53 and beta-catenin, have been identified and researched in-depth as candidates for targeted therapy in breast cancer. TFDP3 is a new, promising candidate for transcriptional regulation in breast cancer, although it was first identified in hepatocellular carcinoma. Here, we demonstrate that TFDP3 is expressed in a variety of malignancies, normal testis tissue and breast cancer cell lines and thus provide evidence that TFDP3 is a cancer-testis antigen. We illustrate that overexpression or silencing TFDP3 interferes with epithelial-mesenchymal transition but does not influence cell proliferation, indicating that the TFDP3 protein acts as a transcription factor during epithelial-mesenchymal transition. These data highlight that TFDP3 is expressed in breast cancer, that it is a member of the cancer-testis antigen family and that it functions as a regulator in epithelial-mesenchymal transition.
Collapse
Affiliation(s)
- Kailin Yin
- Department of Immunology, School of Basic Medical Science, Peking University, Beijing, China
- * E-mail: (KLY); (YDW)
| | - Yanchen Liu
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Liaoning, China
| | - Ming Chu
- Department of Immunology, School of Basic Medical Science, Peking University, Beijing, China
| | - Yuedan Wang
- Department of Immunology, School of Basic Medical Science, Peking University, Beijing, China
- * E-mail: (KLY); (YDW)
| |
Collapse
|
14
|
Salmaninejad A, Zamani MR, Pourvahedi M, Golchehre Z, Hosseini Bereshneh A, Rezaei N. Cancer/Testis Antigens: Expression, Regulation, Tumor Invasion, and Use in Immunotherapy of Cancers. Immunol Invest 2016; 45:619-40. [DOI: 10.1080/08820139.2016.1197241] [Citation(s) in RCA: 101] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
15
|
Amunjela JN, Tucker SJ. POPDC proteins as potential novel therapeutic targets in cancer. Drug Discov Today 2016; 21:1920-1927. [PMID: 27458118 DOI: 10.1016/j.drudis.2016.07.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Revised: 07/10/2016] [Accepted: 07/18/2016] [Indexed: 02/08/2023]
Abstract
Popeye domain-containing (POPDC) proteins are a novel class of cAMP-binding molecules that affect cancer cell behaviour and correlate with poor clinical outcomes. They are encoded by the POPDC genes POPDC1, POPDC2, and POPDC3. The deletion of POPDC genes and the suppression of POPDC proteins correlate with enhanced cancer cell proliferation, migration, invasion, metastasis, drug resistance, and poor patient survival in various human cancers. Overexpression of POPDC proteins inhibits cancer cell migration and invasion in vitro. POPDC proteins present promising anticancer therapeutic targets and here we review their roles in promoting cancer progression and highlight their potential as anticancer therapeutic targets.
Collapse
Affiliation(s)
- Johanna N Amunjela
- Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK
| | - Steven J Tucker
- Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK.
| |
Collapse
|
16
|
Xiang SD, Gao Q, Wilson KL, Heyerick A, Plebanski M. Mapping T and B cell epitopes in sperm protein 17 to support the development of an ovarian cancer vaccine. Vaccine 2015; 33:5950-9. [PMID: 26263201 DOI: 10.1016/j.vaccine.2015.07.094] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Revised: 07/13/2015] [Accepted: 07/23/2015] [Indexed: 11/29/2022]
Abstract
Ovarian cancer (OC) is the seventh most common cancer in women worldwide, and the leading cause of death from gynaecological malignancy. Immunotherapeutic strategies including cancer vaccines are considered less toxic and more specific than current treatments. Sperm surface protein (Sp17) is a protein aberrantly expressed in primary as well as in metastatic lesions in >83% of ovarian cancer patients. Vaccines based on the Sp17 protein are immunogenic and protective in animal models. To map the immunogenic regions and support the development of human Sp17 peptide based vaccines, we used 6 overlapping peptides of the human Sp17 sequence adjuvanted with CpG to immunise humanised HLA-A2.1 transgenic C57BL/6 mice, and assessed immunogenicity by ELISPOT and ELISA. No CD8 T cells were found to be induced to a comprehensive panel of 10 HLA-A2.1 or H-2K(b) binding predicted epitopes. However, one of the 6 peptides, hSp17111-142, induced high levels of antibodies and IFN-γ producing T cells (but not IL-17 or IL-4) both in C57BL/6 and in C57BL/6-HLA-A2.1 transgenic mice. C57BL/6 mice immunised with CpG adjuvanted hSp17111-142 significantly prolonged the life-span of the mice bearing the ovarian carcinoma ID8 cell line. We further mapped the immuno-dominant B and T cell epitope regions within hSp17111-142 using ELISPOT and competition ELISA. Herein, we report the identification of a single immuno-dominant B cell (134-142 aa) epitope and 2 T helper 1 (Th1) cell epitopes (111-124 aa and 124-138 aa). These result together support further exploration of hSp17111-142 peptide formulations as vaccines against ovarian cancer.
Collapse
Affiliation(s)
- Sue D Xiang
- Department of Immunology, Central Clinical School, Faculty of Medicine, Nursing and Health Sciences, 89 Commercial Rd, Melbourne 3004, VIC, Australia.
| | - Qian Gao
- Department of Immunology, Central Clinical School, Faculty of Medicine, Nursing and Health Sciences, 89 Commercial Rd, Melbourne 3004, VIC, Australia.
| | - Kirsty L Wilson
- Department of Immunology, Central Clinical School, Faculty of Medicine, Nursing and Health Sciences, 89 Commercial Rd, Melbourne 3004, VIC, Australia.
| | - Arne Heyerick
- PX Biosolutions Pty Ltd, PO Box 290, South Melbourne 3205, VIC, Australia.
| | - Magdalena Plebanski
- Department of Immunology, Central Clinical School, Faculty of Medicine, Nursing and Health Sciences, 89 Commercial Rd, Melbourne 3004, VIC, Australia.
| |
Collapse
|
17
|
Abstract
Multiple myeloma (MM) is a B-cell malignancy characterized by the clonal proliferation of malignant plasma cells in the bone marrow and the development of osteolytic bone lesions. MM has emerged as a paradigm within the cancers for the success of drug discovery and translational medicine. This article discusses immunotherapy as an encouraging option for the goal of inducing effective and long-lasting therapeutic outcome. Divided into two distinct approaches, passive or active, immunotherapy, which targets tumor-associated antigens has shown promising results in multiple preclinical and clinical studies.
Collapse
Affiliation(s)
- Jooeun Bae
- Dana-Farber Cancer Institute, Harvard Medical School, 450 Brookline Avenue, Boston, MA 02215, USA.
| | - Nikhil C Munshi
- Dana-Farber Cancer Institute, Harvard Medical School, 450 Brookline Avenue, Boston, MA 02215, USA
| | - Kenneth C Anderson
- Dana-Farber Cancer Institute, Harvard Medical School, 450 Brookline Avenue, Boston, MA 02215, USA
| |
Collapse
|
18
|
Grizzi F, Mirandola L, Qehajaj D, Cobos E, Figueroa JA, Chiriva-Internati M. Cancer-Testis Antigens and Immunotherapy in the Light of Cancer Complexity. Int Rev Immunol 2015; 34:143-53. [PMID: 25901859 DOI: 10.3109/08830185.2015.1018418] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
19
|
Figueroa JA, Reidy A, Mirandola L, Trotter K, Suvorava N, Figueroa A, Konala V, Aulakh A, Littlefield L, Grizzi F, Rahman RL, R. Jenkins M, Musgrove B, Radhi S, D'Cunha N, D'Cunha LN, Hermonat PL, Cobos E, Chiriva-Internati M. Chimeric Antigen Receptor Engineering: A Right Step in the Evolution of Adoptive Cellular Immunotherapy. Int Rev Immunol 2015; 34:154-87. [PMID: 25901860 DOI: 10.3109/08830185.2015.1018419] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
20
|
Hoang MD, Jung SH, Lee HJ, Lee YK, Nguyen-Pham TN, Choi NR, Vo MC, Lee SS, Ahn JS, Yang DH, Kim YK, Kim HJ, Lee JJ. Dendritic Cell-Based Cancer Immunotherapy against Multiple Myeloma: From Bench to Clinic. Chonnam Med J 2015; 51:1-7. [PMID: 25914874 PMCID: PMC4406989 DOI: 10.4068/cmj.2015.51.1.1] [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: 03/19/2015] [Revised: 03/19/2015] [Accepted: 03/23/2015] [Indexed: 01/27/2023] Open
Abstract
Although the introduction of stem cell transplantation and novel agents has improved survival, multiple myeloma (MM) is still difficult to cure. Alternative approaches are clearly needed to prolong the survival of patients with MM. Dendritic cell (DC) therapy is a very promising tool immunologically in MM. We developed a method to generate potent DCs with increased Th1 polarization and migration ability for inducing strong myeloma-specific cytotoxic T lymphocytes. In this review, we discuss how the efficacy of cancer immunotherapy using DCs can be improved in MM.
Collapse
Affiliation(s)
- My-Dung Hoang
- Research Center for Cancer Immunotherapy, Chonnam National University Hwasun Hospital, Hwasun, Korea
| | - Sung-Hoon Jung
- Research Center for Cancer Immunotherapy, Chonnam National University Hwasun Hospital, Hwasun, Korea. ; Department of Hematology-Oncology, Chonnam National University Hwasun Hospital, Hwasun, Korea
| | - Hyun-Ju Lee
- Research Center for Cancer Immunotherapy, Chonnam National University Hwasun Hospital, Hwasun, Korea
| | | | - Thanh-Nhan Nguyen-Pham
- Research Center for Cancer Immunotherapy, Chonnam National University Hwasun Hospital, Hwasun, Korea
| | - Nu-Ri Choi
- Research Center for Cancer Immunotherapy, Chonnam National University Hwasun Hospital, Hwasun, Korea
| | - Manh-Cuong Vo
- Research Center for Cancer Immunotherapy, Chonnam National University Hwasun Hospital, Hwasun, Korea
| | - Seung-Shin Lee
- Department of Hematology-Oncology, Chonnam National University Hwasun Hospital, Hwasun, Korea
| | - Jae-Sook Ahn
- Department of Hematology-Oncology, Chonnam National University Hwasun Hospital, Hwasun, Korea
| | - Deok-Hwan Yang
- Department of Hematology-Oncology, Chonnam National University Hwasun Hospital, Hwasun, Korea
| | - Yeo-Kyeoung Kim
- Department of Hematology-Oncology, Chonnam National University Hwasun Hospital, Hwasun, Korea
| | - Hyeoung-Joon Kim
- Department of Hematology-Oncology, Chonnam National University Hwasun Hospital, Hwasun, Korea
| | - Je-Jung Lee
- Research Center for Cancer Immunotherapy, Chonnam National University Hwasun Hospital, Hwasun, Korea. ; Department of Hematology-Oncology, Chonnam National University Hwasun Hospital, Hwasun, Korea. ; Vaxcell-Bio Therapeutics, Hwasun, Korea
| |
Collapse
|
21
|
Grizzi F, Franceschini B, Di Biccari S, Musardo S, Pedretti E, Chiriva-Internati M, Osipov V, Fernández-Aceñero MJ. Sperm protein 17 and AKAP-associated sperm protein cancer/testis antigens are expressed in ciliated hepatic foregut cysts. Histopathology 2015; 67:398-403. [PMID: 25600306 DOI: 10.1111/his.12654] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Accepted: 01/14/2015] [Indexed: 11/30/2022]
Abstract
AIMS Ciliated hepatic foregut cysts (CHFCs) are retained benign lesions of the liver. However, a case of squamous cell metaplasia and five cases of squamous cell carcinoma arising from a CHFC have been described. The potential of malignant transformation makes the identification of new biomarkers necessary. As the cancer/testis antigen sperm protein 17 (Sp17) has been detected in oral and oesophageal squamous cell carcinomas, the aim of this study was to investigate the expression of Sp17 and AKAP-associated sperm protein (ASP), which has a shared N-terminal sequence with Sp17, in four surgically resected CHFCs. METHODS AND RESULTS CHFC specimens were taken from two patients who attended the Medical College of Wisconsin, Milwaukee, USA and two patients who attended the Fundación Jiménez Díaz, Madrid, Spain. CHFCs were found to be immunopositive for Sp17 and ASP. Both proteins were localized to the cytoplasm of ciliated cells lining the cysts, and their cilia. Confocal microscopy demonstrated that Sp17 and ASP overlapped in the same region of the cell. CONCLUSION Sp17 and ASP cancer/testis antigens were found in ciliated cells of four CHFCs. Further characterization of Sp17 and ASP in patients with CHFCs may provide significant clues for understanding the molecular mechanisms underlying their predisposition to develop squamous cell carcinomas.
Collapse
Affiliation(s)
- Fabio Grizzi
- Department of Immunology and Inflammation, Humanitas Clinical and Research Centre, Rozzano, Italy
| | - Barbara Franceschini
- Laboratory of Quantitative Medicine, Humanitas Clinical and Research Centre, Rozzano, Italy
| | - Sonia Di Biccari
- Laboratory of Quantitative Medicine, Humanitas Clinical and Research Centre, Rozzano, Italy
| | - Stefano Musardo
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Elisa Pedretti
- Department of Internal Medicine, Division of Hematology & Oncology, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Maurizio Chiriva-Internati
- Department of Internal Medicine, Division of Hematology & Oncology, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Vladimir Osipov
- Department of Pathology, Medical College of Wisconsin, Milwaukee, WI, USA
| | | |
Collapse
|
22
|
Increased levels of sperm protein 17 mRNA and circulating antibodies in periampullary carcinoma patients. Int J Clin Oncol 2014; 20:736-44. [DOI: 10.1007/s10147-014-0762-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2014] [Accepted: 10/09/2014] [Indexed: 12/13/2022]
|
23
|
Coulie PG, Van den Eynde BJ, van der Bruggen P, Boon T. Tumour antigens recognized by T lymphocytes: at the core of cancer immunotherapy. Nat Rev Cancer 2014; 14:135-46. [PMID: 24457417 DOI: 10.1038/nrc3670] [Citation(s) in RCA: 833] [Impact Index Per Article: 75.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
In this Timeline, we describe the characteristics of tumour antigens that are recognized by spontaneous T cell responses in cancer patients and the paths that led to their identification. We explain on what genetic basis most, but not all, of these antigens are tumour specific: that is, present on tumour cells but not on normal cells. We also discuss how strategies that target these tumour-specific antigens can lead either to tumour-specific or to crossreactive T cell responses, which is an issue that has important safety implications in immunotherapy. These safety issues are even more of a concern for strategies targeting antigens that are not known to induce spontaneous T cell responses in patients.
Collapse
Affiliation(s)
- Pierre G Coulie
- 1] de Duve Institute and the Université catholique de Louvain, B-1200 Brussels, Belgium. [2] WELBIO (Walloon Excellence in Lifesciences and Biotechnology), B-1200 Brussels, Belgium
| | - Benoît J Van den Eynde
- 1] de Duve Institute and the Université catholique de Louvain, B-1200 Brussels, Belgium. [2] Ludwig Institute for Cancer Research, B-1200 Brussels, Belgium. [3] WELBIO (Walloon Excellence in Lifesciences and Biotechnology), B-1200 Brussels, Belgium
| | - Pierre van der Bruggen
- 1] de Duve Institute and the Université catholique de Louvain, B-1200 Brussels, Belgium. [2] Ludwig Institute for Cancer Research, B-1200 Brussels, Belgium. [3] WELBIO (Walloon Excellence in Lifesciences and Biotechnology), B-1200 Brussels, Belgium
| | - Thierry Boon
- 1] de Duve Institute and the Université catholique de Louvain, B-1200 Brussels, Belgium. [2] Ludwig Institute for Cancer Research, B-1200 Brussels, Belgium
| |
Collapse
|
24
|
Rosenblatt J, Bar-Natan M, Munshi NC, Avigan DE. Immunotherapy for multiple myeloma. Expert Rev Hematol 2014; 7:91-6. [DOI: 10.1586/17474086.2014.878226] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
25
|
Avigan D, Hari P, Battiwalla M, Bishop MR, Giralt SA, Hardy NM, Kröger N, Wayne AS, Hsu KC. Proceedings from the National Cancer Institute's Second International Workshop on the Biology, Prevention, and Treatment of Relapse after Hematopoietic Stem Cell Transplantation: part II. Autologous Transplantation-novel agents and immunomodulatory strategies. Biol Blood Marrow Transplant 2013; 19:1661-9. [PMID: 24018393 PMCID: PMC3914636 DOI: 10.1016/j.bbmt.2013.08.011] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2013] [Accepted: 08/30/2013] [Indexed: 12/11/2022]
Abstract
In the National Cancer Institute's Second International Workshop on the Biology, Prevention, and Treatment of Relapse after Hematopoietic Stem Cell Transplantation, the Scientific/Educational Session on Autologous Transplantation addressed the role of novel agents and immunomodulatory strategies in management of relapse after autologous hematopoietic stem cell transplantation (AHSCT). Concepts were illustrated through in-depth discussion of multiple myeloma, with broader discussion of areas relevant for relapse of other malignancies as well as in the setting of allogeneic transplantation. Dr. Hari provided an overview of the epidemiology of relapse after AHSCT in multiple myeloma, addressing clinical patterns, management implications, and treatment options at relapse, highlighting the implications of novel therapeutic agents in initial, maintenance, and relapse treatment. Dr. Avigan discussed current concepts in tumor vaccine design, including whole cell and antigen-specific strategies, use of an AHSCT platform to reverse tumor-associated immunosuppression and tolerance, and combining vaccines with immunomodulatory agents to promote establishment of durable antitumor immunity. Dr. Hsu reviewed the immunogenetics of natural killer (NK) cells and general NK biology, the clinical importance of autologous NK activity (eg, lymphoma and neuroblastoma), the impact of existing therapies on promotion of NK cell activity (eg, immunomodulatory drugs, monoclonal antibodies), and strategies for enhancing autologous and allogeneic NK cell effects through NK cell gene profiling.
Collapse
Affiliation(s)
- David Avigan
- Division of Hematology Oncology, Hematologic Malignancies/Bone Marrow Transplant Program, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Song JX, Li FQ, Cao WL, Jia X, Shi LN, Lu JF, Ma CF, Kong QQ. Anti-Sp17 monoclonal antibody–doxorubicin conjugates as molecularly targeted chemotherapy for ovarian carcinoma. Target Oncol 2013; 9:263-72. [DOI: 10.1007/s11523-013-0293-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2013] [Accepted: 07/15/2013] [Indexed: 10/26/2022]
|
27
|
Arnason J, Avigan D. Evolution of cellular immunotherapy: from allogeneic transplant to dendritic cell vaccination as treatment for multiple myeloma. Immunotherapy 2013; 4:1043-51. [PMID: 23148756 DOI: 10.2217/imt.12.118] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The promise of cellular therapy as treatment for multiple myeloma is highlighted by the observation that allogeneic transplantation results in durable remissions in a subset of patients. The potency of the graft-versus-myeloma effect is supported by the decreased risk of relapse seen in patients with graft-versus-host disease and disease response following donor lymphocyte infusions. However, the lack of specificity of the alloreactive lymphocytes limits their therapeutic efficacy and results in significant treatment-related morbidity and mortality. A major area of investigation is the development of cancer vaccines to generate myeloma-specific immunity that selectively targets malignant cells while minimizing toxicity to normal tissues. Critical elements required to develop an effective vaccine strategy involve the identification of myeloma-associated antigens, enhancement of antigen presentation, and reversing the immunosuppressive milieu induced by the disease. Dendritic cells are potent APCs that represent an ideal platform for vaccination. Strategies for vaccine design include the loading of individual antigens as well as the use of whole tumor cells as a source of myeloma antigens. Vaccination has been examined in the postautologous transplant setting in which disease cytoreduction and depletion of Tregs is associated with enhanced vaccine response. Recent efforts have also included exploration of immune modulatory agents that target inhibitory pathways to enhance vaccine response and create a more durable antitumor immunity.
Collapse
Affiliation(s)
- Jon Arnason
- Beth Israel Deaconess Medical Center, Hematologic Malignancies & Bone Marrow Transplantation Program, Harvard Medical School, MA, USA
| | | |
Collapse
|
28
|
Grizzi F, Chiriva-Internati M. Translating sperm protein 17 as a target for immunotherapy from the bench to the bedside in the light of cancer complexity. TISSUE ANTIGENS 2013; 81:116-118. [PMID: 23330722 DOI: 10.1111/tan.12052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2012] [Accepted: 12/16/2012] [Indexed: 06/01/2023]
|
29
|
Gjerstorff MF, Ditzel HJ. Limited SP17 expression within tumors diminishes its therapeutic potential. ACTA ACUST UNITED AC 2012; 80:523-7. [DOI: 10.1111/tan.12015] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Affiliation(s)
- M. F. Gjerstorff
- Department of Cancer and Inflammation Research, Institute for Molecular Medicine (IMM); University of Southern Denmark; Odense; Denmark
| | | |
Collapse
|
30
|
Novel strategies for immunotherapy in multiple myeloma: previous experience and future directions. Clin Dev Immunol 2012; 2012:753407. [PMID: 22649466 PMCID: PMC3357929 DOI: 10.1155/2012/753407] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2011] [Accepted: 02/27/2012] [Indexed: 12/28/2022]
Abstract
Multiple myeloma (MM) is a life-threatening haematological malignancy for which standard therapy is inadequate. Autologous stem cell transplantation is a relatively effective treatment, but residual malignant sites may cause relapse. Allogeneic transplantation may result in durable responses due to antitumour immunity mediated by donor lymphocytes. However, morbidity and mortality related to graft-versus-host disease remain a challenge. Recent advances in understanding the interaction between the immune system of the patient and the malignant cells are influencing the design of clinically more efficient study protocols for MM.
Cellular immunotherapy using specific antigen-presenting cells (APCs), to overcome aspects of immune incompetence in MM patients, has received great attention, and numerous clinical trials have evaluated the potential for dendritic cell (DC) vaccines as a novel immunotherapeutic approach. This paper will summarize the data investigating aspects of immunity concerning MM, immunotherapy for patients with MM, and strategies, on the way, to target the plasma cell more selectively. We also include the MM antigens and their specific antibodies that are of potential use for MM humoral immunotherapy, because they have demonstrated the most promising preclinical results.
Collapse
|
31
|
Immunotherapy using dendritic cells against multiple myeloma: how to improve? Clin Dev Immunol 2012; 2012:397648. [PMID: 22481968 PMCID: PMC3312256 DOI: 10.1155/2012/397648] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2011] [Accepted: 01/02/2012] [Indexed: 01/10/2023]
Abstract
Multiple myeloma (MM) is a good target disease in which one can apply cellular immunotherapy, which is based on the graft-versus-myeloma effect. This role of immune effector cells provides the framework for the development of immune-based therapeutic options that use antigen-presenting cells (APCs) with increased potency, such as dendritic cells (DCs), in MM. Current isolated idiotype (Id), myeloma cell lysates, myeloma dying cells, DC-myeloma hybrids, or DC transfected with tumor-derived RNA has been used for immunotherapy with DCs. Immunological inhibitory cytokines, such as TGF-β, IL-10, IL-6 and VEGF, which are produced from myeloma cells, can modulate antitumor host immune response, including the abrogation of DC function, by constitutive activation of STAT3. Therefore, even the immune responses have been observed in clinical trials, the clinical response was rarely improved following DC vaccinations in MM patients. We are going to discuss how to improve the efficacy of DC vaccination in MM.
Collapse
|
32
|
Grizzi F, Di Ieva A, Di Biccari S, Ceva-Grimaldi G, Colombo P, Tschabitscher M. Sperm Protein 17: Is It a Useful Target Antigen in Human Pituitary Adenomas? PROCEDIA IN VACCINOLOGY 2012; 6:39-46. [DOI: 10.1016/j.provac.2012.04.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/28/2023]
|
33
|
Chiriva-Internati M, Yu Y, Mirandola L, D'Cunha N, Hardwicke F, Cannon MJ, Cobos E, Kast WM. Identification of AKAP-4 as a new cancer/testis antigen for detection and immunotherapy of prostate cancer. Prostate 2012; 72:12-23. [PMID: 21520158 DOI: 10.1002/pros.21400] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2010] [Accepted: 03/16/2011] [Indexed: 12/27/2022]
Abstract
BACKGROUND Prostate cancer (PC) is the second most common cancer in older men, after skin cancer. PC is difficult to diagnose because the prostate-specific antigen screening method is associated with many false positives. In addition there is a need to develop new and more effective treatments. Among presently available new treatments, immunotherapy is a promising approach. We investigated the expression of the cancer/testis antigen, AKAP-4, in PC patients to evaluate the possibility of exploiting AKAP-4 as a target for immunotherapy. METHODS We analyzed normal prostate tissues, 15 patients with PC and the LnCAP PC cell line by immunohistochemistry. We tested AKAP-4 immunogenicity through indirect ELISA on sera from patients and healthy subjects, and we generated in vitro AKAP-4-specific cytotoxic lymphocytes from peripheral blood mononuclear cells. RESULTS AKAP-4 was shown both at the cytoplasmic and surface levels of the LnCAP PC cell line. AKAP-4 was also highly expressed in PC cells from patients. We detected specific anti-AKAP-4 circulating immunoglobulins in AKAP-4 positive subjects. Using recombinant AKAP-4 loaded autologous dendritic cells, we generated AKAP-4-specific and HLA-I-restricted cytotoxic T lymphocytes able to kill PC cells in vitro. Further characterization indicated a Th-1 skewing in the cytokine secretion profile of these cells. CONCLUSIONS We demonstrate the aberrant expression of AKAP-4 in PC, which will potentially be developed as a biomarker in PC. We provide evidence that AKAP-4 is a potential target for PC adoptive immunotherapy or anti-tumor vaccination.
Collapse
Affiliation(s)
- Maurizio Chiriva-Internati
- Division of Hematology & Oncology, Texas Tech University Health Sciences Center and The Southwest Cancer Treatment and Research Center, Lubbock, Texas 79430, USA.
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Song JX, Cao WL, Li FQ, Shi LN, Jia X. Anti-Sp17 monoclonal antibody with antibody-dependent cell-mediated cytotoxicity and complement-dependent cytotoxicity activities against human ovarian cancer cells. Med Oncol 2011; 29:2923-31. [PMID: 22198696 DOI: 10.1007/s12032-011-0137-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2011] [Accepted: 12/09/2011] [Indexed: 01/25/2023]
Abstract
Sperm protein 17 (Sp17) is a cancer testis antigen that has been shown to be overexpressed in a variety of gynecologic malignancies, in particular ovarian cancer. Emerging evidences indicate that Sp17 is involved in tumorigenesis and in the migration of malignant cells. It has been proposed as a useful target for tumor-vaccine strategies and a novel marker to define tumor subsets and predict drug response. However, the antitumor activity of anti-Sp17 monoclonal antibody (anti-Sp17 mAb) has not been investigated. In this study, the in vitro cytotoxicity, antibody-dependent cell-mediated cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC) activities of anti-Sp17 mAb were evaluated using Sp17-positive ovarian cancer cells as targets, Sp17-negative ovarian cancer cells as the control, and healthy human peripheral blood monocytes and healthy human serum as effectors. Our preliminary results indicate that the direct cytotoxicity of anti-Sp17 mAb against the investigated ovarian cancer cells was very weak. However, the cytotoxicity of anti-Sp17 mAb, mediated by peripheral blood mononuclear cells (PBMCs), as ADCC, or by human serum, as CDC, was relatively strong in the Sp17-positive ovarian cancer cells. This finding suggested that anti-Sp17 mAb could be a useful tool against ovarian cancer and may provide insight into the development of low side-effect targeting therapy for this malignant disease.
Collapse
Affiliation(s)
- Jia-xi Song
- Laboratory of Molecular Biology, Institute of Medical Laboratory Sciences, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, 210002, People's Republic of China.
| | | | | | | | | |
Collapse
|
35
|
Mirandola L, Yu Y, Jenkins MR, Chiaramonte R, Cobos E, John CM, Chiriva-Internati M. Tracking human multiple myeloma xenografts in NOD-Rag-1/IL-2 receptor gamma chain-null mice with the novel biomarker AKAP-4. BMC Cancer 2011; 11:394. [PMID: 21923911 PMCID: PMC3189930 DOI: 10.1186/1471-2407-11-394] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2011] [Accepted: 09/16/2011] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Multiple myeloma (MM) is a fatal malignancy ranking second in prevalence among hematological tumors. Continuous efforts are being made to develop innovative and more effective treatments. The preclinical evaluation of new therapies relies on the use of murine models of the disease. METHODS Here we describe a new MM animal model in NOD-Rag1null IL2rgnull (NRG) mice that supports the engraftment of cell lines and primary MM cells that can be tracked with the tumor antigen, AKAP-4. RESULTS Human MM cell lines, U266 and H929, and primary MM cells were successfully engrafted in NRG mice after intravenous administration, and were found in the bone marrow, blood and spleen of tumor-challenged animals. The AKAP-4 expression pattern was similar to that of known MM markers, such as paraproteins, CD38 and CD45. CONCLUSIONS We developed for the first time a murine model allowing for the growth of both MM cell lines and primary cells in multifocal sites, thus mimicking the disease seen in patients. Additionally, we validated the use of AKAP-4 antigen to track tumor growth in vivo and to specifically identify MM cells in mouse tissues. We expect that our model will significantly improve the pre-clinical evaluation of new anti-myeloma therapies.
Collapse
Affiliation(s)
- Leonardo Mirandola
- Division of Hematology & Oncology, Texas Tech University Health Sciences Center and Southwest Cancer Treatment and Research Center, Lubbock, TX, USA
- The Laura W. Bush Institute for Women's Health and Center for Women's Health and Gender-Based Medicine, Texas Tech University Health Sciences Center, Amarillo, TX, USA
| | - Yuefei Yu
- Division of Hematology & Oncology, Texas Tech University Health Sciences Center and Southwest Cancer Treatment and Research Center, Lubbock, TX, USA
| | - Marjorie R Jenkins
- Division of Hematology & Oncology, Texas Tech University Health Sciences Center and Southwest Cancer Treatment and Research Center, Lubbock, TX, USA
- The Laura W. Bush Institute for Women's Health and Center for Women's Health and Gender-Based Medicine, Texas Tech University Health Sciences Center, Amarillo, TX, USA
- Departments of Internal Medicine and Obstetrics & Gynecology, Texas Tech University Health Sciences Center, Amarillo, TX, USA
| | - Raffaella Chiaramonte
- Division of Hematology & Oncology, Texas Tech University Health Sciences Center and Southwest Cancer Treatment and Research Center, Lubbock, TX, USA
- Department of Medicine, Surgery and Dentistry, Università degli Studi di Milano, Milano, Italy
| | - Everardo Cobos
- Division of Hematology & Oncology, Texas Tech University Health Sciences Center and Southwest Cancer Treatment and Research Center, Lubbock, TX, USA
- The Laura W. Bush Institute for Women's Health and Center for Women's Health and Gender-Based Medicine, Texas Tech University Health Sciences Center, Amarillo, TX, USA
| | | | - Maurizio Chiriva-Internati
- Division of Hematology & Oncology, Texas Tech University Health Sciences Center and Southwest Cancer Treatment and Research Center, Lubbock, TX, USA
- The Laura W. Bush Institute for Women's Health and Center for Women's Health and Gender-Based Medicine, Texas Tech University Health Sciences Center, Amarillo, TX, USA
| |
Collapse
|
36
|
PD-1 blockade by CT-011, anti-PD-1 antibody, enhances ex vivo T-cell responses to autologous dendritic cell/myeloma fusion vaccine. J Immunother 2011; 34:409-18. [PMID: 21577144 DOI: 10.1097/cji.0b013e31821ca6ce] [Citation(s) in RCA: 231] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
We have developed a cancer vaccine in which autologous tumor is fused with dendritic cells (DCs) resulting in the presentation of tumor antigens in the context of DC-mediated costimulation. In clinical trials, immunologic responses have been observed, however responses may be muted by inhibitory pathways. The PD1/PDL1 pathway is an important element contributing to tumor-mediated immune suppression. In this study, we demonstrate that myeloma cells and DC/tumor fusions strongly express PD-L1. Compared with a control population of normal volunteers, increased PD-1 expression was observed on T cells isolated from patients with myeloma. It is interesting to note that after autologous transplantation, T-cell expression of PD-1 returned to levels seen in normal controls. We examined the effect of PD-1 blockade on T-cell response to DC/tumor fusions ex vivo. Presence of CT-011, an anti-PD1 antibody, promoted the vaccine-induced T-cell polarization towards an activated phenotype expressing Th1 compared with Th2 cytokines. A concomitant decrease in regulatory T cells and enhanced killing in a cytotoxicity assay was observed. In summary, we demonstrate that PD-1 expression is increased in T cells of patients with active myeloma, and that CT-011 enhances activated T-cell responses after DC/tumor fusion stimulation.
Collapse
|
37
|
Mirandola L, J Cannon M, Cobos E, Bernardini G, Jenkins MR, Kast WM, Chiriva-Internati M. Cancer testis antigens: novel biomarkers and targetable proteins for ovarian cancer. Int Rev Immunol 2011; 30:127-37. [PMID: 21557639 DOI: 10.3109/08830185.2011.572504] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Ovarian cancer is the fifth leading cause of cancer death in women and the leading cause from gynecological malignancies. Despite the recently improved outcomes of new chemotherapeutical agents in the therapy of ovarian cancer and the increased 5-year survival rate, the mortality of this malignancy disease remains unchanged. Ovarian cancer therapy is often correlated to the stage of the tumor, but the first step is usually surgical treatment. Afterward, various courses of chemotherapy and radiation are suggested. Obviously, the higher the developmental stage of the tumor, the less the probability is in eradicating it surgically, especially in relation to metastasis. It is clear that an early diagnosis of ovarian cancer is important for the survival of these patients. In order to identify ovarian cancer patients in the early stages, a number of studies are focusing on a particular class of antigens called cancer testis antigens. These antigens display high expression in tumors of different histology, but are normally restricted to the testis and have low or no expression in normal tissues. The testes are an immunologically-privileged site due to the presence of tight junctions between adjacent Sertoli cells that constitute the blood-testis barrier, which prevents auto-immune reactions. In the past few years, some of these antigens were demonstrated to be very promising for the early diagnosis and development of vaccines for ovarian cancer. This review aims to underline the most reliable cancer testis antigens under investigation at this moment.
Collapse
Affiliation(s)
- Leonardo Mirandola
- Division of Hematology & Oncology and Texas Tech University Health Sciences Center, Lubbock, Texas 79430, USA
| | | | | | | | | | | | | |
Collapse
|
38
|
Cheng YH, Wong EW, Cheng CY. Cancer/testis (CT) antigens, carcinogenesis and spermatogenesis. SPERMATOGENESIS 2011; 1:209-220. [PMID: 22319669 PMCID: PMC3271663 DOI: 10.4161/spmg.1.3.17990] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2011] [Revised: 09/01/2011] [Accepted: 09/05/2011] [Indexed: 02/07/2023]
Abstract
During spermatogenesis, spermatogonial stem cells, undifferentiated and differentiated spermatogonia, spermatocytes, spermatids and spermatozoa all express specific antigens, yet the functions of many of these antigens remain unexplored. Studies in the past three decades have shown that many of these transiently expressed genes in developing germ cells are proto-oncogenes and oncogenes, which are expressed only in the testis and various types of cancers in humans and rodents. As such, these antigens are designated cancer/testis antigens (CT antigens). Since the early 1980s, about 70 families of CT antigens have been identified with over 140 members are known to date. Due to their restricted expression in the testis and in various tumors in humans, they have been used as the target of immunotherapy. Multiple clinical trials at different phases are now being conducted with some promising results. Interestingly, in a significant number of cancer patients, antibodies against some of these CT antigens were detected in their sera. However, antibodies against these CT antigens in humans under normal physiological conditions have yet to be reported even though many of these antigens are residing outside of the blood-testis barrier (BTB), such as in the basal compartment of the seminiferous epithelium and in the stem cell niche in the testis. In this review, we summarize latest findings in the field regarding several selected CT antigens which may be intimately related to spermatogenesis due to their unusual restricted expression during different discrete events of spermatogenesis, such as cell cycle progression, meiosis and spermiogenesis. This information should be helpful to investigators in the field to study the roles of these oncogenes in spermatogenesis.
Collapse
Affiliation(s)
- Yan-Ho Cheng
- Center for Biomedical Research; The Population Council; New York, NY USA
- Richmond University Medical Center; Staten Island, NY USA
| | - Elissa Wp Wong
- Center for Biomedical Research; The Population Council; New York, NY USA
| | - C Yan Cheng
- Center for Biomedical Research; The Population Council; New York, NY USA
| |
Collapse
|
39
|
Chiriva-Internati M. Sperm Protein 17: Clinical Relevance of a Cancer/Testis Antigen, from Contraception to Cancer Immunotherapy, and Beyond. Int Rev Immunol 2011; 30:138-49. [DOI: 10.3109/08830185.2011.569903] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
40
|
In vivo molecular targeting effects of anti-Sp17- ICG-Der-02 on hepatocellular carcinoma evaluated by an optical imaging system. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2011; 30:25. [PMID: 21366930 PMCID: PMC3062613 DOI: 10.1186/1756-9966-30-25] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/10/2010] [Accepted: 03/03/2011] [Indexed: 01/22/2023]
Abstract
Background As the expression of human sperm protein 17 (Sp17) in normal tissue is limited and the function is obscure, its aberrant expression in malignant tumors makes it to be a candidated molecular marker for tumor imaging diagnosis and targeting therapy of the diseases.The aim of this research is to evaluate the targeting effects of anti-sperm protein 17 monoclonal antibody (anti-Sp17) on cancer in vivo and investigate its usefulness as a reagent for molecular imaging diagnosis. Methods Immunohistochemistry was used to identify the expression of Sp17 in a hepatocellular carcinoma cell line and tumor xenograft specimens. A near infrared fluorescence dye, ICG-Der-02, was covalently linked to anti-Sp17 for in vivo imaging. The immuno-activity of the anti-Sp17-ICG-Der-02 complex was tested in vitro by ELISA; it was then injected into tumor-bearing nude mice through the caudal vein to evaluate its tumor targeting effect by near infrared imaging system. Results Overexpression of Sp17 on the surface of the hepatocellular carcinoma cell line SMMC-7721 was demonstrated. Anti-Sp17-ICG-Der-02 with immuno-activity was successfully synthesized. The immuno-activity and photo stability of anti-Sp17- ICG-Der-02 showed good targeting capability for Sp17 expressing tumor models (SMMC-7721) in vivo, and its accumulation in the tumor lasted for at least 7 days. Conclusions Anti-Sp17 antibody targeted and accumulated in Sp17 positive tumors in vivo, which demonstrated its capability of serving as a diagnostic reagent.
Collapse
|
41
|
Abstract
Multiple myeloma is still a fatal disease. Despite advances in high-dose chemotherapy and stem-cell transplantation and the development of novel therapeutics, relapse of the underlying disease remains the primary cause of treatment failure. Strategies for posttransplantation immunomodulation are desirable for eradication of remaining tumor cells. To this end, immunotherapy aimed at inducing myeloma-specific immunity in patients has been explored. Idiotype protein, secreted by myeloma cells, has been the primary target for immunotherapy as it is the best defined tumor-specific antigen. This chapter focuses on novel immunotherapies that are being developed to treat patients with myeloma. I will discuss potential myeloma antigens, antigen-specific T cells, and their function on myeloma tumor cells, and T-cell-based and antibody-based immunotherapies for myeloma. Furthermore, clinical studies of T-cell-based immunotherapy in the form of vaccination, allogeneic stem-cell transplantation and donor lymphocyte infusions, with or without donor vaccination using patient-derived idiotype, and future application of donor-derived or patient-derived, antigen-specific T-cell infusion in this disease are also discussed. Based on the specificity of the immune effector molecules and cells, immunotherapies with specific T cells or therapeutic antibodies may represent novel strategies for the treatment of multiple myeloma in the near future.
Collapse
|
42
|
Li FQ, Liu Q, Han YL, Wu B, Yin HL. Sperm protein 17 is highly expressed in endometrial and cervical cancers. BMC Cancer 2010; 10:429. [PMID: 20712874 PMCID: PMC2931487 DOI: 10.1186/1471-2407-10-429] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2010] [Accepted: 08/16/2010] [Indexed: 12/13/2022] Open
Abstract
Background Sperm protein 17 (Sp17) is a highly conserved mammalian protein in the testis and spermatozoa and has been characterized as a tumor-associated antigen in a variety of human malignancies. Many studies have examined the role of Sp17 in tumorigenesis and the migration of malignant cells. It has been proposed as a useful target for tumor-vaccine strategies and a novel marker to define tumor subsets and predict drug response. This study aimed to investigate the expression of Sp17 in endometrial and cervical cancer specimens, its possible correlation with the pathological characteristics, and its value in the diagnosis and immunotherapy of the related cancers. Methods The monoclonal antibodies against human Sp17 were produced as reagents for the analysis and immunohistochemistry was used to study two major kinds of paraffin-embedded gynecological cancer specimens, including 50 cases of endometrial cancer (44 adenous and 6 adenosquamous) and 31 cases of cervical cancer (15 adenous and 16 squamous). Normal peripheral endometrial and cervical tissues were used as controls. Results Sp17 was found in 66% (33/50) of the patients with endometrial cancer and 61% (19/31) of those with cervical cancer. Its expression was found in a heterogeneous pattern in the cancer tissues. The expression was not correlated with the histological subtype and grade of malignancy, but the staining patterns were different in endometrial and cervical cancers. The hyperplastic glands were positive for Sp17 in the normal peripheral endometrial and cervical tissues in 10% (8/81) of the patients. Conclusions Sp17 is highly expressed in human endometrial and cervical cancers in a heterogeneous pattern. Although the expression frequency of Sp17 is not correlated with the histological subtype, the staining pattern may help to define endometrial and cervical cancers. Sp17 targeted immunotherapy of tumors needs more accurate validation.
Collapse
Affiliation(s)
- Fang-Qiu Li
- Laboratory of Molecular Biology, Institute of Medical Laboratory Sciences, Jinling Hospital, School of Medicine, Nanjing University, Nanjing 210002, China.
| | | | | | | | | |
Collapse
|
43
|
Kausar T, Ahsan A, Hasan MR, Lin L, Beer DG, Ralhan R. Sperm protein 17 is a novel marker for predicting cisplatin response in esophageal squamous cancer cell lines. Int J Cancer 2010; 126:1494-503. [PMID: 19685492 DOI: 10.1002/ijc.24828] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Expression of sperm protein 17 (Sp17) mRNA has been reported in various malignancies. In an earlier study, we reported the upregulation of Sp17 transcripts in primary esophageal squamous cell carcinomas (ESCCs) using differential display and detected Sp17 transcripts in 86% of ESCCs by RT-PCR, whereas no transcripts were detected in the paired normal esophageal tissues. Herein we hypothesized that Sp17 might be used as a marker for detecting the response of anticancer therapies in ESCCs. Our results indicated that Sp17 protein levels in esophageal squamous cancer cell lines decreased in response to treatment with (i) the HSP90 activity inhibitor geldanamycin, (ii) the tyrosine kinase inhibitor erlotinib and (iii) cisplatin (chemotherapeutic agent commonly used in management of ESCC). In contrast, the Sp17 levels did not decrease in response to radiation therapy and treatment with the chemotherapeutic agent, gemcitabine. Further investigations showed that cisplatin induced decrease in Sp17 levels was due to transcriptional inhibition and cisplatin-resistant cell lines did not show this decrease in Sp17 levels in response to cisplatin treatment. In addition, we also carried our mass spectophotometric analysis to identify the binding partners of Sp17 to characterize its possible involvement in esophageal tumorigenesis and chemoresistance.
Collapse
Affiliation(s)
- Tasneem Kausar
- Department of Biochemistry, All India Institute of Medical Science, New Delhi, India
| | | | | | | | | | | |
Collapse
|
44
|
Li FQ, Han YL, Liu Q, Wu B, Huang WB, Zeng SY. Overexpression of human sperm protein 17 increases migration and decreases the chemosensitivity of human epithelial ovarian cancer cells. BMC Cancer 2009; 9:323. [PMID: 19744347 PMCID: PMC2753635 DOI: 10.1186/1471-2407-9-323] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2008] [Accepted: 09/11/2009] [Indexed: 11/20/2022] Open
Abstract
Background Most deaths from ovarian cancer are due to metastases that are resistant to conventional therapies. But the factors that regulate the metastatic process and chemoresistance of ovarian cancer are poorly understood. In the current study, we investigated the aberrant expression of human sperm protein 17 (HSp17) in human epithelial ovarian cancer cells and tried to analyze its influences on the cell behaviors like migration and chemoresistance. Methods Immunohistochemistry and immunocytochemistry were used to identify HSp17 in paraffin embedded ovarian malignant tumor specimens and peritoneal metastatic malignant cells. Then we examined the effect of HSp17 overexpression on the proliferation, migration, and chemoresistance of ovarian cancer cells to carboplatin and cisplatin in a human ovarian carcinoma cell line, HO8910. Results We found that HSp17 was aberrantly expressed in 43% (30/70) of the patients with primary epithelial ovarian carcinomas, and in all of the metastatic cancer cells of ascites from 8 patients. The Sp17 expression was also detected in the metastatic lesions the same as in ovarian lesions. None of the 7 non-epithelial tumors primarily developed in the ovaries was immunopositive for HSp17. Overexpression of HSp17 increased the migration but decreased the chemosensitivity of ovarian carcinoma cells to carboplatin and cisplatin. Conclusion HSp17 is aberrantly expressed in a significant proportion of epithelial ovarian carcinomas. Our results strongly suggest that HSp17 plays a role in metastatic disease and resistance of epithelial ovarian carcinoma to chemotherapy.
Collapse
Affiliation(s)
- Fang-Qiu Li
- Laboratory of Molecular Biology, Institute of Medical Laboratory Sciences, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, PR China.
| | | | | | | | | | | |
Collapse
|
45
|
Chiriva-Internati M, Cobos E, Kast WM. Advances in Immunotherapy of Multiple Myeloma: From the Discovery of Tumor-Associated Antigens to Clinical Trials. Int Rev Immunol 2009; 26:197-222. [PMID: 17558744 DOI: 10.1080/08830180701365966] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Tumors aberrantly express tumor-associated antigens that can be specifically recognized by T-cells, thereby providing a scientific rationale for the design and clinical testing of immunotherapeutic strategies targeting these antigens. Multiple myeloma is a fatal hematologic malignancy. Here, we review techniques to discover new tumor-associated antigens in multiple myeloma and the latest immunotherapeutic strategies employed in this disease.
Collapse
Affiliation(s)
- Maurizio Chiriva-Internati
- Department of Microbiology and Immunology and Division of Hematology/Oncology, Texas Tech University Health Sciences Center and Southwest Cancer Treatment and Research Center, Lubbock, Texas 79430, USA.
| | | | | |
Collapse
|
46
|
Ait-Tahar K, Liggins AP, Collins GP, Campbell A, Barnardo M, Lawrie C, Moir D, Hatton C, Banham AH, Pulford K. Cytolytic T-cell response to the PASD1 cancer testis antigen in patients with diffuse large B-cell lymphoma. Br J Haematol 2009; 146:396-407. [DOI: 10.1111/j.1365-2141.2009.07761.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
47
|
Chiriva-Internati M, Gagliano N, Donetti E, Costa F, Grizzi F, Franceschini B, Albani E, Levi-Setti PE, Gioia M, Jenkins M, Cobos E, Kast WM. Sperm protein 17 is expressed in the sperm fibrous sheath. J Transl Med 2009; 7:61. [PMID: 19604394 PMCID: PMC2727497 DOI: 10.1186/1479-5876-7-61] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2008] [Accepted: 07/15/2009] [Indexed: 11/23/2022] Open
Abstract
Background Sperm protein 17 (Sp17) is a highly conserved mammalian protein characterized in rabbit, mouse, monkey, baboon, macaque, human testis and spermatozoa. mRNA encoding Sp17 has been detected in a range of murine and human somatic tissues. It was also recognized in two myeloma cell lines and in neoplastic cells from patients with multiple myeloma and ovarian carcinoma. These data all indicate that Sp17 is widely distributed in humans, expressed not only in germinal cells and in a variety of somatic tissues, but also in neoplastic cells of unrelated origin. Methods Sp17 expression was analyzed by immunocytochemistry and transmission electron microscopy on spermatozoa. Results Here, we demonstrate the ultrastructural localization of human Sp17 throughout the spermatozoa flagellar fibrous sheath, and its presence in spermatozoa during in vitro states from their ejaculation to the oocyte fertilization. Conclusion These findings suggest a possible role of Sp17 in regulating sperm maturation, capacitation, acrosomal reaction and interactions with the oocyte zona pellucida during the fertilization process. Further, the high degree of sequence conservation throughout its N-terminal half, and the presence of an A-kinase anchoring protein (AKAP)-binding motif within this region, suggest that Sp17 might play a regulatory role in a protein kinase A-independent AKAP complex in both germinal and somatic cells.
Collapse
|
48
|
Melan-A/MART1 Analog Peptide Triggers Anti-myeloma T-cells Through Crossreactivity With HM1.24. J Immunother 2009; 32:613-21. [DOI: 10.1097/cji.0b013e3181a95198] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
49
|
Sze DMY, Brown RD, Yuen E, Gibson J, Ho J, Raitakari M, Basten A, Joshua DE, Fazekas de St Groth B. Clonal Cytotoxic T Cells in Myeloma. Leuk Lymphoma 2009; 44:1667-74. [PMID: 14692517 DOI: 10.1080/1042819031000097438] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Multiple myeloma (MM) is a malignant disease characterized by accumulation of morphologically recognizable plasma cells producing immunoglobulin (Ig) in the bone marrow. The occurrence of clonal T cells in MM, as defined by the presence of rearrangements in the T-cell receptor (TCR)-beta chains detected on Southern blotting, is associated with an improved prognosis. This review aims to describe the various ways in which we have demonstrated the presence of such T cell clones, and to describe the phenotype of these cells. Finally, the specificities of these clinically important CD8+ T cell populations will be discussed in the context of immunotherapy.
Collapse
Affiliation(s)
- Daniel M Y Sze
- Institute of Haematology, Royal Prince Alfred Hospital, Camperdown, NSW, Australia.
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Zhang Y, Shahriar M, Zhang J, Ahmed SU, Lim SH. Clofarabine induces hypomethylation of DNA and expression of Cancer-Testis antigens. Leuk Res 2009; 33:1678-83. [PMID: 19427036 DOI: 10.1016/j.leukres.2009.04.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2009] [Revised: 02/22/2009] [Accepted: 04/04/2009] [Indexed: 11/16/2022]
Abstract
In this study, treatment of lymphoid tumor cells with low dose clofarabine upregulated the expression of Sp17 and SPAN-Xb. This was associated with an increase in hypomethylated CpG dinucleotides and a decrease in global DNA methylation, as demonstrated by decreases in the percent of methylated Alu repeats. The most optimal concentration of clofarabine to induce DNA hypomethylation and CT antigen expression was between 1x10(-9) and 1x10(-8)M. Above this, clofarabine resulted in tumor cell growth inhibition and apoptosis. Our results provide the first evidence for the CT antigen-inducing and DNA hypomethylating property of low concentration clofarabine.
Collapse
Affiliation(s)
- Yana Zhang
- Cancer Research Program, Harrington Regional Medical Center Inc., United States
| | | | | | | | | |
Collapse
|