1
|
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
|
2
|
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: 9] [Impact Index Per Article: 3.0] [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
|
3
|
Xie K, Fu C, Wang S, Xu H, Liu S, Shao Y, Gong Z, Wu X, Xu B, Han J, Xu J, Xu P, Jia X, Wu J. Cancer-testis antigens in ovarian cancer: implication for biomarkers and therapeutic targets. J Ovarian Res 2019; 12:1. [PMID: 30609934 PMCID: PMC6318940 DOI: 10.1186/s13048-018-0475-z] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 12/11/2018] [Indexed: 12/21/2022] Open
Abstract
Ovarian cancer remains the most fatal gynecologic malignancy worldwide due to delayed diagnosis as well as recurrence and drug resistance. Thus, the development of new tumor-related molecules with high sensitivity and specificity to replace or supplement existing tools is urgently needed. Cancer-testis antigens (CTAs) are exclusively expressed in normal testis tissues but abundantly found in several types of cancers, including ovarian cancer. Numerous novel CTAs have been identified by high-throughput sequencing techniques, and some aberrantly expressed CTAs are associated with ovarian cancer initiation, clinical outcomes and chemotherapy resistance. More importantly, CTAs are immunogenic and may be novel targets for antigen-specific immunotherapy in ovarian cancer. In this review, we attempt to characterize the expression of candidate CTAs in ovarian cancer and their clinical significance as biomarkers, activation mechanisms, function in malignant phenotypes and applications in immunotherapy.
Collapse
Affiliation(s)
- Kaipeng Xie
- The Affiliated Obstetrics and Gynecology Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Women's Hospital of Nanjing Medical University, Nanjing, 210004, China.
| | - Chenyang Fu
- The Affiliated Obstetrics and Gynecology Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Women's Hospital of Nanjing Medical University, Nanjing, 210004, China
| | - Suli Wang
- The Affiliated Obstetrics and Gynecology Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Women's Hospital of Nanjing Medical University, Nanjing, 210004, China
| | - Hanzi Xu
- Jiangsu Institute of Cancer Research The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, 210009, China
| | - Siyu Liu
- The Affiliated Obstetrics and Gynecology Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Women's Hospital of Nanjing Medical University, Nanjing, 210004, China
| | - Yang Shao
- The Affiliated Obstetrics and Gynecology Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Women's Hospital of Nanjing Medical University, Nanjing, 210004, China
| | - Zhen Gong
- The Affiliated Obstetrics and Gynecology Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Women's Hospital of Nanjing Medical University, Nanjing, 210004, China
| | - Xiaoli Wu
- The Affiliated Obstetrics and Gynecology Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Women's Hospital of Nanjing Medical University, Nanjing, 210004, China
| | - Bo Xu
- State Key Laboratory of Reproductive Medicine, Institute of Toxicology, Nanjing Medical University, Nanjing, 211166, China
| | - Jing Han
- Jiangsu Institute of Cancer Research The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, 210009, China
| | - Juan Xu
- The Affiliated Obstetrics and Gynecology Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Women's Hospital of Nanjing Medical University, Nanjing, 210004, China
| | - Pengfei Xu
- The Affiliated Obstetrics and Gynecology Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Women's Hospital of Nanjing Medical University, Nanjing, 210004, China
| | - Xuemei Jia
- The Affiliated Obstetrics and Gynecology Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Women's Hospital of Nanjing Medical University, Nanjing, 210004, China.
| | - Jiangping Wu
- The Affiliated Obstetrics and Gynecology Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Women's Hospital of Nanjing Medical University, Nanjing, 210004, China.
| |
Collapse
|
4
|
Gao Q, Xiang SD, Wilson K, Madondo M, Stephens AN, Plebanski M. Sperm Protein 17 Expression by Murine Epithelial Ovarian Cancer Cells and Its Impact on Tumor Progression. Cancers (Basel) 2018; 10:cancers10080276. [PMID: 30127274 PMCID: PMC6115966 DOI: 10.3390/cancers10080276] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 08/13/2018] [Accepted: 08/14/2018] [Indexed: 12/12/2022] Open
Abstract
The cancer testis antigen sperm protein 17 (Sp17) is a promising antigenic target in epithelial ovarian cancer (EOC) vaccine development. However, its role in ovarian cancer is unclear. We isolated and expanded Sp17+ and Sp17− clones from the murine EOC cell line ID8, and compared their in-vitro cell growth characteristics and in-vivo tumorigenicity. We also examined the potential co-expression of molecules that may influence cancer cell survival and interaction with immune cells. These include stimulatory and immunosuppressive molecules, such as major histocompatibility class I molecules (MHC I), MHC II, cytotoxic T lymphocyte associated antigen-4 (CTLA-4), CD73, CD39, tumor necrosis factor receptor II (TNFRII), signal transducer and activator of transcription 3 (STAT3) and programmed death-ligand 1 (PD-L1). Whilst the presence of Sp17 was not correlated with the ID8 cell proliferation/growth capacity in vitro, it was critical to enable progressive tumor formation in vivo. Flow cytometry revealed that Sp17+ ID8 cells displayed higher expression of both STAT3 and PD-L1, whilst MHC II expression was lower. Moreover, Sp17high (PD-L1+MHCII−) cell populations showed significantly enhanced resistance to Paclitaxel-induced cell death in vitro compared to Sp17low (PD-L1−MHCII+) cells, which was associated in turn with increased STAT3 expression. Together, the data support Sp17 as a factor associated with in-vivo tumor progression and chemo-resistance, validating it as a suitable target for vaccine development.
Collapse
Affiliation(s)
- Qian Gao
- Department of Immunology and Pathology, Central Clinical School, Faculty of Medicine, Nursing and Health Science, Monash University, Melbourne, Victoria 3004, Australia.
- Department of Clinical Laboratory, Xiangya Hospital, Central South University, Changsha 410008, China.
| | - Sue D Xiang
- Department of Immunology and Pathology, Central Clinical School, Faculty of Medicine, Nursing and Health Science, Monash University, Melbourne, Victoria 3004, Australia.
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, Victoria 3168, Australia.
- Department of Molecular and Translational Sciences, Monash University, Clayton, Victoria 3168, Australia.
| | - Kirsty Wilson
- Department of Immunology and Pathology, Central Clinical School, Faculty of Medicine, Nursing and Health Science, Monash University, Melbourne, Victoria 3004, Australia.
| | - Mutsa Madondo
- Department of Immunology and Pathology, Central Clinical School, Faculty of Medicine, Nursing and Health Science, Monash University, Melbourne, Victoria 3004, Australia.
| | - Andrew N Stephens
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, Victoria 3168, Australia.
- Department of Molecular and Translational Sciences, Monash University, Clayton, Victoria 3168, Australia.
| | - Magdalena Plebanski
- Department of Immunology and Pathology, Central Clinical School, Faculty of Medicine, Nursing and Health Science, Monash University, Melbourne, Victoria 3004, Australia.
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria 3083, Australia.
| |
Collapse
|
5
|
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
|
6
|
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: 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: 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
|
7
|
Colombo M, Mirandola L, Reidy A, Suvorava N, Konala V, Chiaramonte R, Grizzi F, Rahman RL, Jenkins MR, Nugyen DD, Dalhbeck S, Cobos E, Figueroa JA, Chiriva-Internati M. Targeting Tumor Initiating Cells through Inhibition of Cancer Testis Antigens and Notch Signaling: A Hypothesis. Int Rev Immunol 2016; 34:188-99. [PMID: 25901861 DOI: 10.3109/08830185.2015.1027629] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Tumor initiating cells (TICs) differ from normal stem cells (SCs) in their ability to initiate tumorigenesis, invasive growth, metastasis and the acquisition of chemo and/or radio-resistance. Over the past years, several studies have indicated the potential role of the Notch system as a key regulator of cellular stemness and tumor development. Furthermore, the expression of cancer testis antigens (CTA) in TICs, and their role in SC differentiation and biology, has become an important area of investigation. Here, we propose a model in which CTA expression and Notch signaling interacts to maintain the sustainability of self-replicating tumor populations, ultimately leading to the development of metastasis, drug resistance and cancer progression. We hypothesize that Notch-CTA interactions in TICs offer a novel opportunity for meaningful therapeutic interventions in cancer.
Collapse
Affiliation(s)
- Michela Colombo
- Department of Health Sciences, Università degli Studi di Milano , Milano , Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
8
|
Liu Y, Tian X, Jiang S, Ren X, Liu F, Yang J, Chen Y, Jiang Y. Umbilical cord blood-derived dendritic cells infected by adenovirus for SP17 expression induce antigen-specific cytotoxic T cells against NSCLC cells. Cell Immunol 2015; 298:18-24. [DOI: 10.1016/j.cellimm.2015.08.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Revised: 08/09/2015] [Accepted: 08/17/2015] [Indexed: 12/25/2022]
|
9
|
Tumour antigen expression in hepatocellular carcinoma in a low-endemic western area. Br J Cancer 2015; 112:1911-20. [PMID: 26057582 PMCID: PMC4580401 DOI: 10.1038/bjc.2015.92] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Revised: 01/21/2015] [Accepted: 02/12/2015] [Indexed: 02/06/2023] Open
Abstract
Background: Identification of tumour antigens is crucial for the development of
vaccination strategies against hepatocellular carcinoma (HCC). Most studies
come from eastern-Asia, where hepatitis-B is the main cause of HCC. However,
tumour antigen expression is poorly studied in low-endemic, western areas
where the aetiology of HCC differs. Methods: We constructed tissue microarrays from resected HCC tissue of 133 patients.
Expression of a comprehensive panel of cancer-testis (MAGE-A1,
MAGE-A3/4, MAGE-A10, MAGE-C1, MAGE-C2, NY-ESO-1, SSX-2, sperm protein
17), onco-fetal (AFP, Glypican-3) and overexpressed tumour antigens
(Annexin-A2, Wilms tumor-1, Survivin, Midkine, MUC-1) was determined by
immunohistochemistry. Results: A higher prevalence of MAGE antigens was observed in patients with
hepatitis-B. Patients with expression of more tumour antigens in general had
better HCC-specific survival (P=0.022). The four tumour
antigens with high expression in HCC and no, or weak, expression in
surrounding tumour-free-liver tissue, were Annexin-A2, GPC-3, MAGE-C1 and
MAGE-C2, expressed in 90, 39, 17 and 20% of HCCs, respectively.
Ninety-five percent of HCCs expressed at least one of these four tumour
antigens. Interestingly, GPC-3 was associated with SALL-4 expression
(P=0.001), an oncofetal transcription factor highly
expressed in embryonal stem cells. SALL-4 and GPC-3 expression levels were
correlated with vascular invasion, poor differentiation and higher AFP
levels before surgery. Moreover, patients who co-expressed higher levels of
both GPC-3 and SALL-4 had worse HCC-specific survival
(P=0.018). Conclusions: We describe a panel of four tumour antigens with excellent coverage and good
tumour specificity in a western area, low-endemic for hepatitis-B. The
association between GPC-3 and SALL-4 is a novel finding and suggests that
GPC-3 targeting may specifically attack the tumour stem-cell
compartment.
Collapse
|
10
|
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]
|
11
|
Arnaboldi F, Menon A, Menegola E, Di Renzo F, Mirandola L, Grizzi F, Figueroa JA, Cobos E, Jenkins M, Barajon I, Chiriva-Internati M. Sperm protein 17 is an oncofetal antigen: a lesson from a murine model. Int Rev Immunol 2014; 33:367-74. [PMID: 24811209 DOI: 10.3109/08830185.2014.911856] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Sperm protein 17 (Sp17) was originally identified in the flagellum of spermatozoa and subsequently included in the subfamily of tumor-associated antigens known as cancer-testes antigens (CTA). Sp17 has been associated with the motility and migratory capacity in tumor cells, representing a link between gene expression patterns in germinal and tumor cells of different histological origins. Here we review the relevance of Sp17 expression in the mouse embryo and cancerous tissues, and present additional data demonstrating Sp17 complex expression pattern in this murine model. The expression of Sp17 in embryonic as well as adult neoplastic cells, but not normal tissues, suggests this protein should be considered an "oncofetal antigen." Further investigations are necessary to elucidate the mechanisms and functional significance of Sp17 aberrant expression in human adult cells and its implication in the pathobiology of cancer.
Collapse
Affiliation(s)
- F Arnaboldi
- 1Department of Human Morphology and Biomedical Sciences, University of Milan, Milan, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
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]
|
13
|
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
|
14
|
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: 1.1] [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
|
15
|
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: 5.3] [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
|
16
|
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: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
17
|
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
|
18
|
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: 1.7] [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
|
19
|
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: 0.4] [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
|
20
|
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: 1.5] [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
|
21
|
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: 2.7] [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
|
22
|
Gupta G, Sharma R, Chattopadhyay TK, Gupta SD, Ralhan R. Clinical significance of sperm protein 17 expression and immunogenicity in esophageal cancer. Int J Cancer 2007; 120:1739-47. [PMID: 17230514 DOI: 10.1002/ijc.22463] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
We recently identified sperm protein 17 (Sp17) transcripts in esophageal squamous cell carcinomas (ESCCs) by differential display. This study was designed to determine the clinical significance of Sp17 protein in different stages of esophageal tumorigenesis and to test the hypothesis that aberrant localization of Sp17 protein to immunosurveillant site may lead to production of anti-Sp17 antibodies in serum, which may be of clinical relevance in ESCCs. Sp17 transcripts were detected by RT-PCR in 26 of 30 (86%) ESCCs, while no transcripts were detected in normal esophageal tissues. Rabbit polyclonal antibody was raised against an immunogenic peptide of Sp17 and used to evaluate protein expression by immunohistochemistry. Expression of Sp17 protein was observed in 60/80 (75%) of ESCCs and 27/30 (90%) dysplastic tissues, while no detectable Sp17 expression was observed in 13 distant histologically normal epithelia. Sixteen of the 60 immunopositive ESCCs showed nuclear expression in addition to cytoplasmic localization of the protein. The circulating levels of anti-Sp17 antibodies, determined by ELISA, were significantly elevated in ESCC patients when compared with normal subjects (p < 0.001). Increasing Sp17 antibody titers were observed to be associated with the progressive disease in 4 patients. In conclusion, the study demonstrates expression of Sp17 protein in esophageal tumor as well as dysplastic tissues, suggesting it to be an early event in the development of ESCC. To our knowledge, this is the first report showing elevated levels of anti-Sp17 antibodies in ESCC patients.
Collapse
MESH Headings
- Adult
- Animals
- Antibodies, Neoplasm/immunology
- Antibodies, Neoplasm/metabolism
- Antigens, Surface/genetics
- Antigens, Surface/immunology
- Antigens, Surface/metabolism
- Autoantigens/immunology
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/immunology
- Biomarkers, Tumor/metabolism
- Blotting, Northern
- Blotting, Western
- Calmodulin-Binding Proteins
- Carcinoma, Squamous Cell/genetics
- Carcinoma, Squamous Cell/immunology
- Carcinoma, Squamous Cell/metabolism
- Carrier Proteins/genetics
- Carrier Proteins/immunology
- Carrier Proteins/metabolism
- Cell Nucleus/metabolism
- Cytoplasm/metabolism
- Esophageal Neoplasms/genetics
- Esophageal Neoplasms/immunology
- Esophageal Neoplasms/metabolism
- Esophagus/immunology
- Esophagus/metabolism
- Female
- Gene Expression Regulation, Neoplastic
- Humans
- Male
- Membrane Proteins
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Rabbits
- Reverse Transcriptase Polymerase Chain Reaction
Collapse
Affiliation(s)
- Garima Gupta
- Department of Biochemistry, All India Institute of Medical Sciences, Ansari Nagar, New Delhi 110029, India
| | | | | | | | | |
Collapse
|
23
|
Nakazato T, Kanuma T, Tamura T, Faried LS, Aoki H, Minegishi T. Sperm protein 17 influences the tissue-specific malignancy of clear cell adenocarcinoma in human epithelial ovarian cancer. Int J Gynecol Cancer 2007; 17:426-32. [PMID: 17309563 DOI: 10.1111/j.1525-1438.2007.00815.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Clear cell adenocarcinoma of the ovary has a poor prognosis due to chemoresistance and early metastasis to the lymph nodes. It also can result in endometriosis and is the second most frequent type of ovarian cancer in Japan. Serous adenocarcinoma of the ovary is another common epithelial cancer tissue subtype in Japan, and it is highly sensitive to chemotherapy. In the current study, we examined the differential expression of genes in these types of ovarian cancer and tried to analyze their functions, especially as they relate to chemoresistance. We used differential display to compare clear cell carcinoma and serous adenocarcinoma of the ovary. We identified sperm protein 17 (SP17) as a candidate gene related to the chemoresistance of clear cell carcinoma. Its differential expression was confirmed by real-time polymerase chain reaction. Because the function of the SP17 gene in ovarian cancer is not known, we examined the effect of small interfering RNA targeting the SP17 gene on the chemoresistance and proliferation of ES-2 ovarian cancer cells to paclitaxel, currently the most effective treatment for ovarian cancer. We found that this treatment decreased the chemoresistance of these cells to paclitaxel. Our results strongly suggest that SP17 plays a role in the resistance of clear cell carcinoma to chemotherapy without influencing their ability to proliferate
Collapse
MESH Headings
- Adenocarcinoma, Clear Cell/genetics
- Adenocarcinoma, Clear Cell/metabolism
- Adenocarcinoma, Clear Cell/pathology
- Antigens, Surface/genetics
- Antigens, Surface/metabolism
- Calmodulin-Binding Proteins
- Carrier Proteins/antagonists & inhibitors
- Carrier Proteins/genetics
- Carrier Proteins/metabolism
- Cells, Cultured
- Cystadenocarcinoma, Serous/genetics
- Cystadenocarcinoma, Serous/metabolism
- Drug Resistance, Neoplasm/genetics
- Female
- Gene Expression Profiling
- Gene Expression Regulation, Neoplastic/drug effects
- Humans
- Membrane Proteins
- Ovarian Neoplasms/genetics
- Ovarian Neoplasms/metabolism
- Ovarian Neoplasms/pathology
- RNA, Small Interfering/pharmacology
- Reverse Transcriptase Polymerase Chain Reaction
Collapse
Affiliation(s)
- T Nakazato
- Department of Gynecology and Reproductive Medicine, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | | | | | | | | | | |
Collapse
|
24
|
Gattei V, Fonsatti E, Sigalotti L, Degan M, Di Giacomo AM, Altomonte M, Calabrò L, Maio M. Epigenetic immunomodulation of hematopoietic malignancies. Semin Oncol 2005; 32:503-10. [PMID: 16210091 DOI: 10.1053/j.seminoncol.2005.07.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Significant progress has been made in the clinical management of hematologic malignancies; nevertheless, a proportion of patients still remains unresponsive to available therapeutic options. Furthermore, patients who respond to specific therapeutic regimens may still require additional treatment to eradicate minimal residual disease. In this scenario, novel immunotherapeutic strategies may significantly impact on the clinical course of hematopoietic tumors in different clinical stages of disease. Among immunotherapeutic approaches under development, promising clinical results are being obtained with vaccination of patients with solid malignancies against cancer testis antigens (CTA), which belong to a growing family of methylation-regulated tumor-associated antigens (TAA) shared among human malignancies of different histologies. Based on these notions, the emerging preclinical and clinical evidence suggest that an immunomodulatory role for epigenetic drugs is highly relevant; in fact, by interfering with DNA methylation, these compounds induce or upregulate the constitutive expression of CTA on actively proliferating neoplastic cells. This novel activity of epigenetic drugs combines with their well-known cytotoxic, pro-apoptotic and differentiating activities in hematopoietic tumors that are extensively described in other chapters of this issue. This review will focus on the expression of CTA in hematopoietic malignancies, on their epigenetic regulation, and on the foreseeable immunotherapeutic implications of DNA hypometylating drugs to design new CTA-based chemo-immunotherapeutic approaches in patients with hematopoietic tumors.
Collapse
Affiliation(s)
- Valter Gattei
- Clinical and Experimental Hematology Research Unit, Centro di Riferimento Oncologico, IRCCS, Aviano, Italy
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Dadabayev AR, Wang Z, Zhang Y, Zhang J, Robinson WR, Lim SH. Cancer immunotherapy targeting Sp17: when should the laboratory findings be translated to the clinics? Am J Hematol 2005; 80:6-11. [PMID: 16138340 DOI: 10.1002/ajh.20415] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Despite advances in chemotherapeutic agents, the prognosis for some cancers remains extremely poor, suggesting the need for other treatment modalities. Immunotherapy appears an ideal approach because the mechanisms of tumor cell killing induced by tumor vaccines are different from those from chemotherapy. Various investigations are ongoing to identify suitable targets for this purpose. Sperm protein 17 (Sp17) was originally identified by our group as a novel cancer-testis antigen in various malignancies, including multiple myeloma. Sp17 is a highly immunogenic protein and the observation that more than 90% of vasectomized males develop immunity against Sp17 suggests the opportunity and safety of Sp17 for tumor vaccines. Recent works by other workers suggest a low level of expression of Sp17 in some normal tissues, and investigators have questioned whether Sp17 is in fact a suitable target for immunotherapy. In this paper, we review the general principles of immunotherapy and provide evidence supporting the highly immunogenic nature of Sp17. We also address the discrepancies between the objectives of oncologists involved in treating cancer patients and their familiarity with acceptable levels of toxicity of any effective therapy and those of pure laboratory-based investigators. Finally, we present some early clinical data supporting the rationale for further investigations of Sp17 for tumor vaccines.
Collapse
Affiliation(s)
- Alisher R Dadabayev
- Division of Hematology and Oncology, Texas Tech University Health Sciences Center, Amarillo, 79106, USA
| | | | | | | | | | | |
Collapse
|
26
|
Zhang Y, Wang Z, Robinson WR, Lim SH. Combined real time PCR and immunohistochemical evaluation of sperm protein 17 as a cancer-testis antigen. Eur J Haematol 2004; 73:280-4. [PMID: 15347315 DOI: 10.1111/j.1600-0609.2004.00308.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
We previously identified sperm protein 17 (Sp17) as a normal testicular protein aberrantly expressed in a proportion of multiple myeloma (MM). However, recent studies have generated controversies on the normal tissue expression of Sp17 and whether or not it is a suitable target for immunotherapy. In this study, we have used a combination of real time polymerase chain reaction and immunohistochemistry on a large panel of normal tissues. Although Sp17 transcripts could be detected in some normal tissues, the levels of expression were <2% of those in normal testis. In contrast, Sp17+ myeloma cells expressed 3-18% of normal testis levels of Sp17 transcript. Immunohistochemistry using two Sp17 murine monoclonal antibodies, each directed at a non-overlapping B-cell epitope, showed Sp17 protein to be expressed only in testis and not any other normal tissues. Specificity of binding of the antibodies to testis was also confirmed in competitive binding assays. Our results therefore further suggest Sp17 as a cancer-testis antigen in MM and support its suitability as a target for immunotherapy.
Collapse
Affiliation(s)
- Yana Zhang
- Division of Hematology and Oncology, Texas Tech University Health Sciences Center, Amarillo, USA
| | | | | | | |
Collapse
|
27
|
Wang Z, Zhang Y, Ramsahoye B, Bowen D, Lim SH. Sp17 gene expression in myeloma cells is regulated by promoter methylation. Br J Cancer 2004; 91:1597-603. [PMID: 15381930 PMCID: PMC2409933 DOI: 10.1038/sj.bjc.6602160] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
The mechanisms underlying sperm protein 17 (Sp17) gene expression in myeloma cells remained unclear. Using reverse transcription–polymerase chain reaction (RT–PCR), Sp17 transcripts were detected in ARK-B, ARP-1, RPMI-8226 and KMS-11 but not in H929, IM-9, MM1-R and U266 cells. Using a panel of primer pairs in methylation-sensitive PCR to amplify overlapping gene segments, our screening studies showed that the HpaII sites at −359 and −350 are involved in the regulation of Sp17 gene expression. To confirm the differences in methylation status between Sp17-positive and Sp17-negative cell lines, KMS-11 cells (Sp17-positive) and IM-9 cells (Sp17-negative) were subjected to the more accurate method of bisulphite conversion. KMS-11 cells were more hypomethylated at these HpaII sites of exon 1 compared to IM-9 cells, indicating the association of hypomethylated promoter with Sp17 gene expression. In addition, the level of methylation at other CpG sites within the promoter sequence was also higher in IM-9 than KMS-11. Exon 1 was cloned into a reporter vector, pCAT*3 Enhancer. Chloramphenicol acetyl transferase (CAT) activity was restored in cells transfected with the recombinant plasmid, indicating the promoter function of exon 1. Exposure of Sp17-negative cell lines to the hypomethylating agent, 5-azacytidine, resulted in the upregulation of Sp17 gene expression. Our results therefore provide evidence for the regulation of Sp17 gene expression by promoter methylation.
Collapse
MESH Headings
- Antigens, Surface
- Antimetabolites, Antineoplastic/pharmacology
- Azacitidine/pharmacology
- Calmodulin-Binding Proteins
- Carrier Proteins/genetics
- Carrier Proteins/metabolism
- Chloramphenicol O-Acetyltransferase/metabolism
- CpG Islands
- DNA Methylation
- Gene Expression Regulation, Neoplastic
- Humans
- Membrane Proteins
- Multiple Myeloma/genetics
- Multiple Myeloma/metabolism
- Promoter Regions, Genetic/genetics
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- RNA, Neoplasm/genetics
- RNA, Neoplasm/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Transcription, Genetic
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- Z Wang
- Division of Hematology and Oncology, Texas Tech University Health Sciences Center, Amarillo, TX, USA
- Biotherapy and Stem Cell Transplant Program, Don and Sybil Harrington Cancer Center, 1500 Wallace Boulevard, Amarillo, TX 79106, USA
| | - Y Zhang
- Division of Hematology and Oncology, Texas Tech University Health Sciences Center, Amarillo, TX, USA
- Biotherapy and Stem Cell Transplant Program, Don and Sybil Harrington Cancer Center, 1500 Wallace Boulevard, Amarillo, TX 79106, USA
| | - B Ramsahoye
- Division of Hematology and Oncology, Texas Tech University Health Sciences Center, Amarillo, TX, USA
- Biotherapy and Stem Cell Transplant Program, Don and Sybil Harrington Cancer Center, 1500 Wallace Boulevard, Amarillo, TX 79106, USA
| | - D Bowen
- Division of Hematology and Oncology, Texas Tech University Health Sciences Center, Amarillo, TX, USA
- Biotherapy and Stem Cell Transplant Program, Don and Sybil Harrington Cancer Center, 1500 Wallace Boulevard, Amarillo, TX 79106, USA
| | - S H Lim
- Division of Hematology and Oncology, Texas Tech University Health Sciences Center, Amarillo, TX, USA
- Biotherapy and Stem Cell Transplant Program, Don and Sybil Harrington Cancer Center, 1500 Wallace Boulevard, Amarillo, TX 79106, USA
- Division of Hematology and Oncology, Texas Tech University Health Sciences Center, Amarillo, TX, USA. E-mail:
| |
Collapse
|
28
|
Abstract
Most patients with multiple myeloma (MM) cannot be cured with currently available therapies. Although complete remission could be achieved in about 50% of newly diagnosed patients with high-dose chemotherapy and tandem transplantation, relapses of the underlying disease occur frequently. To realize long-term disease-free survival, it will be necessary to develop complementary therapies that are non-cross-resistant with chemotherapy. To this end, immunotherapy aimed at inducing or enhancing tumor-specific immunity that may control or eradicate remaining tumor cells may be an appealing method. Dendritic cells (DCs) are professional antigen-presenting cells and considered the best natural adjuvants for immunotherapy in malignancies. Vaccination with tumor antigen-pulsed DCs has been shown to be protective and therapeutic in animal tumor models, and induced a strong tumor-specific immunity and durable tumor regression in human solid tumors and B-cell lymphoma. As a result, clinical trials in various human malignancies have been initiated. This review will focus on DC-based immunotherapy in MM. I will discuss myeloma antigens and antigen-specific immune responses, the capacity of DCs to present myeloma antigens and induce cytotoxic T-cell responses, and clinical experience of DC vaccination in myeloma patients.
Collapse
Affiliation(s)
- Qing Yi
- Myeloma Institute for Research and Therapy, Arkansas Cancer Research Center University of Arkansas for Medical Sciences, 4301 West Markham Street, Slot #776, Little Rock, AR 72205, USA.
| |
Collapse
|
29
|
Lea IA, Widgren EE, O'Rand MG. Association of sperm protein 17 with A-kinase anchoring protein 3 in flagella. Reprod Biol Endocrinol 2004; 2:57. [PMID: 15257753 PMCID: PMC484205 DOI: 10.1186/1477-7827-2-57] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2004] [Accepted: 07/16/2004] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Sperm protein 17 (Sp17) is a three-domain protein that contains: 1) a highly conserved N-terminal domain that is 45% identical to the human type II alpha regulatory subunit (RII alpha) of protein kinase A (PKA); 2) a central sulphated carbohydrate-binding domain; and 3) a C-terminal Ca++/calmodulin (CaM) binding domain. Although Sp17 was originally discovered and characterized in spermatozoa, its mRNA has now been found in a variety of normal mouse and human tissues. However, Sp17 protein is found predominantly in spermatozoa, cilia and human neoplastic cell lines. This study demonstrates that Sp17 from spermatozoa binds A-kinase anchoring protein 3 (AKAP3), confirming the functionality of the N-terminal domain. METHODS In this study in vitro precipitation and immunolocalization demonstrate that Sp17 binds to AKAP3 (AKAP110) in spermatozoa. RESULTS Sp17 is present in the head and tail of spermatozoa, in the tail it is in the fibrous sheath, which contains AKAP3 and AKAP4. Recombinant AKAP3 and AKAP4 RII binding domains were synthesized as glutathione S-transferase (GST) fusion proteins immobilized on glutathione-agarose resin and added to CHAPS extracts of human spermatozoa. Western blots of bound and eluted proteins probed with anti-Sp17 revealed that AKAP3 bound and precipitated a significant level of Sp17 while AKAP4 did not. AKAP4 binds AKAP3 and expression of AKAP3 is reduced in AKAP4 knockout sperm, therefore we tested AKAP4 knockout spermatozoa for Sp17 and found that there was a reduction in the amount of Sp17 expressed when compared to wild type spermatozoa. Co-localization of AKAP3 and Sp17 by immunofluorescence was demonstrated along the length of the principal piece of the flagella. CONCLUSIONS As predicted by its N-terminal domain that is 45% identical to the human RIIalpha of PKA, Sp17 from spermatozoa binds the RII binding domain of AKAP3 along the length of the flagella.
Collapse
Affiliation(s)
- Isabel A Lea
- University of North Carolina at Chapel Hill, Department of Cell and Developmental Biology, Chapel Hill, NC 27599, USA
| | - Esther E Widgren
- University of North Carolina at Chapel Hill, Department of Cell and Developmental Biology, Chapel Hill, NC 27599, USA
| | - Michael G O'Rand
- University of North Carolina at Chapel Hill, Department of Cell and Developmental Biology, Chapel Hill, NC 27599, USA
| |
Collapse
|
30
|
Abstract
Therapy for patients with multiple myeloma (MM) is currently unsatisfactory and most patients eventually succumb to relapsed disease. DCs are a subset of leukocytes with the capacity to initiate and control the adaptive immune response against many cancers, including MM. In MM patients, in vivo DC function is often abnormal, however, it appears that it can be restored by in vitro manipulation. This has led to the development of DC immunotherapy for MM patients. We review the background research leading to the recognition of an anti-MM immune response, and discuss abnormalities in DC function, potential tumor-associated Ags, and the results of clinical trials of DC immunotherapy in MM patients.
Collapse
Affiliation(s)
- C J Turtle
- Dendritic Cell Laboratory, Mater Medical Research Institute, Raymond Terrace, South Brisbane, Queensland, Australia
| | | | | | | |
Collapse
|
31
|
Straughn JM, Shaw DR, Guerrero A, Bhoola SM, Racelis A, Wang Z, Chiriva-Internati M, Grizzle WE, Alvarez RD, Lim SH, Strong TV. Expression of sperm protein 17 (Sp17) in ovarian cancer. Int J Cancer 2004; 108:805-11. [PMID: 14712480 DOI: 10.1002/ijc.11617] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Sperm protein 17 (Sp17) is an antigenic protein highly expressed in spermatozoa. Sp17 expression was demonstrated recently in multiple myeloma, suggesting that it may be a novel cancer-testis antigen. Expression of Sp17 mRNA and protein was examined in human ovarian tumors. Sp17 mRNA was evaluated by reverse transcription-polymerase chain reaction (RT-PCR) and Northern blot analysis of RNA derived from epithelial ovarian tumors and normal tissues. RT-PCR analysis detected Sp17 transcripts in 15 of 18 (83%) primary ovarian tumors. The transcript was not detected in RNA derived from normal uterus or cervix, whereas weak expression was noted in some normal ovarian tissue samples. Northern blot analysis showed no detectable Sp17 mRNA expression in normal tissues, including normal ovary, but showed Sp17 expression in 17 of 25 ovarian tumors (68%). To evaluate protein expression, mouse monoclonal antibodies were produced against recombinant Sp17 protein and used in Western blot and immunohistochemical analyses of normal reproductive tissue and primary ovarian tumor samples. Sp17 protein was detected by Western blot analysis in normal spermatozoa and in 8 of 19 ovarian tumor samples. Immunohistochemical studies showed Sp17 expression in spermatozoa, ciliated cells of the female reproductive tract, and most ovarian tumors evaluated. Tumors showed a predominantly nuclear localization of Sp17 expression, with some cytoplasmic staining. These results demonstrate that Sp17, a protein with restricted expression in somatic tissues, is expressed in ovarian tumors. Because Sp17 is immunogenic, it may represent a novel target for immunotherapeutic interventions for ovarian cancer patients.
Collapse
Affiliation(s)
- J Michael Straughn
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, The University of Alabama, Birmingham, AL 35294, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Chiriva-Internati M, Wang Z, Pochopien S, Salati E, Lim SH. Identification of a sperm protein 17 CTL epitope restricted by HLA-A1. Int J Cancer 2003; 107:863-5. [PMID: 14566839 DOI: 10.1002/ijc.11486] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Sperm protein 17 (Sp17) is a novel cancer-testis antigen. We previously reported the successful generation of Sp17-specific HLA-A1-restricted cytotoxic T-lymphocytes (CTLs) from the peripheral blood of a healthy donor using a recombinant Sp17 protein. These CTLs were able to kill not only target cells pulsed with the recombinant protein but also fresh Sp17+ tumor cells. In the present study, we have identified a nonapeptide sequence within the Sp17 protein that is predicted to have a high binding affinity for the HLA-A1 molecules. We generated the synthetic nonapeptide and successfully propagated a peptide-specific CTL line. We confirmed that peptide Sp17(103-111) (ILDSSEEDK) contains an Sp17 CTL epitope restricted by HLA-A1 and identified amino acid residues 104, 107, 109 and 110 as crucial for the CTL lysis. Our findings therefore provide the tool for the characterization of CD8 T-cell function in vivo and generation of epitope-specific treatment strategies.
Collapse
Affiliation(s)
- Maurizio Chiriva-Internati
- Division of Hematology and Oncology, Texas Tech University Health Sciences Center, Amarillo, Texas 79106, USA
| | | | | | | | | |
Collapse
|
33
|
Grizzi F, Chiriva-Internati M, Franceschini B, Hermonat PL, Soda G, Lim SH, Dioguardi N. Immunolocalization of sperm protein 17 in human testis and ejaculated spermatozoa. J Histochem Cytochem 2003; 51:1245-8. [PMID: 12923251 DOI: 10.1177/002215540305100916] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Sperm protein 17 (Sp17) is a highly conserved mammalian protein whose primary function is still poorly understood. Immunohistochemistry (IHC) in the human testis reveals the presence of Sp17 in some spermatocytes and abundantly in spermatids. All spermatogonia, Sertoli cells, and Leydig cells appear to be immunonegative for Sp17, whereas some interstitial cells are immunopositive. IHC recognized two distinct populations (immunopositive or not for Sp17) in the ejaculated spermatozoa. Although it will be necessary to clarify why some ejaculated spermatozoa do not contain Sp17, its distribution suggests that this protein may be associated with some phases of germinal cell differentiation.
Collapse
Affiliation(s)
- Fabio Grizzi
- Scientific Direction, Istituto Clinico Humanitas, Rozzano, Milan, Italy.
| | | | | | | | | | | | | |
Collapse
|
34
|
Abstract
Cell surface heparan sulfate (HS) influences a multitude of molecules, cell types, and processes relevant to inflammation. HS binds to cell surface and matrix proteins, cytokines, and chemokines. These interactions modulate inflammatory cell maturation and activation, leukocyte rolling, and tight adhesion to endothelium, as well as extravasation and chemotaxis. The syndecan family of transmembrane proteoglycans is the major source of cell surface HS on all cell types. Recent in vitro and in vivo data suggest the involvement of syndecans in the modulation of leukocyte-endothelial interactions and extravasation, the formation of chemokine and kininogen gradients, participation in chemokine and growth factor signaling, as well as repair processes. Thus, the complex role of HS in inflammation is reflected by multiple functions of its physiological carriers, the syndecans. Individual and common functions of the four mammalian syndecan family members can be distinguished. Recently generated transgenic and knockout mouse models will facilitate analysis of the individual processes that each syndecan is involved in.
Collapse
|
35
|
Zendman AJW, Ruiter DJ, Van Muijen GNP. Cancer/testis-associated genes: identification, expression profile, and putative function. J Cell Physiol 2003; 194:272-88. [PMID: 12548548 DOI: 10.1002/jcp.10215] [Citation(s) in RCA: 194] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Cancer/testis-associated genes (CTAs) are a subgroup of tumor antigens with a restricted expression in testis and malignancies. During the last decade, many of these immunotherapy candidate genes have been discovered using various approaches. Most of these genes are localized on the X-chromosome, often as multigene families. Methylation status seems to be the main, but not the only regulator of their specific expression pattern. In testis, CTAs are exclusively present in cells of the germ cell lineage, though there is a lot of variation in the moment of expression during different stages of sperm development. Likewise, there is also a lot of heterogeneity in the expression of CTAs in melanoma samples. Clues regarding functionality of CTAs for many of these proteins point to a role in cell cycle regulation or transcriptional control. Better insights in the function of these genes may shed light on the link between spermatogenesis and tumor growth and could be of use in anti-tumor therapies. This review outlines the CTA family and focuses on their expression and putative function during male germ cell development and melanocytic tumor progression.
Collapse
Affiliation(s)
- Albert J W Zendman
- Department of Pathology, University Medical Center St. Radboud, Nijmegen, The Netherlands.
| | | | | |
Collapse
|
36
|
|
37
|
De Jong A, Buchli R, Robbins D. Characterization of sperm protein 17 in human somatic and neoplastic tissue. Cancer Lett 2002; 186:201-9. [PMID: 12213290 DOI: 10.1016/s0304-3835(02)00350-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Sperm protein 17 (Sp17) is a highly antigenic, testes-specific protein whose known function is to bind sperm to the zona pellucida. However, the Sp17 gene has been recently detected in normal non-testes tissues and malignant neoplasias. As the role of Sp17 in non-testes tissues is unknown, the characterization of the Sp17 gene in highly proliferating tissues may provide further insight into the regulation and alternative function of Sp17. Reverse transcriptase-polymerase chain reaction (RT-PCR) was used to amplify the Sp17-1 transcript in multiple normal human tissues and cancer cell lines. Similarly, the Sp17-2 gene was examined by PCR. In addition, Northern and Western blot analyses were used to detect Sp17 mRNA and protein expression. The Sp17-1a and Sp17-1b transcripts were amplified from cancer cell lines. Similarly, an Sp17-2 transcript was also detected in cancer cell lines. Furthermore, Northern blot analysis revealed Sp17 mRNA expression in all cancer cell lines examined. However, Sp17 protein expression was not detected. The differential detection of the Sp17 transcripts in cancer cell lines as compared to normal non-testes tissues, suggests a potential pathogenic role for Sp17 in diseased cells. Moreover, the Sp17-2 transcript may be a marker for highly proliferating cells. Collectively, these data implicate Sp17 as a cancer testis antigen.
Collapse
Affiliation(s)
- Ann De Jong
- Department of Internal Medicine, School of Medicine, University of California, Davis, CA 95616, USA
| | | | | |
Collapse
|
38
|
Buchli R, De Jong A, Robbins DL. Genomic organization of an intron-containing sperm protein 17 gene (Sp17-1) and an intronless pseudogene (Sp17-2) in humans: a new model. BIOCHIMICA ET BIOPHYSICA ACTA 2002; 1578:29-42. [PMID: 12393185 DOI: 10.1016/s0167-4781(02)00478-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Sp17 was initially thought to be a sperm specific protein involved in the interaction of the spermatozoon with the oocyte's surrounding extracellular glycoprotein matrix. Recent reports, however, indicate that Sp17 expression is neither testis-specific nor is it exclusively used for binding to the zona pellucida of the oocyte. In this study, we provide comprehensive characterization of the genomic structure of Sp17. We identified an intron-containing gene (Sp17-1) containing five exonic and four intronic sequences. Analysis of Sp17 transcripts using rapid amplification of DNA complementary to RNA (cDNA) ends (RACE) and polymerase chain reaction (PCR) techniques showed the presence of alternative polyadenylation resulting in the production of varying lengths of mRNAs as well as the usage of different transcriptional start sites. Moreover, an earlier description of the human Sp17 mRNA describing a splice variant could not be confirmed. Comparison to mouse Sp17 gene organization demonstrated a high degree of conservation, suggesting selective evolutionary pressure for this protein to retain a conserved gene architecture. Additionally, we identified a second gene (Sp17-2), whose most striking characteristic was the complete absence of introns. This Sp17-2 gene has likely arisen by reverse transcription (RT) of a spliced Sp17-1 mRNA with subsequent integration into the human genome. Its open reading frame (ORF) is interrupted by stop codons, giving rise to a pseudogene. Furthermore, Southern blot analysis of human genomic DNA indicated the possibility of additional Sp17 species within the human genome.
Collapse
Affiliation(s)
- Rico Buchli
- Division of Rheumatology/Allergy and Clinical Immunology, Department of Internal Medicine, University of California-Davis, 1 Shields Avenue, TB 192, Davis, CA 95616, USA
| | | | | |
Collapse
|
39
|
Chiriva-Internati M, Wang Z, Salati E, Wroblewski D, Lim SH. Successful generation of sperm protein 17 (Sp17)-specific cytotoxic T lymphocytes from normal donors: implication for tumour-specific adoptive immunotherapy following allogeneic stem cell transplantation for Sp17-positive multiple myeloma. Scand J Immunol 2002; 56:429-33. [PMID: 12234264 DOI: 10.1046/j.1365-3083.2002.01138.x] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Sperm protein 17 (Sp17) is a highly immunogenic cancer-testis antigen expressed by tumour cells from up to 30% of patients with multiple myeloma (MM). We recently successfully generated Sp17-specific human leucocyte antigen (HLA)-A1 and B27-restricted cytotoxic T lymphocytes (CTLs) from the peripheral blood of a healthy donor. Because CTLs were able to kill HLA-matched fresh myeloma cells, it may be possible to generate and administer myeloma-specific donor T cells to MM patients following allogeneic stem cell transplantation to enhance graft-versus-myeloma (GVM) without inducing graft-versus-host disease (GVHD). To determine how widely applicable this approach is, we have determined the ability to generate Sp17-specific CTLs from four consecutive healthy donors with other HLA class I phenotypes. We found that Sp17-specific HLA class I-restricted CTLs could be easily generated from all four donors. Sp17-specific CTLs were primarily CD8 in phenotype and produced interferon-gamma and very little interleukin-4. These T cells killed target cells primarily via the perforin-mediated route. These results therefore suggest that myeloma-specific donor T-cell infusion that targets Sp17 to selectively enhance GVM could be applicable to patients with Sp17+ MM.
Collapse
Affiliation(s)
- M Chiriva-Internati
- Division of Hematology and Oncology, Texas Tech University School of Medicine at Amarillo and Blood Stem Cell Transplant Program, Don and Sybil Harrington Cancer Center, Amarillo, TX 79106, USA
| | | | | | | | | |
Collapse
|
40
|
Chiriva-Internati M, Wang Z, Salati E, Bumm K, Barlogie B, Lim SH. Sperm protein 17 (Sp17) is a suitable target for immunotherapy of multiple myeloma. Blood 2002; 100:961-5. [PMID: 12130509 DOI: 10.1182/blood-2002-02-0408] [Citation(s) in RCA: 76] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Sperm protein 17 (Sp17) is a protein recently identified as a novel cancer-testis (CT) antigen in multiple myeloma (MM). Because this tumor antigen demonstrates a very restricted normal tissue expression, Sp17 may be an excellent target for tumor vaccine of MM. In this study, we determined the ability to generate Sp17-specific HLA class I-restricted cytotoxic T lymphocytes (CTLs) from the peripheral blood of 4 patients with MM, 3 consecutive Sp17(+) patients, and 1 Sp17(-) patient. Dendritic cells were generated from monocytes of 4 patients with MM and used to present a recombinant Sp17 protein to autologous T cells. Following 4 rounds of antigen stimulation, the CTLs were tested for their ability to kill autologous targets in an Sp17-dependent and HLA-class I- restricted manner in standard cytotoxicity assays. Despite previous chemotherapy and the immunosuppression so often associated with MM, CTL generation was successful in all 4 patients, irrespective of the Sp17 status of their tumors. Most importantly, the CTLs were able to lyse autologous tumor cells that expressed Sp17. Tumor cell lysis in all cases appeared to be mainly mediated by perforin and could be blocked by concanamycin A. We conclude that Sp17 is a suitable target for immunotherapy of MM. Our findings provide the basis for a clinical study aimed at inducing a cellular immune response directed at Sp17(+) MM.
Collapse
|
41
|
Grizzi F, Lim SH, Chiriva-Internati M, Franceschini B, Wang Z, Lawrence D, Dioguardi N. Sperm protein 17 is not expressed on normal leukocytes. Blood 2002; 99:3479-80; author reply 3480-1. [PMID: 12001904 DOI: 10.1182/blood-2001-11-0064] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|