1
|
Liu X, Shen L, Wang H. Decreased Expression of PLD2 Promotes EMT in Colorectal Cancer Invasion and Metastasis. J Cancer 2024; 15:2981-2993. [PMID: 38706911 PMCID: PMC11064252 DOI: 10.7150/jca.89970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 03/09/2024] [Indexed: 05/07/2024] Open
Abstract
Background and Objectives: PLD2 has been identified as playing a critical role in cancer cell motility and migration and other pathophysiological processes. We investigated the expression of PLD2 and its biological functions and clinical implications in human colorectal cancer. Materials and Methods: In this study, the expressions of PLD2 were analyzed in CRC cell lines and CRC samples by RT-PCR, western blot and immunohistochemistry. The PLD enzyme activity was studied using an PLD detection kit. We also performed matrigel invasion assay to evaluate the invasive capabilities in CRC cells. The expressions of EMT-related markers were quantified at mRNA and protein level using RT-PCR and western blot. We performed high-throughput RNA sequencing on PLD2 knockdown and overexpression CRC cell lines to explore the changes in gene expression associated with PLD2. Result: Herein, we showed that PLD2 expression was relatively low in CRC cell lines and CRC samples and PLD2 deficiency was significantly correlated with more advanced clinical phenotype regarding lymphatic and distant metastasis and poor patient survival. We also detected that PLD2 knockdown favored epithelial-mesenchymal transition (EMT) and thus promoted CRC invasion and metastasis. Further exploration uncovered that the expressions of several important genes closely related to metabolic pathways in CRC were noticeably altered due to PLD2 deficiency, including ID1, IFIT4, OASL, IFIT2 and CTAG2. Conclusion: Our results revealed that PLD2 deficiency promotes cell invasion and metastasis in CRC via EMT indicating PLD2 might have an important implication in carcinogenesis and progression and would be a new therapeutic target for cancer treatment.
Collapse
Affiliation(s)
- Xuan Liu
- Department of General Surgery, Central Hospital of Xuhui District, Shanghai, China
| | - Lei Shen
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Haiyu Wang
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
2
|
Misir Z, Glavčić G, Janković S, Kruljac I, Čugura-Filipović J, Čimić K, Ulamec M. The role of the NY-ESO-1 in the prognosis of gastric cancer. BIOMOLECULES & BIOMEDICINE 2023; 24:813-820. [PMID: 38153439 PMCID: PMC11293239 DOI: 10.17305/bb.2023.9937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 12/17/2023] [Accepted: 12/28/2023] [Indexed: 12/29/2023]
Abstract
Gastric cancer (GC) is one of the most common malignancies worldwide and the fourth leading cause of cancer-related deaths. GC is a multifactorial disease influenced by both environmental and genetic factors. Its most critical features include invasiveness and high metastatic potential. Metastasis is a complex process, and our understanding of the mechanisms involved remains incomplete. Growing evidence suggests that cancer-testis antigens (CTAs) play a crucial role in the metastatic potential of various tumors. Several studies have linked CTA expression with lower tumor differentiation, higher metastatic potential, and poor chemotherapy response. New York esophageal squamous cell carcinoma-1 (NY-ESO-1) antigen, part of the CTA group, is expressed in tumor tissues, while its expression in normal tissues is restricted to spermatogonia. This study aimed to determine the expression of NY-ESO-1 in primary adenocarcinoma of the stomach, both with and without metastasis in regional lymph nodes, and to compare it with TNM stage, age, gender, and survival. We analyzed GC tissue from 53 node-negative and 55 node-positive primary gastric carcinoma patients for NY-ESO-1 expression using immunohistochemical assay. The results were correlated with clinicopathological parameters and survival. Patients with positive NY-ESO-1 expression in primary tumors had a median survival of 19.0 months (range 14.1-24.0), in contrast to those with negative expression, who had a median survival of 52.0 months (range 0.0-133.3) (chi-square 7.99, P=0.005). T status, N status, and NY-ESO-1 expression were all independently associated with shorter survival. No significant difference in NY-ESO-1 expression in primary tumors was observed concerning lymph node metastasis status. In summary, our findings suggest that increased expression of NY-ESO-1 could potentially serve as a prognostic biomarker for GC.
Collapse
Affiliation(s)
- Zvonimir Misir
- Department of Surgery, University Hospital Center “Sestre milosrdnice”, Zagreb, Croatia
| | - Goran Glavčić
- Department of Surgery, University Hospital Center “Sestre milosrdnice”, Zagreb, Croatia
| | - Suzana Janković
- Department of Surgery, University Hospital Center “Sestre milosrdnice”, Zagreb, Croatia
| | - Ivan Kruljac
- Department of Clinical Research and Biostatistics, Poliklinika Solmed d.o.o., Zagreb, Croatia
| | | | - Kristina Čimić
- Department of Gynecology, General Hospital “Dr Ivo Pedišić”, Sisak, Croatia
| | - Monika Ulamec
- Clinical Department of Pathology and Cytology “Ljudevit Jurak”, University Hospital Center “Sestre milosrdnice”, Zagreb, Croatia
- Department of Pathology and Center of Excellence for Reproductive and Regenerative Medicine, School of Medicine, University of Zagreb, Zagreb, Croatia
| |
Collapse
|
3
|
Julve M, Kennedy O, Frampton AE, Bagwan I, Lythgoe MP. Gene of the month: cancer testis antigen gene 1b (NY-ESO-1). J Clin Pathol 2023; 77:1-7. [PMID: 37857483 DOI: 10.1136/jcp-2023-209053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/04/2023] [Indexed: 10/21/2023]
Abstract
Cancer testis antigen gene 1B (CTAG1B) and its associated gene product; New York oesophageal squamous carcinoma 1 (NY-ESO-1), represent a unique and promising target for cancer immunotherapy. As a member of the cancer testis antigen family (CTA), the protein's restricted expression pattern and ability to elicit spontaneous humoural and cellular immune responses has resulted in a plethora of novel modalities and approaches attempting to harness its immunotherapeutic anti-cancer potential. Here, we discuss the structure and function of CTAG1B/NY-ESO-1 in both health and disease, immunohistochemical detection, as well as the most promising advances in the development of associated anti-cancer therapies. From cancer vaccines to engineered cellular therapy approaches, a multitude of immunotherapies targeting CTA's are coming to the forefront of oncology. Although the efficacy of such approaches have yet to provide convincing evidence of durable response, early phase clinical trial data has resulted in some exciting findings which will have significant potential to act as a platform for future practice changing technologies.
Collapse
Affiliation(s)
- Max Julve
- Department of Surgery and Cancer, Imperial College London, London, UK
| | - Oliver Kennedy
- Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Adam Enver Frampton
- Department of Surgery and Cancer, Imperial College London, London, UK
- Section of Oncology, Deptartment of Clinical and Experimental Medicine, FHMS, University of Surrey, Guildford, UK
| | - Izhar Bagwan
- Department of Cellular Pathology, Royal Surrey Hospital, Guildford, UK
| | - Mark P Lythgoe
- Department of Surgery and Cancer, Imperial College London, London, UK
| |
Collapse
|
4
|
Li D, Xia L, Huang P, Wang Z, Guo Q, Huang C, Leng W, Qin S. Serine protease PRSS56, a novel cancer-testis antigen activated by DNA hypomethylation, promotes colorectal and gastric cancer progression via PI3K/AKT axis. Cell Biosci 2023; 13:124. [PMID: 37400936 DOI: 10.1186/s13578-023-01060-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 05/27/2023] [Indexed: 07/05/2023] Open
Abstract
BACKGROUND Cancer/testis (CT) antigens/genes are usually overexpressed in cancers and exhibit high immunogenicity, making them promising targets for immunotherapy and cancer vaccines. The role of serine protease PRSS56 in cancers remains unknown to date. METHODS RNA sequencing studies were performed to screen CT genes in gastric cancer (GC) and colorectal cancer (CRC) cells exposed to DNA methyltransferase inhibitor 5-aza-2'-deoxycytidine (5-AZA-CdR). Bioinformatics analysis was conducted to analyze the correlation between PRSS56 expression and DNA methylation. Functional experiments were performed to explore the biological function of PRSS56 in GC and CRC. RESULTS In this study, we identified the testis-specific serine proteases PRSS56 as a novel CT antigen. PRSS56 was frequently overexpressed in various cancers, especially in gastrointestinal cancer. PRSS56 expression was negatively associated with promoter DNA methylation level, and positively associated with gene body methylation level. PRSS56 expression was significantly activated in colorectal and gastric cancer cells exposed to DNA methyltransferase inhibitors. Importantly, our finding highlights that the decreased methylation level of the CpG site cg10242318 in the PRSS56 promoter region resulted in its overexpression in GC and CRC. Additionally, functional assays verified that PRSS56 overexpression activated PI3K-AKT signaling in GC and CRC. CONCLUSION Serine protease PRSS56 is a novel CT antigen that is reactivated in cancers by promoter DNA hypomethylation. PRSS56 functions oncogenic roles in GC and CRC by activating of PI3K/AKT axis. Our results presented here represent the first data on the function of the serine protease PRSS56 in cancers.
Collapse
Affiliation(s)
- Dandan Li
- Department of Stomatology, Taihe Hospital and Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei, 442000, P.R. China
- Laboratory of Tumor biology, Academy of Bio-Medicine Research, Hubei University of Medicine, Shiyan, Hubei, 442000, P.R. China
| | - Lingyun Xia
- Department of Stomatology, Taihe Hospital and Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei, 442000, P.R. China
| | - Pan Huang
- Department of Stomatology, Taihe Hospital and Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei, 442000, P.R. China
- Laboratory of Tumor biology, Academy of Bio-Medicine Research, Hubei University of Medicine, Shiyan, Hubei, 442000, P.R. China
| | - Zidi Wang
- Laboratory of Tumor biology, Academy of Bio-Medicine Research, Hubei University of Medicine, Shiyan, Hubei, 442000, P.R. China
| | - Qiwei Guo
- Laboratory of Tumor biology, Academy of Bio-Medicine Research, Hubei University of Medicine, Shiyan, Hubei, 442000, P.R. China
| | - Congcong Huang
- Department of Stomatology, Taihe Hospital and Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei, 442000, P.R. China
- Laboratory of Tumor biology, Academy of Bio-Medicine Research, Hubei University of Medicine, Shiyan, Hubei, 442000, P.R. China
| | - Weidong Leng
- Department of Stomatology, Taihe Hospital and Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei, 442000, P.R. China.
| | - Shanshan Qin
- Department of Stomatology, Taihe Hospital and Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei, 442000, P.R. China.
- Laboratory of Tumor biology, Academy of Bio-Medicine Research, Hubei University of Medicine, Shiyan, Hubei, 442000, P.R. China.
| |
Collapse
|
5
|
Alsadat Mahmoudian R, Amirhosein M, Mahmoudian P, Fardi Golyan F, Mokhlessi L, Maftooh M, Khazaei M, Nassiri M, Mahdi Hassanian S, Ghayour-Mobarhan M, Ferns GA, Shahidsales S, Avan A. The therapeutic potential value of Cancer-testis antigens in immunotherapy of gastric cancer. Gene 2023; 853:147082. [PMID: 36464170 DOI: 10.1016/j.gene.2022.147082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 11/15/2022] [Accepted: 11/28/2022] [Indexed: 12/05/2022]
Abstract
Gastric cancer (GC) is the fourth most common cause of mortality and the fifth for incidence, globally. Diagnosis, early prognosis, and therapy remains challenging for this condition, and new tumor-associated antigens are required for its detection and immunotherapy. Cancer-testis antigens (CTAs) are a subfamily of tumor-associated antigens (TAAs) that have been identified as potential biomarkers and targets for cancer immunotherapy. The CTAs-restricted expression pattern in tumor cells and their potential immunogenicity identify them as attractive target candidates in CTA-based diagnosis or prognosis or immunotherapy. To date, numerous studies have reported the dysregulation of CTAs in GC. Several clinical trials have been done to assess CTA-based immunotherapeutic potential in the treatment of GC patients. NY-ESO-1, MAGE, and KK-LC-1 have been used in GC clinical trials. We review recent studies that have investigated the potential of the CTAs in GC regarding the expression, function, aggressive phenotype, prognosis, and immunological responses as well as their possible clinical significance as immunotherapeutic targets with a focus on challenges and future interventions.
Collapse
Affiliation(s)
- Reihaneh Alsadat Mahmoudian
- Cancer Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Basic Sciences Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Maharati Amirhosein
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Parvaneh Mahmoudian
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Fatemeh Fardi Golyan
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Leila Mokhlessi
- Centre for Biomedical Education and Research, Institute of Pharmacology and Toxicology, Witten/Herdecke University, Witten, Germany.
| | - Mina Maftooh
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Majid Khazaei
- Basic Sciences Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Mohammadreza Nassiri
- Recombinant Proteins Research Group, The Research Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran.
| | - Seyed Mahdi Hassanian
- Basic Sciences Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Majid Ghayour-Mobarhan
- Basic Sciences Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Gordon A Ferns
- Brighton & Sussex Medical School, Department of Medical Education, Falmer, Brighton, Sussex BN1 9PH, UK.
| | | | - Amir Avan
- Basic Sciences Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
6
|
Hoyos V, Vasileiou S, Kuvalekar M, Watanabe A, Tzannou I, Velazquez Y, French-Kim M, Leung W, Lulla S, Robertson C, Foreman C, Wang T, Bulsara S, Lapteva N, Grilley B, Ellis M, Osborne CK, Coscio A, Nangia J, Heslop HE, Rooney CM, Vera JF, Lulla P, Rimawi M, Leen AM. Multi-antigen-targeted T-cell therapy to treat patients with relapsed/refractory breast cancer. Ther Adv Med Oncol 2022; 14:17588359221107113. [PMID: 35860837 PMCID: PMC9290161 DOI: 10.1177/17588359221107113] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 05/25/2022] [Indexed: 11/16/2022] Open
Abstract
Purpose Adoptively transferred, ex vivo expanded multi-antigen-targeted T cells (multiTAA-T) represent a new, potentially effective, and nontoxic therapeutic approach for patients with breast cancer (BC). In this first-in-human trial, we investigated the safety and clinical effects of administering multiTAA T cells targeting the tumor-expressed antigens, Survivin, NY-ESO-1, MAGE-A4, SSX2, and PRAME, to patients with relapsed/refractory/metastatic BC. Materials and methods MultiTAA T-cell products were generated from the peripheral blood of heavily pre-treated patients with metastatic or locally recurrent unresectable BC of all subtypes and infused at a fixed dose level of 2 × 107/m2. Patients received two infusions of cells 4 weeks apart and safety and clinical activity were determined. Cells were administered in an outpatient setting and without prior lymphodepleting chemotherapy. Results All patients had estrogen receptor/progesterone receptor positive BC, with one patient also having human epidermal growth factor receptor 2-positive. There were no treatment-related toxicities and the infusions were well tolerated. Of the 10 heavily pre-treated patients enrolled and infused with multiTAA T cells, nine had disease progression while one patient with 10 lines of prior therapies experienced prolonged (5 months) disease stabilization that was associated with the in vivo expansion and persistence of T cells directed against the targeted antigens. Furthermore, antigen spreading and the endogenous activation of T cells directed against a spectrum of non-targeted tumor antigens were observed in 7/10 patients post-multiTAA infusion. Conclusion MultiTAA T cells were well tolerated and induced disease stabilization in a patient with refractory BC. This was associated with in vivo T-cell expansion, persistence, and antigen spreading. Future directions of this approach may include additional strategies to enhance the therapeutic benefit of multiTAA T cells in patients with BC.
Collapse
Affiliation(s)
- Valentina Hoyos
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children’s Hospital and Houston Methodist Hospital, Houston, TX, USA Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, 1102 Bates Ave, Feigin Center 17th Floor. Houston, TX 77030, USA
| | - Spyridoula Vasileiou
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children’s Hospital and Houston Methodist Hospital, Houston, TX, USA
| | - Manik Kuvalekar
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children’s Hospital and Houston Methodist Hospital, Houston, TX, USA
| | - Ayumi Watanabe
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children’s Hospital and Houston Methodist Hospital, Houston, TX, USA
| | - Ifigeneia Tzannou
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children’s Hospital and Houston Methodist Hospital, Houston, TX, USA
| | - Yovana Velazquez
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children’s Hospital and Houston Methodist Hospital, Houston, TX, USA
| | - Matthew French-Kim
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children’s Hospital and Houston Methodist Hospital, Houston, TX, USA
| | - Wingchi Leung
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children’s Hospital and Houston Methodist Hospital, Houston, TX, USA
| | - Suhasini Lulla
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children’s Hospital and Houston Methodist Hospital, Houston, TX, USA
| | - Catherine Robertson
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children’s Hospital and Houston Methodist Hospital, Houston, TX, USA
| | - Claudette Foreman
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Tao Wang
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Shaun Bulsara
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Natalia Lapteva
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children’s Hospital and Houston Methodist Hospital, Houston, TX, USA
| | - Bambi Grilley
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children’s Hospital and Houston Methodist Hospital, Houston, TX, USA
| | - Matthew Ellis
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Charles Kent Osborne
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Angela Coscio
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Julie Nangia
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Helen E. Heslop
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children’s Hospital and Houston Methodist Hospital, Houston, TX, USA
| | - Cliona M. Rooney
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children’s Hospital and Houston Methodist Hospital, Houston, TX, USA
| | - Juan F. Vera
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children’s Hospital and Houston Methodist Hospital, Houston, TX, USA
| | - Premal Lulla
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children’s Hospital and Houston Methodist Hospital, Houston, TX, USA
| | - Mothaffar Rimawi
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Lester and Sue Smith Breast Center, Baylor College of Medicine
| | - Ann M. Leen
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children’s Hospital and Houston Methodist Hospital, Houston, TX, USA
| |
Collapse
|
7
|
Lingg L, Rottenberg S, Francica P. Meiotic Genes and DNA Double Strand Break Repair in Cancer. Front Genet 2022; 13:831620. [PMID: 35251135 PMCID: PMC8895043 DOI: 10.3389/fgene.2022.831620] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 02/02/2022] [Indexed: 12/16/2022] Open
Abstract
Tumor cells show widespread genetic alterations that change the expression of genes driving tumor progression, including genes that maintain genomic integrity. In recent years, it has become clear that tumors frequently reactivate genes whose expression is typically restricted to germ cells. As germ cells have specialized pathways to facilitate the exchange of genetic information between homologous chromosomes, their aberrant regulation influences how cancer cells repair DNA double strand breaks (DSB). This drives genomic instability and affects the response of tumor cells to anticancer therapies. Since meiotic genes are usually transcriptionally repressed in somatic cells of healthy tissues, targeting aberrantly expressed meiotic genes may provide a unique opportunity to specifically kill cancer cells whilst sparing the non-transformed somatic cells. In this review, we highlight meiotic genes that have been reported to affect DSB repair in cancers derived from somatic cells. A better understanding of their mechanistic role in the context of homology-directed DNA repair in somatic cancers may provide useful insights to find novel vulnerabilities that can be targeted.
Collapse
Affiliation(s)
- Lea Lingg
- Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
- Cancer Therapy Resistance Cluster, Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Sven Rottenberg
- Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
- Cancer Therapy Resistance Cluster, Department for BioMedical Research, University of Bern, Bern, Switzerland
- Bern Center for Precision Medicine, University of Bern, Bern, Switzerland
- *Correspondence: Sven Rottenberg, ; Paola Francica,
| | - Paola Francica
- Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
- Cancer Therapy Resistance Cluster, Department for BioMedical Research, University of Bern, Bern, Switzerland
- *Correspondence: Sven Rottenberg, ; Paola Francica,
| |
Collapse
|
8
|
Lai JP, Rosenberg AZ, Miettinen MM, Lee CCR. NY-ESO-1 expression in sarcomas: A diagnostic marker and immunotherapy target. Oncoimmunology 2021; 1:1409-1410. [PMID: 23243610 PMCID: PMC3518519 DOI: 10.4161/onci.21059] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
NY-ESO-1 (CTAG 1B) is highly expressed in the majority of synovial sarcomas and myxoid/round cell liposarcomas as well as in a subset of melanomas, but only rarely in other mesenchymal tumors. This points to a potential for using NY-ESO-1 in the differential diagnosis of these lesions. Furthermore, promising results have been obtained in clinical trials testing NY-ESO-1-targeted immunotherapy in subsets of melanoma and synovial sarcoma patients.
Collapse
Affiliation(s)
- Jin-Ping Lai
- Laboratory of Pathology; National Cancer Institute; NIH; Bethesda, MD USA
| | | | | | | |
Collapse
|
9
|
Hosoya N, Miyagawa K. Synaptonemal complex proteins modulate the level of genome integrity in cancers. Cancer Sci 2021; 112:989-996. [PMID: 33382503 PMCID: PMC7935773 DOI: 10.1111/cas.14791] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 12/25/2020] [Accepted: 12/28/2020] [Indexed: 01/09/2023] Open
Abstract
The synaptonemal complex (SC) is a proteinaceous structure that is transiently formed during meiosis to promote homologous recombination between maternal and paternal chromosomes. As this structure is required only for meiotic recombination, the proteins constituting the complex are almost undetectable in normal somatic cells, but they can be expressed under the conditions in which the transcriptional machinery is deregulated. Accumulating evidence indicates that they are epigenetically expressed in cancers of various origin. Not surprisingly, in contrast to their meiotic roles, the somatic roles of the SC proteins remain to be investigated. However, it has recently been reported that SYCP3 and SYCE2 control DNA double‐strand break repair negatively and positively, respectively, suggesting that the ectopic expression of the SC proteins in somatic cells could be associated with the maintenance of genomic instability. Thus, it is highly likely that the investigation of the somatic roles of the SC proteins would improve our understanding of the mechanisms underlying tumor development.
Collapse
Affiliation(s)
- Noriko Hosoya
- Laboratory of Molecular Radiology, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Kiyoshi Miyagawa
- Laboratory of Molecular Radiology, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
10
|
Gjerstorff MF. Novel Insights Into Epigenetic Reprogramming and Destabilization of Pericentromeric Heterochromatin in Cancer. Front Oncol 2020; 10:594163. [PMID: 33251148 PMCID: PMC7674669 DOI: 10.3389/fonc.2020.594163] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 10/15/2020] [Indexed: 12/17/2022] Open
Abstract
Pericentromeric heterochromatin is maintained in a condensed structure by repressive epigenetic control mechanisms and perturbation of these may cause diseases. The chromosome 1q12 region harbors the largest pericentromeric heterochromatin domain in the genome and is among the most common breakpoints in both solid and hematopoietic cancers. Furthermore, the 1q arm is frequently amplified in cancer and this may support tumorigenesis by increasing the dosage of the many oncogenes of this genomic region. Recent studies have provided insight into the mechanisms leading to loss of 1q12 stability and 1q amplification and DNA hypomethylation seems to play a prominent role. This may be the result of decreased activity of DNA methyltransferases and instrumental for 1q12 destabilization or arise secondary to perturbation of other important epigenetic mechanisms that control repression of pericentromeric heterochromatin. Polycomb proteins were recently demonstrated to epigenetically reprogram demethylated 1q12 pericentromeric heterochromatin in premalignant and malignant cells to form large subnuclear structures known as polycomb bodies. This may influence the regulation and stability of 1q12 pericentromeric heterochromatin and/or the distribution of polycomb factors to support tumorigenesis. This review will discuss recent insight into the epigenetic perturbations causing the destabilization of 1q12 pericentromeric heterochromatin and its possible implications for tumor biology.
Collapse
Affiliation(s)
- Morten Frier 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
|
11
|
Safavi A, Kefayat A, Mahdevar E, Ghahremani F, Nezafat N, Modarressi MH. Efficacy of co-immunization with the DNA and peptide vaccines containing SYCP1 and ACRBP epitopes in a murine triple-negative breast cancer model. Hum Vaccin Immunother 2020; 17:22-34. [PMID: 32497486 DOI: 10.1080/21645515.2020.1763693] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Multiepitope cancer vaccines have gained lots of attention for prophylactic and therapeutic purposes in cancer patients. In our previous study, multiepitope DNA and peptide cancer vaccines consisted of the most immunodominant epitopes of ACRBP and SYCP1 antigens were designed by bioinformatic tools. In this study, the effect of prophylactic co-immunization with these DNA and peptide cancer vaccines in the 4T1 breast cancer animal model was assessed. Serum levels of the peptide-specific IgG total, IgG2a and IgG1 were measured by enzyme-linked immunosorbent assay (ELISA). Also, the efficacy of the immunized mice splenocytes' for producing interleukin-4 (IL-4) and interferon-γ (IFN-γ) was evaluated. The co-immunization caused a significant (P < .05) increase in the serum levels of IgG1 and IgG2a. The co-immunized mice splenocytes exhibited significantly enhanced IL-4 (6.6-fold) and IFN-γ (19-fold) production. Also, their lymphocytes exhibited higher proliferation rate (3-fold) and granzyme B production (6.5-fold) in comparison with the control. The prophylactic co-immunization significantly decreased the breast tumors' volume (78%) and increased the tumor-bearing mice survival time (37.5%) in comparison with the control. Taking together, prophylactic co-immunization with these multiepitope DNA and peptide cancer vaccines can activate the immune system against breast cancer. However, further experiments are needed to evaluate their efficacy from different angles.
Collapse
Affiliation(s)
- Ashkan Safavi
- Department of Biology, Science and Research Branch, Islamic Azad University , Tehran, Iran
| | - Amirhosein Kefayat
- Department of Oncology, Cancer Prevention Research Center, Isfahan University of Medical Sciences , Isfahan, Iran
| | - Elham Mahdevar
- Department of Biology, Faculty of Science and Engineering, Science and Arts University , Yazd, Iran
| | - Fatemeh Ghahremani
- Department of Medical Physics and Radiotherapy, Arak School of Paramedicine, Arak University of Medical Sciences , Arak, Iran
| | - Navid Nezafat
- Pharmaceutical Sciences Research Center, Faculty of Pharmacy, Shiraz University of Medical Sciences , Shiraz, Iran
| | | |
Collapse
|
12
|
Gantchev J, Martínez Villarreal A, Gunn S, Zetka M, Ødum N, Litvinov IV. The ectopic expression of meiCT genes promotes meiomitosis and may facilitate carcinogenesis. Cell Cycle 2020; 19:837-854. [PMID: 32223693 DOI: 10.1080/15384101.2020.1743902] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Cancer meiomitosis is defined as the concurrent activation of both mitotic and meiotic machineries in neoplastic cells that confer a selective advantage together with increased genomic instability. MeiCT (meiosis-specific cancer/testis) genes that perform specialized functions in the germline events required for the first meiotic division are ectopically expressed in several cancers. Here we describe the expression profiles of meiCT genes and proteins across a number of cancers and review the proposed mechanisms that increase aneuploidy and elicit reduction division in polyploid cells. These mechanisms are centered on the overexpression and function of meiCT proteins in cancers under various conditions that includes a response to genotoxic stress. Since meiCT genes are transcriptionally repressed in somatic cells, their target offers a promising therapeutic approach with limited toxicity to healthy tissues. Throughout the review, we provide a detailed description of the roles for each gene in the context of meiosis and we discuss proposed functions and outcomes resulting from their ectopic reactivation in cancer.
Collapse
Affiliation(s)
- Jennifer Gantchev
- Division of Dermatology, Department of Medicine, McGill University Health Centre, Montreal, QC, Canada
| | | | - Scott Gunn
- Division of Dermatology, Department of Medicine, McGill University Health Centre, Montreal, QC, Canada
| | - Monique Zetka
- Department of Biology, McGill University, Montreal, QC, Canada
| | - Neils Ødum
- Department of Microbiology and Immunology, The University of Copenhagen, Copenhagen, Denmark
| | - Ivan V Litvinov
- Division of Dermatology, Department of Medicine, McGill University Health Centre, Montreal, QC, Canada
| |
Collapse
|
13
|
Wang Q, Lu P, Wang T, Zheng Q, Li Y, Leng SX, Meng X, Wang B, Xie J, Zhang H. Sitagliptin affects gastric cancer cells proliferation by suppressing Melanoma-associated antigen-A3 expression through Yes-associated protein inactivation. Cancer Med 2020; 9:3816-3828. [PMID: 32227453 PMCID: PMC7286447 DOI: 10.1002/cam4.3024] [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: 12/02/2019] [Revised: 02/18/2020] [Accepted: 03/08/2020] [Indexed: 12/18/2022] Open
Abstract
Sitagliptin is an emerging oral hypoglycemic agent that inhibits the development of a wide variety of tumors. Current researches indicate that the abnormal activation of Yes-associated protein (YAP) promotes the proliferation and poor prognosis of multiple tumors. However, the ability of sitagliptin to regulate YAP and its effects on gastric cancer (GC) cells remain unclear. Here, we first showed that sitagliptin inhibited the proliferation of GC cells, and this inhibition was regulated by Hippo pathway. Sitagliptin phosphorylated YAP in a large tumor suppressor homolog-dependent manner, thereby inhibiting YAP nuclear translocation, and promoted YAP cytoplasm retention. This inhibition can be blocked by adenosine 5'-monophosphate-activated protein kinase (AMPK). Moreover, sitagliptin could reduce the expression of tumor-testis antigen Melanoma-associated antigen-A3 through YAP. In conclusion, sitagliptin may have a potential inhibitory effect on GC by AMPK/YAP/melanoma-associated antigen-A3 pathway.
Collapse
Affiliation(s)
- Qi Wang
- Department of Geriatrics, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Pan Lu
- Department of Urinary Surgery, The Central Hospital of Xiaogan, Xiaogan, Hubei, China
| | - Tao Wang
- Department of Pathology, Shenyang KingMed Center for Clinical Laboratory Co., Ltd., Shenyang, Liaoning, China
| | - Qianqian Zheng
- Department of Pathophysiology, College of Basic Medical Science, China Medical University, Shenyang, Liaoning, China
| | - Yan Li
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Sean X Leng
- Division of Geriatric Medicine and Gerontology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Xin Meng
- Department of Biochemistry and Molecular Biology, School of Life Sciences, China Medical University, Shenyang, Liaoning, China
| | - Biao Wang
- Department of Biochemistry and Molecular Biology, School of Life Sciences, China Medical University, Shenyang, Liaoning, China
| | - Jisheng Xie
- Department of Histology and Embryology, Youjiang Medical College for Nationalities, Baise, Guangxi, China
| | - Haiyan Zhang
- Department of Geriatrics, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
14
|
Wu C, Tuo Y. SYCP2 expression is a novel prognostic biomarker in luminal A/B breast cancer. Future Oncol 2019; 15:817-826. [PMID: 30511892 DOI: 10.2217/fon-2018-0821] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: To explore the prognostic value of synaptonemal complex protein-2 (SYCP2) in different subtypes of breast cancer. Patients & materials: In silico bioinformatic analysis was conducted using large databases. Results: SYCP2 was only significantly upregulated in luminal B tumors compared with the adjacent normal tissues. SYCP2 expression was an independent indicator of shorter overall survival in luminal A patients (hazard ratio: 2.269; 95% CI: 1.059–4.862; p = 0.035) and luminal B patients (hazard ratio: 2.546; 95% CI: 1.020–6.355; p = 0.045). Its expression was negatively correlated with the methylation status of multiple CpG sites (cg22214414, cg23241473 and cg07347645) in both luminal A and luminal B tumors. Conclusion: Increased SYCP2 expression might be an independent indicator of shorter overall survival in both luminal A and luminal B patients.
Collapse
Affiliation(s)
- Chihua Wu
- Department of Breast Surgery, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Clinical Medicine, University of Electronic Science & Technology of China, Chengdu, 610072, Sichuan, PR China
| | - Youlin Tuo
- Department of Breast Surgery, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Clinical Medicine, University of Electronic Science & Technology of China, Chengdu, 610072, Sichuan, PR China
| |
Collapse
|
15
|
Zhang Y, Zhang Y, Zhang L. Expression of cancer-testis antigens in esophageal cancer and their progress in immunotherapy. J Cancer Res Clin Oncol 2019; 145:281-291. [PMID: 30656409 PMCID: PMC6373256 DOI: 10.1007/s00432-019-02840-3] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 01/03/2019] [Indexed: 12/17/2022]
Abstract
PURPOSE Esophageal cancer is a common disease in China with low survival rate due to no obvious early symptoms and lack of effective screening strategies. Traditional treatments usually do not produce desirable results in patients with advanced esophageal cancer, so immunotherapy which relies on tumor-related antigens is needed to combat low survival rates effectively. Cancer-testis antigens (CTA), a large family of tumor-related antigens, have a strong in vivo immunogenicity and tumor-restricted expressing patterns in normal adult tissues. These two characteristics are ideal features of anticancer immunotherapy targets and, therefore, promoted the development of some studies of CTA-based therapy. To provide ideas for the role of the cancer-testis antigens MAGE-A, NY-ESO-1, LAGE-1, and TTK in esophageal cancer, we summarized their expression, prognostic value, and development in immunotherapy. METHODS The relevant literature from PubMed is reviewed in this study. RESULTS In esophageal cancer, although the relationship between expression of MAGE-A, NY-ESO-1, LAGE-1, and TTK and prognosis value is still in a controversial situation, MAGE-A, NY-ESO-1, LAGE-1, and TTK are highly expressed and can induce specific CTL cells to produce particular killing effect on tumor cells, and some clinical trials have demonstrated that immunotherapy for esophageal cancer patients is effective and safe, which provides a new therapeutic strategy for the treatment of esophageal cancer in the future. CONCLUSION In this review, we summarize expression and prognostic value of MAGE-A, NY-ESO-1, LAGE-1, and TTK in esophageal cancer and point out recent advances in immunotherapy about them.
Collapse
Affiliation(s)
- Yujie Zhang
- Department of Oncology, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan, 430030, Hubei, China
| | - Yuxin Zhang
- Hepatic Surgery Center, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan, 430030, Hubei, China
| | - Li Zhang
- Department of Oncology, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan, 430030, Hubei, China.
| |
Collapse
|
16
|
Schlößer HA, Thelen M, Lechner A, Wennhold K, Garcia-Marquez MA, Rothschild SI, Staib E, Zander T, Beutner D, Gathof B, Gilles R, Cukuroglu E, Göke J, Shimabukuro-Vornhagen A, Drebber U, Quaas A, Bruns CJ, Hölscher AH, Von Bergwelt-Baildon MS. B cells in esophago-gastric adenocarcinoma are highly differentiated, organize in tertiary lymphoid structures and produce tumor-specific antibodies. Oncoimmunology 2018; 8:e1512458. [PMID: 30546950 PMCID: PMC6287776 DOI: 10.1080/2162402x.2018.1512458] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 08/09/2018] [Accepted: 08/11/2018] [Indexed: 12/29/2022] Open
Abstract
Tumor-infiltrating lymphocytes (TILs) are correlated to prognosis of several kinds of cancer. Most studies focused on T cells, while the role of tumor-associated B cells (TABs) has only recently gained more attention. TABs contain subpopulations with distinct functions, potentially promoting or inhibiting immune responses. This study provides a detailed analysis of TABs in gastro-esophageal adenocarcinoma (EAC). Flow cytometric analyses of single cell suspensions of tumor samples, mucosa, lymph nodes and peripheral blood mononuclear cells (PBMC) of EAC patients and healthy controls revealed a distinct B cell compartment in cancer patients. B cells were increased in tumor samples and subset-analyses of TILs showed increased proportions of differentiated and activated B cells and an enrichment for follicular T helper cells. Confocal microscopy demonstrated that TABs were mainly organized in tertiary lymphoid structures (TLS), which resemble lymphoid follicles in secondary lymphoid organs. A panel of 34 tumor-associated antigens (TAAs) expressed in EAC was identified based on public databases and TCGA data to analyze tumor-specific B cell responses using a LUMINEXTM bead assay and flow cytometry. Structural analyses of TLS and the detection of tumor-specific antibodies against one or more TAAs in 48.1% of analyzed serum samples underline presence of anti-tumor B cell responses in EAC. Interestingly, B cells were decreased in tumors with expression of Programmed Death Ligand 1 or impaired HLA-I expression. These data demonstrate that anti-tumor B cell responses are an additional and underestimated aspect of EAC. Our results are of immediate translational relevance to emerging immunotherapies.
Collapse
Affiliation(s)
- Hans A. Schlößer
- Department of General, Visceral and Cancer Surgery, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
| | - Martin Thelen
- Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
| | - Axel Lechner
- Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
- Department of Head and Neck Surgery, University of Göttingen, Göttingen, Germany
| | - Kerstin Wennhold
- Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
| | | | | | - Elena Staib
- Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
| | - Thomas Zander
- Department I of Internal Medicine I, University of Cologne, Cologne, Germany
| | - Dirk Beutner
- Department of Head and Neck Surgery, University of Göttingen, Göttingen, Germany
| | - Birgit Gathof
- Institute of Transfusion Medicine, University of Cologne, Cologne, Germany
| | - Ramona Gilles
- Institute of Transfusion Medicine, University of Cologne, Cologne, Germany
| | | | | | | | - Uta Drebber
- Institute of Pathology, University of Cologne, Cologne, Germany
| | - Alexander Quaas
- Institute of Pathology, University of Cologne, Cologne, Germany
| | - Christiane J. Bruns
- Department of General, Visceral and Cancer Surgery, University of Cologne, Cologne, Germany
| | - Arnulf H. Hölscher
- Department of General, Visceral and Cancer Surgery, University of Cologne, Cologne, Germany
| | - Michael S. Von Bergwelt-Baildon
- German Cancer Consortium (DKTK), Heidelberg, Germany
- Department of Internal Medicine III, University Hospital, Munich, Germany
| |
Collapse
|
17
|
In Silico Analysis of Synaptonemal Complex Protein 1 (SYCP1) and Acrosin Binding Protein (ACRBP) Antigens to Design Novel Multiepitope Peptide Cancer Vaccine Against Breast Cancer. Int J Pept Res Ther 2018. [DOI: 10.1007/s10989-018-9780-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
18
|
Gordeeva O. Cancer-testis antigens: Unique cancer stem cell biomarkers and targets for cancer therapy. Semin Cancer Biol 2018; 53:75-89. [PMID: 30171980 DOI: 10.1016/j.semcancer.2018.08.006] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2018] [Revised: 08/15/2018] [Accepted: 08/17/2018] [Indexed: 02/07/2023]
Abstract
Cancer-testis antigens (CTAs) are considered as unique and promising cancer biomarkers and targets for cancer therapy. CTAs are multifunctional protein group with specific expression patterns in normal embryonic and adult cells and various types of cancer cells. CTAs are involved in regulating of the basic cellular processes during development, stem cell differentiation and carcinogenesis though the biological roles and cell functions of CTA families remain largely unclear. Analysis of CTA expression patterns in embryonic germ and somatic cells, pluripotent and multipotent stem cells, cancer stem cells and their cell descendants indicates that rearrangements of characteristic CTA profiles (aberrant expression) could be associated with cancer transformation and failure of the developmental program of cell lineage specification and germ line restriction. Therefore, aberrant CTA profiles can be used as panels of biomarkers for diagnoses and the selection of cancer treatment strategies. Moreover, immunogenic CTAs are prospective targets for cancer immunotherapy. Clinical trials testing broad range of cancer therapeutic vaccines against antigens of MAGEA and NY-ESO-1 families for treating various cancers have shown mixed clinical efficiency, safety and tolerability, suggesting the requirement of in-depth research of CTA expression in normal and cancer stem cells and extensive clinical trials for improving cancer immunotherapy technologies. This review focuses on recent advancement in study of CTAs in normal and cancer cells, particularly in normal and cancer stem cells, and provides a new insight into CTA expression patterns during normal and cancer stem cell lineage development. Additionally, new approaches in development of effective CTA-based therapies exclusively targeting cancer stem cells will be discussed.
Collapse
Affiliation(s)
- Olga Gordeeva
- Laboratory of Cell and Molecular Mechanisms of Histogenesis, Kol'tsov Institute of Developmental Biology, Russian Academy of Sciences, 26 Vavilov Street, Moscow, 119334, Russia.
| |
Collapse
|
19
|
Szaryńska M, Olejniczak A, Kobiela J, Łaski D, Śledziński Z, Kmieć Z. Cancer stem cells as targets for DC-based immunotherapy of colorectal cancer. Sci Rep 2018; 8:12042. [PMID: 30104575 PMCID: PMC6089981 DOI: 10.1038/s41598-018-30525-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 08/01/2018] [Indexed: 12/11/2022] Open
Abstract
The therapy of colorectal cancer (CRC) patients is often unsuccessful because of the presence of cancer stem cells (CSCs) resistant to conventional approaches. Dendritic cells (DC)-based protocols are believed to effectively supplement CRC therapy. Our study was aimed to assess how the number and properties of CSCs isolated from tumor tissue of CRC patients will affect the biological characteristics of in vitro modified DCs. Similar procedures were conducted with the using of CRC HCT116 and HT29 cell lines. We found that the detailed configuration of CSC-like markers significantly influenced the maturation and activation of DCs after stimulation with cancer cells lysates or culture supernatants. This basic stimulatory effect was enhanced by LPS that is normally present in CRC CSCs niche. The increased number of CD29+ and CD44+ CSCs presented the opposite impact on treated DCs as showed by many significant correlations. The CD133+ CSCs seemed to impair the functions of DCs. The more CD133+ CSCs in tumor sample the lower number of activated DCs evidenced after stimulation. Moreover, our results showed superiority of the spherical culture model over the adherent one since spherical HCT116 and HT29 cells presented similar influence on DCs properties as CRC patients cancer cells. We concluded that the DCs features may depend directly on the properties of CSCs affected by progression status of tumor.
Collapse
Affiliation(s)
- Magdalena Szaryńska
- Department of Histology, Medical University of Gdansk, 80-210, Gdansk, Poland.
| | - Agata Olejniczak
- Department of Histology, Medical University of Gdansk, 80-210, Gdansk, Poland
| | - Jarosław Kobiela
- Department of General, Endocrine and Transplant Surgery, Medical University of Gdansk, 80-214, Gdansk, Poland
| | - Dariusz Łaski
- Department of General, Endocrine and Transplant Surgery, Medical University of Gdansk, 80-214, Gdansk, Poland
| | - Zbigniew Śledziński
- Department of General, Endocrine and Transplant Surgery, Medical University of Gdansk, 80-214, Gdansk, Poland
| | - Zbigniew Kmieć
- Department of Histology, Medical University of Gdansk, 80-210, Gdansk, Poland
| |
Collapse
|
20
|
Wierinckx A, Delgrange E, Bertolino P, François P, Chanson P, Jouanneau E, Lachuer J, Trouillas J, Raverot G. Sex-Related Differences in Lactotroph Tumor Aggressiveness Are Associated With a Specific Gene-Expression Signature and Genome Instability. Front Endocrinol (Lausanne) 2018; 9:706. [PMID: 30555413 PMCID: PMC6283894 DOI: 10.3389/fendo.2018.00706] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Accepted: 11/09/2018] [Indexed: 12/24/2022] Open
Abstract
Sex-related differences have been reported in various cancers, in particular men with lactotroph tumors have a worse prognosis than women. While the underlying mechanism of this sexual dimorphism remains unclear, it has been suggested that a lower estrogen receptor alpha expression may drive the sex differences observed in aggressive and malignant lactotroph tumors that are resistant to dopamine agonists. Based on this observation, we aimed to explore the molecular importance of the estrogen pathway through a detailed analysis of the transcriptomic profile of lactotroph tumors from 20 men and 10 women. We undertook gene expression analysis of the selected lactotroph tumors following their pathological grading using the five-tiered classification. Chromosomic alterations were further determined in 13 tumors. Functional analysis showed that there were differences between tumors from men and women in gene signatures associated with cell morphology, cell growth, cell proliferation, development, and cell movement. Hundred-forty genes showed an increased or decreased expression with a minimum 2-fold change. A large subset of those genes belonged to the estrogen receptor signaling pathway, therefore confirming the potent role of this pathway in lactotroph tumor sex-associated aggressiveness. Genes belonging to the X chromosome, such as CTAG2, FGF13, and VEGF-D, were identified as appealing candidates with a sex-linked dysregulation in lactotroph tumors. Through our comparative genomic hybridization analyses (CGH), chromosomic gain, in particular chromosome 19p, was found only in tumors from men, while deletion of chromosome 11 was sex-independent, as it was found in most (5/6) of the aggressive and malignant tumors. Comparison of transcriptomic and CGH analysis revealed four genes (CRB3, FAM138F, MATK, and STAP2) located on gained regions of chromosome 19 and upregulated in lactotroph tumors from men. MATK and STAP2 are both implicated in cell growth and are reported to be associated with the estrogen signaling pathway. Our work confirms the proposed involvement of the estrogen signaling pathway in favoring the increased aggressiveness of lactotroph tumors in men. More importantly, we highlight a number of ER-related candidate genes and further identify a series of target molecules with sex-specific expression that could contribute to the aggressive behavior of lactotroph tumors in men.
Collapse
Affiliation(s)
- Anne Wierinckx
- Institut Universitaire de Technologie, Université Lyon 1, Université de Lyon, Lyon, France
- Centre de Recherche en Cancérologie de Lyon (CRCL), INSERM U1052, CNRS UMR5286, Université de Lyon, Lyon, France
- ProfileXpert, SFR-Est, CNRS UMR-S3453, INSERM US7, Lyon, France
- *Correspondence: Anne Wierinckx
| | - Etienne Delgrange
- Service d'Endocrinologie, CHU UCL Namur, Université catholique de Louvain, Ottignies-Louvain-la-Neuve, Belgium
| | - Philippe Bertolino
- Centre de Recherche en Cancérologie de Lyon (CRCL), INSERM U1052, CNRS UMR5286, Université de Lyon, Lyon, France
| | | | - Philippe Chanson
- Service d'Endocrinologie et des Maladies de la Reproduction, Assistance Publique-Hôpitaux de Paris, Centre de Référence des Maladies Rares de l'Hypophyse, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
- Faculté de Médecine Paris-Sud, UMR S-1185, Université Paris-Sud, Université Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Emmanuel Jouanneau
- Service de Neurochirurgie Groupement Hospitalier Est, Hospices Civils de Lyon, Bron, France
- Faculté de Médecine Lyon-Est, Université Lyon 1, Université de Lyon, Lyon, France
| | - Joël Lachuer
- Institut Universitaire de Technologie, Université Lyon 1, Université de Lyon, Lyon, France
- Centre de Recherche en Cancérologie de Lyon (CRCL), INSERM U1052, CNRS UMR5286, Université de Lyon, Lyon, France
- ProfileXpert, SFR-Est, CNRS UMR-S3453, INSERM US7, Lyon, France
| | - Jacqueline Trouillas
- Institut Universitaire de Technologie, Université Lyon 1, Université de Lyon, Lyon, France
- Faculté de Médecine Lyon-Est, Université Lyon 1, Université de Lyon, Lyon, France
| | - Gérald Raverot
- Centre de Recherche en Cancérologie de Lyon (CRCL), INSERM U1052, CNRS UMR5286, Université de Lyon, Lyon, France
- Faculté de Médecine Lyon-Est, Université Lyon 1, Université de Lyon, Lyon, France
- Département d'Endocrinologie, Centre de Référence pour les Maladies Hypophysaires Rares (HYPO), Groupement Hospitalier EST, Hospices Civils de Lyon, Université de Lyon, Lyon, France
| |
Collapse
|
21
|
Futawatari N, Fukuyama T, Yamamura R, Shida A, Takahashi Y, Nishi Y, Ichiki Y, Kobayashi N, Yamazaki H, Watanabe M. Early gastric cancer frequently has high expression of KK-LC-1, a cancer-testis antigen. World J Gastroenterol 2017; 23:8200-8206. [PMID: 29290656 PMCID: PMC5739926 DOI: 10.3748/wjg.v23.i46.8200] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Revised: 10/30/2017] [Accepted: 11/14/2017] [Indexed: 02/07/2023] Open
Abstract
AIM To assess cancer-testis antigens (CTAs) expression in gastric cancer patients and examined their associations with clinicopathological factors.
METHODS Eighty-three gastric cancer patients were evaluated in this study. Gastric cancer specimens were evaluated for the gene expression of CTAs, Kitakyushu lung cancer antigen-1 (KK-LC-1), melanoma antigen (MAGE)-A1, MAGE-A3 and New York esophageal cancer-1 (NY-ESO-1), by reverse transcription PCR. Clinicopathological background information, such as gender, age, tumor size, macroscopic type, tumor histology, depth of invasion, lymph node metastasis, lymphatic invasion, venous invasion, and pathological stage, was obtained. Statistical comparisons between the expression of each CTA and each clinicopathological background were performed using the χ2 test.
RESULTS The expression rates of KK-LC-1, MAGE-A1, MAGE-A3, and NY-ESO-1 were 79.5%, 32.5%, 39.8%, and 15.7%, respectively. In early stage gastric cancer specimens, the expression of KK-LC-1 was 79.4%, which is comparable to the 79.6% observed in advanced stage specimens. The expression of KK-LC-1 was not significantly associated with clinicopathological factors, while there were considerable differences in the expression rates of MAGE-A1 and MAGE-A3 with vs without lymphatic invasion (MAGE-A1, 39.3% vs 13.6%, P = 0.034; MAGE-A3, 47.5% vs 18.2%, P = 0.022) and/or vascular invasion (MAGE-A1, 41.5% vs 16.7%, P = 0.028; MAGE-A3, 49.1% vs 23.3%, P = 0.035) and, particularly, MAGE-A3, in patients with early vs advanced stage (36.5% vs 49.0%, P = 0.044), respectively. Patients expressing MAGE-A3 and NY-ESO-1 were older than those not expressing MAGE-A3 and NY-ESO-1 (MAGE-A3, 73.7 ± 7.1 vs 67.4 ± 12.3, P = 0.009; NY-ESO-1, 75.5 ± 7.2 vs 68.8 ± 11.2, P = 0.042).
CONCLUSION The KK-LC-1 expression rate was high even in patients with stage I cancer, suggesting that KK-LC-1 is a useful biomarker for early diagnosis of gastric cancer.
Collapse
Affiliation(s)
- Nobue Futawatari
- Department of Surgery, Sagamihara National Hospital, Sagamihara, Kanagawa 252-0392, Japan
- Department of Surgery, School of Medicine, Kitasato University, Sagamihara, Kanagawa 252-0374, Japan
| | - Takashi Fukuyama
- Division of Biomedical Research, Kitasato University Medical Center, Kitamoto, Saitama 364-8501, Japan
| | - Rui Yamamura
- Division of Biomedical Research, Kitasato University Medical Center, Kitamoto, Saitama 364-8501, Japan
| | - Akiko Shida
- Department of Surgery, School of Medicine, Kitasato University, Sagamihara, Kanagawa 252-0374, Japan
| | - Yoshihito Takahashi
- Department of Surgery, Kitasato University Medical Center, Kitamoto, Saitama 364-8501, Japan
| | - Yatsushi Nishi
- Department of Surgery, Kitasato University Medical Center, Kitamoto, Saitama 364-8501, Japan
| | - Yoshinobu Ichiki
- Second Department of Surgery, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Fukuoka 807-8555, Japan
| | - Noritada Kobayashi
- Division of Biomedical Research, Kitasato University Medical Center, Kitamoto, Saitama 364-8501, Japan
| | - Hitoshi Yamazaki
- Department of Pathology, Kitasato University Medical Center, Kitamoto, Saitama 364-8501, Japan
| | - Masahiko Watanabe
- Department of Surgery, School of Medicine, Kitasato University, Sagamihara, Kanagawa 252-0374, Japan
| |
Collapse
|
22
|
Szender JB, Papanicolau-Sengos A, Eng KH, Miliotto AJ, Lugade AA, Gnjatic S, Matsuzaki J, Morrison CD, Odunsi K. NY-ESO-1 expression predicts an aggressive phenotype of ovarian cancer. Gynecol Oncol 2017; 145:420-425. [PMID: 28392127 DOI: 10.1016/j.ygyno.2017.03.509] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2016] [Revised: 03/11/2017] [Accepted: 03/29/2017] [Indexed: 10/19/2022]
Abstract
OBJECTIVES NY-ESO-1 is a cancer testis antigen and a promising target for immunotherapy. The purpose of this study was to determine the expression frequency, immunogenicity, and clinical impact of NY-ESO-1 in ovarian cancer. METHODS Immunohistochemistry (IHC), reverse-transcription polymerase chain reaction (RT-PCR), and quantitative-PCR (qRT-PCR) were utilized in an ovarian cancer (including Fallopian tube and primary peritoneal cancers) patient cohort; humoral responses against NY-ESO-1 were determined by ELISA. Clinicopathologic outcomes including progression-free (PFS) and overall (OS) survival were evaluated based on NY-ESO-1 expression. Cohen's kappa (κ) tested agreement between expression tests. RESULTS NY-ESO-1 expression was detected by any method in 40.7% of 1002 patients' tumors (NY-ESO-1+) and baseline humoral response was identified in 19.0% of 689 tested patients. NY-ESO-1+ patients were older (p<0.001), higher stage (85% stage III/IV vs. 76.4%, p=0.015), less likely to have a complete response to initial therapy (53.9% vs. 68.9%, p=0.002), had more serous histotype (74.5% vs. 66.9%, p=0.011), and had more grade 3 tumors (83.7% vs. 70.8%, p<0.001). There was a trend towards shorter PFS (22.2 vs. 25.0months, p=0.07) and significantly shorter OS (42.9 vs. 50.0months, p=0.003) among NY-ESO-1+ patients. A subset analysis of NY-ESO-1+ patients that received immunotherapy demonstrated improved OS by >2years (52.6 vs. 27.2months, p<0.001). CONCLUSIONS This study is the first demonstration of an association between NY-ESO-1 expression and an aggressive cancer phenotype. The relatively high expression frequency of NY-ESO-1 in ovarian cancer patients coupled with the poor clinical outcomes in NY-ESO-1+ patients reveals an underappreciated need for targeted therapy against this antigen. In support, our study reveals that NY-ESO-1+ patients enrolled on immunotherapy trials targeting the antigen exhibited an improvement in OS.
Collapse
Affiliation(s)
- J Brian Szender
- Department of Gynecologic Oncology, Roswell Park Cancer Institute, Buffalo, NY 14263, United States
| | | | - Kevin H Eng
- Biostatistics and Bioinformatics, Roswell Park Cancer Institute, Buffalo, NY 14263, United States
| | - Anthony J Miliotto
- Center for Immunotherapy, Roswell Park Cancer Institute, Buffalo, NY 14263, United States
| | - Amit A Lugade
- Center for Immunotherapy, Roswell Park Cancer Institute, Buffalo, NY 14263, United States
| | - Sacha Gnjatic
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States
| | - Junko Matsuzaki
- Center for Immunotherapy, Roswell Park Cancer Institute, Buffalo, NY 14263, United States
| | | | - Kunle Odunsi
- Department of Gynecologic Oncology, Roswell Park Cancer Institute, Buffalo, NY 14263, United States; Center for Immunotherapy, Roswell Park Cancer Institute, Buffalo, NY 14263, United States.
| |
Collapse
|
23
|
Giavina-Bianchi MH, Giavina-Bianchi Junior PF, Festa Neto C. Melanoma: tumor microenvironment and new treatments. An Bras Dermatol 2017; 92:156-166. [PMID: 28538872 PMCID: PMC5429098 DOI: 10.1590/abd1806-4841.20176183] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Accepted: 08/28/2016] [Indexed: 01/22/2023] Open
Abstract
In the recent past years, many discoveries in the tumor microenvironment have led to changes in the management of melanoma and it is rising up hopes, specially, to those in advanced stages. FDA approved seven new drugs from 2011 to 2014. They are: Vemurafenib, Dabrafenib and Trametinib, kinases inhibitors used for patients that have BRAFV600E mutation; Ipilimumab (anti-CTLA4), Pembrolizumab (anti-PD-1) and Nivolumab (anti-PD-1), monoclonal antibodies that stimulate the immune system; and Peginterferon alfa-2b, an anti-proliferative cytokine used as adjuvant therapy. In this article, we will review the molecular bases for these new metastatic melanoma therapeutic agents cited above and also analyze new molecular discoveries in melanoma study, as Cancer-Testis antigens (CT). They are capable of induce humoral and cellular immune responses in cancer patients and because of this immunogenicity and their restrict expression in normal tissues, they are considered an ideal candidate for vaccine development against cancer. Among CT antigens, NY-ESO-1 is the best characterized in terms of expression patterns and immunogenicity. It is expressed in 20-40% of all melanomas, more in metastatic lesions than in primary ones, and it is very heterogeneous inter and intratumoral. Breslow index is associate with NY-ESO-1 expression in primary cutaneous melanomas, but its relation to patient survival remains controversial.
Collapse
Affiliation(s)
| | | | - Cyro Festa Neto
- Dermatology Department of Universidade de São Paulo Medical
School (FMUSP) – São Paulo (SP), Brazil
| |
Collapse
|
24
|
Goode EF, Smyth EC. Immunotherapy for Gastroesophageal Cancer. J Clin Med 2016; 5:jcm5100084. [PMID: 27669318 PMCID: PMC5086586 DOI: 10.3390/jcm5100084] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Revised: 09/07/2016] [Accepted: 09/14/2016] [Indexed: 12/29/2022] Open
Abstract
Survival for patients with advanced oesophageal and stomach cancer is poor; together these cancers are responsible for more than a million deaths per year globally. As chemotherapy and targeted therapies such as trastuzumab and ramucirumab result in modest improvements in survival but not long-term cure for such patients, development of alternative treatment approaches is warranted. Novel immunotherapy drugs such as checkpoint inhibitors have been paradigm changing in melanoma, non-small cell lung cancer and urothelial cancers. In this review, we assess the early evidence for efficacy of immunotherapy in patients with gastroesophageal cancer in addition to considering biomarkers associated with response to these treatments. Early results of Anti- Programmed Cell Death Protein-1 (anti-PD-1), anti-PD-L1 and anti-Cytotoxic T-lymphocyte assosciated protein-4 (anti-CTLA4) trials are examined, and we conclude with a discussion on the future direction for immunotherapy for gastroesophageal cancer patients.
Collapse
Affiliation(s)
- Emily F Goode
- The Royal Marsden Hospital, NHS Foundation Trust, London SW3 6JJ, UK.
| | - Elizabeth C Smyth
- The Royal Marsden Hospital, NHS Foundation Trust, London SW3 6JJ, UK.
| |
Collapse
|
25
|
Abstract
Melanoma-associated antigen-A (MAGE-A) and New York esophageal squamous cell cancer-1 (NY-ESO-1) are 2 cancer testis antigens (CTA) demonstrating potential for use in targeted immunotherapy. Clinical trials in melanoma and synovial sarcomas targeting these antigens in immune-based therapies have demonstrated durable tumor regression. Although protein expression of NY-ESO-1 has been assessed in a variety of cancer types, the expression of MAGE-A has not been studied in depth. In this study we analyzed MAGE-A and NY-ESO-1 expression in 314 melanoma specimens from 301 melanoma patients, 38 patients with squamous cell cancers and 111 patients with adenocarcinomas. Our results demonstrated higher expression of MAGE-A compared with NY-ESO-1 in melanomas (32% vs. 13%) and squamous cell carcinomas (45% vs. 7.9%), and higher expression of both CTAs in metastatic versus primary tumors. CTA expression in adenocarcinomas was low (MAGE-A: 10%, NY-ESO-1: 0.9%). In addition, we looked at concordance of expression among metastatic melanoma lesions within the same patient and found concordant expression in 38 of 47 patients for MAGE-A and 43 of 47 patients for NY-ESO-1. Our study demonstrated that the MAGE-A family may be of greater utility than NY-ESO-1 for targeted immunotherapy in a variety of cancer histologies, in particular metastatic melanomas and squamous cell carcinomas.
Collapse
|
26
|
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: 12.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
27
|
Expression of Cancer Testis Antigens in Colorectal Cancer: New Prognostic and Therapeutic Implications. DISEASE MARKERS 2016; 2016:1987505. [PMID: 27635108 PMCID: PMC5007337 DOI: 10.1155/2016/1987505] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Accepted: 07/28/2016] [Indexed: 12/11/2022]
Abstract
Background. While cancer/testis antigens (CTAs) are restricted in postnatal tissues to testes and germ line-derived cells, their role in cancer development and the clinical significance of their expression still remain to be better defined. Objective. The aim of this study was to investigate the level of CTA expression in colon samples from patients with colorectal cancer (CRC) in relation to patient clinical status. Methods. Forty-five patients with newly diagnosed colorectal cancer were included in the study. We selected a panel of 18 CTAs that were previously detected in CRC as well as some new gene candidates, and their expression was detected at the mRNA level by employing RQ-PCR. Additionally, we evaluated CTA expression in three colon cancer cell lines (CL-188, HTB-39, and HTB-37) after exposure to the DNA methylation-modifying drug 5-azacytidine. Results. We report that 6 out of 18 (33%) CTAs tested (MAGEA3, OIP5, TTK, PLU1, DKKL1, and FBXO39) were significantly (p < 0.05) overexpressed in tumor tissue compared with healthy colon samples isolated from the same patients. Conclusions. Moreover, we found that MAGEA3, PLU-1, and DKKL expression positively correlated with disease progression, evaluated according to the Dukes staging system. Finally, 5-azacytidine exposure significantly upregulated expression of CTAs on CRC cells, which indicates that this demethylation agent could be employed therapeutically to enhance the immune response against tumor cells.
Collapse
|
28
|
Rastgoosalami M, Memar B, Aledavood SA, Fanipakdel A. Evaluation of MAGE-1 Cancer-Testis Antigen Expression in Invasive Breast Cancer and its Correlation with Prognostic Factors. IRANIAN JOURNAL OF CANCER PREVENTION 2016; 9:e4404. [PMID: 27761208 PMCID: PMC5056020 DOI: 10.17795/ijcp-4404] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Revised: 11/14/2015] [Accepted: 08/10/2016] [Indexed: 12/14/2022]
Abstract
Background Aberrant expression of cancer-testis antigens (CTA) in breast carcinoma tissue, and its natural expression in the testis, the tissue away from the immune system, makes them good candidates for cancer immunotherapy and vaccines designing. Objectives The aim of this study was to assess the expression of a CTA (MAGE-1) in invasive breast cancer and its correlation with prognostic factors. Methods Paraffin blocks of breast cancer tissues from 113 patients operated in 2011 - 2013 were stained for MAGE-1expression by immunohistochemistry (IHC). The associations of MAGE-1 expression with known prognostic factors were assessed by statistical analysis using SPSS 16. Results MAGE-1 expression was found in cancer cell cytoplasms of 30.1% of patients, with different degrees of intensity, (23.9% moderate and 6.2% strong). Nuclear staining turned positive in 31.8%, stratified from moderate in 26.5%to to strong in 5.3%. There was a significant association between the number of lymph nodes involved and both nuclear (P = 0.042) and cytoplasmic (P = 0.003) MAGE-1 expression. There was also a significant correlation between the nuclear expression of MAGE-1 and tumor size (P = 0.018). Cytoplasmic expression of MAGE-1 increased with increasing pathologic grade of tumors although the association was not statistically significant (P = 0.119). Conclusions CTA MAGE-1 has significant association with some prognostic factors in breast cancer and may have the role of a prognostic factor.
Collapse
Affiliation(s)
| | - Bahram Memar
- Cancer Research Center, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, IR Iran
| | - Seyed Amir Aledavood
- Cancer Research Center, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, IR Iran
| | - Azar Fanipakdel
- Cancer Research Center, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, IR Iran
| |
Collapse
|
29
|
Kajihara M, Takakura K, Kanai T, Ito Z, Saito K, Takami S, Shimodaira S, Okamoto M, Ohkusa T, Koido S. Dendritic cell-based cancer immunotherapy for colorectal cancer. World J Gastroenterol 2016; 22:4275-86. [PMID: 27158196 PMCID: PMC4853685 DOI: 10.3748/wjg.v22.i17.4275] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Revised: 03/15/2016] [Accepted: 04/07/2016] [Indexed: 02/06/2023] Open
Abstract
Colorectal cancer (CRC) is one of the most common cancers and a leading cause of cancer-related mortality worldwide. Although systemic therapy is the standard care for patients with recurrent or metastatic CRC, the prognosis is extremely poor. The optimal sequence of therapy remains unknown. Therefore, alternative strategies, such as immunotherapy, are needed for patients with advanced CRC. This review summarizes evidence from dendritic cell-based cancer immunotherapy strategies that are currently in clinical trials. In addition, we discuss the possibility of antitumor immune responses through immunoinhibitory PD-1/PD-L1 pathway blockade in CRC patients.
Collapse
|
30
|
Hagiwara Y, Sieverling L, Hanif F, Anton J, Dickinson ER, Bui TTT, Andreeva A, Barran PE, Cota E, Nikolova PV. Consequences of point mutations in melanoma-associated antigen 4 (MAGE-A4) protein: Insights from structural and biophysical studies. Sci Rep 2016; 6:25182. [PMID: 27121989 PMCID: PMC4848555 DOI: 10.1038/srep25182] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 04/12/2016] [Indexed: 02/01/2023] Open
Abstract
The Melanoma-Associated Antigen A4 (MAGE-A4) protein is a target for cancer therapy. The function of this protein is not well understood. We report the first comprehensive study on key cancer-associated MAGE-A4 mutations and provide analysis on the consequences of these mutations on the structure, folding and stability of the protein. Based on Nuclear Magnetic Resonance and Circular Dichroism, these mutations had no significant effects on the structure and the folding of the protein. Some mutations affected the thermal stability of the protein remarkably. Native mass spectrometry of wild-type MAGE-A4 showed a broad charge state distribution suggestive of a structurally dynamic protein. Significant intensity was found in relatively low charge states, indicative of a predominantly globular form and some population in more extended states. The latter is supported by Ion Mobility measurements. The MAGE-A4 mutants exhibited similar features. These novel molecular insights shed further light on better understanding of these proteins, which are implicated in a wide range of human cancers.
Collapse
Affiliation(s)
- Yoshio Hagiwara
- King's College London, Faculty of Life Sciences &Medicine, Institute of Pharmaceutical Science, Franklin-Wilkins Building, 150 Stamford St, London, SE1 9NH, UK
| | - Lina Sieverling
- King's College London, Faculty of Life Sciences &Medicine, Institute of Pharmaceutical Science, Franklin-Wilkins Building, 150 Stamford St, London, SE1 9NH, UK
| | - Farina Hanif
- King's College London, Faculty of Life Sciences &Medicine, Institute of Pharmaceutical Science, Franklin-Wilkins Building, 150 Stamford St, London, SE1 9NH, UK
| | - Jensy Anton
- King's College London, Faculty of Life Sciences &Medicine, Institute of Pharmaceutical Science, Franklin-Wilkins Building, 150 Stamford St, London, SE1 9NH, UK
| | - Eleanor R Dickinson
- Michael Barber Centre for Collaborative Mass Spectrometry, Manchester Institute of Biotechnology, The University of Manchester, 131 Princess Street, Manchester, M1 7DN, UK
| | - Tam T T Bui
- Biomolecular Spectroscopy Centre, King's College London, The Wolfson Wing, Hodgkin Building, London SE1 1UL
| | | | - Perdita E Barran
- Michael Barber Centre for Collaborative Mass Spectrometry, Manchester Institute of Biotechnology, The University of Manchester, 131 Princess Street, Manchester, M1 7DN, UK
| | - Ernesto Cota
- Imperial College London, Faculty of Natural Sciences, London, SW7 2AZ
| | - Penka V Nikolova
- King's College London, Faculty of Life Sciences &Medicine, Institute of Pharmaceutical Science, Franklin-Wilkins Building, 150 Stamford St, London, SE1 9NH, UK
| |
Collapse
|
31
|
Oshima Y, Shimada H, Yajima S, Nanami T, Matsushita K, Nomura F, Kainuma O, Takiguchi N, Soda H, Ueda T, Iizasa T, Yamamoto N, Yamamoto H, Nagata M, Yokoi S, Tagawa M, Ohtsuka S, Kuwajima A, Murakami A, Kaneko H. NY-ESO-1 autoantibody as a tumor-specific biomarker for esophageal cancer: screening in 1969 patients with various cancers. J Gastroenterol 2016; 51:30-4. [PMID: 25906289 DOI: 10.1007/s00535-015-1078-8] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Accepted: 04/06/2015] [Indexed: 02/04/2023]
Abstract
BACKGROUND Although serum NY-ESO-1 antibodies (s-NY-ESO-1-Abs) have been reported in patients with esophageal carcinoma, this assay system has not been used to study a large series of patients with various other cancers. PATIENTS AND METHODS Serum samples of 1969 cancer patients [esophageal cancer (n = 172), lung cancer (n = 269), hepatocellular carcinoma (n = 91), prostate cancer (n = 358), gastric cancer (n = 313), colorectal cancer (n = 262), breast cancer (n = 365)] and 74 healthy individuals were analyzed using an originally developed enzyme-linked immunosorbent assay system for s-NY-ESO-1-Abs. The optical density cut-off value, determined as the mean plus three standard deviations for serum samples from the healthy controls, was fixed at 0.165. Conventional tumor markers were also evaluated in patients with esophageal carcinoma. RESULTS The positive rate of s-NY-ESO-1-Abs in patients with esophageal cancer (31 %) was significantly higher than that in the other groups: patients with lung cancer (13 %), patients with hepatocellular carcinoma (11 %), patients with prostate cancer (10 %), patients with gastric cancer (10 %), patients with colorectal cancer (8 %), patients with breast cancer (7 %), and healthy controls (0 %). The positive rate of s-NY-ESO-1-Abs was comparable to that of serum p53 antibodies (33 %), squamous cell carcinoma antigen (36 %), carcinoembryonic antigen (26 %), and CYFRA 21-1 (18 %) and gradually increased with the tumor stage. CONCLUSIONS The positive rate of s-NY-ESO-1-Abs was significantly higher in patients with esophageal cancer than in patients with the other types of cancers. On the basis of its high specificity and sensitivity, even in patients with stage I tumors, s-NY-ESO-1-Abs may be one of the first choices for esophageal cancer.
Collapse
Affiliation(s)
- Yoko Oshima
- Department of Surgery, School of Medicine, Toho University, Tokyo, Japan
| | - Hideaki Shimada
- Department of Surgery, School of Medicine, Toho University, Tokyo, Japan.
| | - Satoshi Yajima
- Department of Surgery, School of Medicine, Toho University, Tokyo, Japan
| | - Tatsuki Nanami
- Department of Surgery, School of Medicine, Toho University, Tokyo, Japan
| | - Kazuyuki Matsushita
- Molecular Diagnosis, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Fumio Nomura
- Molecular Diagnosis, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Osamu Kainuma
- Division of Gastroenterological Surgery, Chiba Cancer Center, Chiba, Japan
| | - Nobuhiro Takiguchi
- Division of Gastroenterological Surgery, Chiba Cancer Center, Chiba, Japan
| | - Hiroaki Soda
- Division of Gastroenterological Surgery, Chiba Cancer Center, Chiba, Japan
| | - Takeshi Ueda
- Division of Urology, Chiba Cancer Center, Chiba, Japan
| | | | - Naoto Yamamoto
- Division of Breast Surgery, Chiba Cancer Center, Chiba, Japan
| | - Hiroshi Yamamoto
- Division of Gastroenterological Surgery, Chiba Cancer Center, Chiba, Japan
| | - Matsuo Nagata
- Division of Gastroenterological Surgery, Chiba Cancer Center, Chiba, Japan
| | - Sana Yokoi
- Division of Clinical Genomics, Chiba Cancer Center, Chiba, Japan
| | - Masatoshi Tagawa
- Division of Pathology and Cell Therapy, Chiba Cancer Center, Chiba, Japan
| | - Seiko Ohtsuka
- Department of Surgery, School of Medicine, Toho University, Tokyo, Japan
| | - Akiko Kuwajima
- Medical & Biological Laboratories Co., Ltd, Nagoya, Japan
| | | | - Hironori Kaneko
- Department of Surgery, School of Medicine, Toho University, Tokyo, Japan
| |
Collapse
|
32
|
Lee HJ, Kim JY, Song IH, Park IA, Yu JH, Gong G. Expression of NY-ESO-1 in Triple-Negative Breast Cancer Is Associated with Tumor-Infiltrating Lymphocytes and a Good Prognosis. Oncology 2015; 89:337-44. [DOI: 10.1159/000439535] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Accepted: 08/13/2015] [Indexed: 11/19/2022]
|
33
|
Ghafouri-Fard S, Shamsi R, Seifi-Alan M, Javaheri M, Tabarestani S. Cancer-testis genes as candidates for immunotherapy in breast cancer. Immunotherapy 2014; 6:165-79. [PMID: 24491090 DOI: 10.2217/imt.13.165] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Cancer-testis (CT) antigens are tumor-associated antigens attracting immunologists for their possible application in the immunotherapy of cancer. Several clinical trials have assessed their therapeutic potentials in cancer patients. Breast cancers, especially triple-negative cancers are among those with significant expression of CT genes. Identification of CT genes with high expression in cancer patients is the prerequisite for any immunotherapeutic approach. CT genes have gained attention not only for immunotherapy of cancer patients, but also for immunoprevention in high-risk individuals. Many CT genes have proved to be immunogenic in breast cancer patients suggesting the basis for the development of polyvalent vaccines.
Collapse
Affiliation(s)
- Soudeh Ghafouri-Fard
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran 19857-17443, Iran
| | | | | | | | | |
Collapse
|
34
|
Wang L, Jin N, Schmitt A, Greiner J, Malcherek G, Hundemer M, Mani J, Hose D, Raab MS, Ho AD, Chen BA, Goldschmidt H, Schmitt M. T cell-based targeted immunotherapies for patients with multiple myeloma. Int J Cancer 2014; 136:1751-68. [PMID: 25195787 DOI: 10.1002/ijc.29190] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Revised: 08/28/2014] [Accepted: 09/03/2014] [Indexed: 12/17/2022]
Abstract
Despite high-dose chemotherapy followed by autologs stem-cell transplantation as well as novel therapeutic agents, multiple myeloma (MM) remains incurable. Following the general trend towards personalized therapy, targeted immunotherapy as a new approach in the therapy of MM has emerged. Better progression-free survival and overall survival after tandem autologs/allogeneic stem cell transplantation suggest a graft versus myeloma effect strongly supporting the usefulness of immunological therapies for MM patients. How to induce a powerful antimyeloma effect is the key issue in this field. Pivotal is the definition of appropriate tumor antigen targets and effective methods for expansion of T cells with clinical activity. Besides a comprehensive list of tumor antigens for T cell-based approaches, eight promising antigens, CS1, Dickkopf-1, HM1.24, Human telomerase reverse transcriptase, MAGE-A3, New York Esophageal-1, Receptor of hyaluronic acid mediated motility and Wilms' tumor gene 1, are described in detail to provide a background for potential clinical use. Results from both closed and on-going clinical trials are summarized in this review. On the basis of the preclinical and clinical data, we elaborate on three encouraging therapeutic options, vaccine-enhanced donor lymphocyte infusion, chimeric antigen receptors-transfected T cells as well as vaccines with multiple antigen peptides, to pave the way towards clinically significant immune responses against MM.
Collapse
Affiliation(s)
- Lei Wang
- Department of Internal Medicine V, University Clinic Heidelberg, University of Heidelberg, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Yilmaz-Ozcan S, Sade A, Kucukkaraduman B, Kaygusuz Y, Senses KM, Banerjee S, Gure AO. Epigenetic mechanisms underlying the dynamic expression of cancer-testis genes, PAGE2, -2B and SPANX-B, during mesenchymal-to-epithelial transition. PLoS One 2014; 9:e107905. [PMID: 25229454 PMCID: PMC4168264 DOI: 10.1371/journal.pone.0107905] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2014] [Accepted: 08/22/2014] [Indexed: 12/14/2022] Open
Abstract
Cancer-testis (CT) genes are expressed in various cancers but not in normal tissues other than in cells of the germline. Although DNA demethylation of promoter-proximal CpGs of CT genes is linked to their expression in cancer, the mechanisms leading to demethylation are unknown. To elucidate such mechanisms we chose to study the Caco-2 colorectal cancer cell line during the course of its spontaneous differentiation in vitro, as we found CT genes, in particular PAGE2, -2B and SPANX-B, to be up-regulated during this process. Differentiation of these cells resulted in a mesenchymal-to-epithelial transition (MET) as evidenced by the gain of epithelial markers CDX2, Claudin-4 and E-cadherin, and a concomitant loss of mesenchymal markers Vimentin, Fibronectin-1 and Transgelin. PAGE2 and SPAN-X up-regulation was accompanied by an increase in Ten-eleven translocation-2 (TET2) expression and cytosine 5-hydroxymethylation as well as the disassociation of heterochromatin protein 1 and the polycomb repressive complex 2 protein EZH2 from promoter-proximal regions of these genes. Reversal of differentiation resulted in down-regulation of PAGE2, -2B and SPANX-B, and induction of epithelial-to-mesenchymal transition (EMT) markers, demonstrating the dynamic nature of CT gene regulation in this model.
Collapse
Affiliation(s)
- Sinem Yilmaz-Ozcan
- Department of Molecular Biology and Genetics, Bilkent University, Ankara, Turkey
| | - Asli Sade
- Department of Biological Sciences, Middle East Technical University, Ankara, Turkey
| | - Baris Kucukkaraduman
- Department of Molecular Biology and Genetics, Bilkent University, Ankara, Turkey
| | - Yasemin Kaygusuz
- Department of Molecular Biology and Genetics, Bilkent University, Ankara, Turkey
| | - Kerem Mert Senses
- Department of Molecular Biology and Genetics, Bilkent University, Ankara, Turkey
| | - Sreeparna Banerjee
- Department of Biological Sciences, Middle East Technical University, Ankara, Turkey
| | - Ali Osmay Gure
- Department of Molecular Biology and Genetics, Bilkent University, Ankara, Turkey
- * E-mail:
| |
Collapse
|
36
|
Islamian JP, Mohammadi M, Baradaran B. Inhibition of human esophageal squamous cell carcinomas by targeted silencing of tumor enhancer genes: an overview. Cancer Biol Med 2014; 11:78-85. [PMID: 25009749 PMCID: PMC4069799 DOI: 10.7497/j.issn.2095-3941.2014.02.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2013] [Accepted: 04/08/2014] [Indexed: 12/12/2022] Open
Abstract
Esophageal cancer has been reported as the ninth most common malignancy and ranks as the sixth most frequent cause of death worldwide. Esophageal cancer treatment involves surgery, chemotherapy, radiation therapy, or combination therapy. Novel strategies are needed to boost the oncologic outcome. Recent advances in the molecular biology of esophageal cancer have documented the role of genetic alterations in tumorigenesis. Oncogenes serve a pivotal function in tumorigenesis. Targeted therapies are directed at the unique molecular signature of cancer cells for enhanced efficacy with low toxicity. RNA interference (RNAi) technology is a powerful tool for silencing endogenous or exogenous genes in mammalian cells. Related results have shown that targeting oncogenes with siRNAs, specifically the mRNA, effectively reduces tumor cell proliferation and induces apoptotic cell death. This article will briefly review studies on silencing tumor enhancer genes related to the induction of esophageal cancer.
Collapse
Affiliation(s)
- Jalil Pirayesh Islamian
- 1 Tabriz University of Medical Sciences, School of Medicine, Tabriz, East Asarbeidjan, Iran ; 2 Department of Radiation, School of Paramedicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran ; 3 Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohsen Mohammadi
- 1 Tabriz University of Medical Sciences, School of Medicine, Tabriz, East Asarbeidjan, Iran ; 2 Department of Radiation, School of Paramedicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran ; 3 Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- 1 Tabriz University of Medical Sciences, School of Medicine, Tabriz, East Asarbeidjan, Iran ; 2 Department of Radiation, School of Paramedicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran ; 3 Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
37
|
Barøy T, Kresse SH, Skårn M, Stabell M, Castro R, Lauvrak S, Llombart-Bosch A, Myklebost O, Meza-Zepeda LA. Reexpression of LSAMP inhibits tumor growth in a preclinical osteosarcoma model. Mol Cancer 2014; 13:93. [PMID: 24885297 PMCID: PMC4029956 DOI: 10.1186/1476-4598-13-93] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2013] [Accepted: 03/11/2014] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Osteosarcomas are the most common primary malignant tumors of bone, showing complex chromosomal rearrangements with multiple gains and losses. A frequent deletion within the chromosomal region 3q13.31 has been identified by us and others, and is mainly reported to be present in osteosarcomas. The purpose of the study was to further characterize the frequency and the extent of the deletion in an extended panel of osteosarcoma samples, and the expression level of the affected genes within the region. We have identified LSAMP as the target gene for the deletion, and have studied the functional implications of LSAMP-reexpression. METHODS LSAMP copy number, expression level and protein level were investigated by quantitative PCR and western blotting in an osteosarcoma panel. The expression of LSAMP was restored in an osteosarcoma cell line, and differences in proliferation rate, tumor formation, gene expression, migration rate, differentiation capabilities, cell cycle distribution and apoptosis were investigated by metabolic dyes, tumor formation in vivo, gene expression profiling, time-lapse photography, differentiation techniques and flow cytometry, respectively. RESULTS We found reduced copy number of LSAMP in 45/76 osteosarcoma samples, reduced expression level in 25/42 samples and protein expression in 9/42 samples. By restoring the expression of LSAMP in a cell line with a homozygous deletion of the gene, the proliferation rate in vitro was significantly reduced and tumor growth in vivo was significantly delayed. In response to reexpression of LSAMP, mRNA expression profiling revealed consistent upregulation of the genes hairy and enhancer of split 1 (HES1), cancer/testis antigen 2 (CTAG2) and kruppel-like factor 10 (KLF10). CONCLUSIONS The high frequency and the specificity of the deletion indicate that it is important for the development of osteosarcomas. The deletion targets the tumor suppressor LSAMP, and based on the functional evidence, the tumor suppressor function of LSAMP is most likely exerted by reducing the proliferation rate of the tumor cells, possibly by indirectly upregulating one or more of the genes HES1, CTAG2 or KLF10. To our knowledge, this study describes novel functions of LSAMP, a first step to understanding the functional role of this specific deletion in osteosarcomas.
Collapse
MESH Headings
- Antigens, Neoplasm/genetics
- Antigens, Neoplasm/metabolism
- Antigens, Surface/genetics
- Antigens, Surface/metabolism
- Basic Helix-Loop-Helix Transcription Factors/genetics
- Basic Helix-Loop-Helix Transcription Factors/metabolism
- Bone Neoplasms/genetics
- Bone Neoplasms/metabolism
- Bone Neoplasms/mortality
- Bone Neoplasms/pathology
- Cell Adhesion Molecules, Neuronal/genetics
- Cell Adhesion Molecules, Neuronal/metabolism
- Cell Line, Tumor
- Cell Proliferation/genetics
- Chromosome Mapping
- Chromosomes, Human, Pair 3
- Early Growth Response Transcription Factors/genetics
- Early Growth Response Transcription Factors/metabolism
- Female
- GPI-Linked Proteins/genetics
- GPI-Linked Proteins/metabolism
- Gene Deletion
- Gene Dosage
- Gene Expression Regulation, Neoplastic
- Genetic Complementation Test
- Homeodomain Proteins/genetics
- Homeodomain Proteins/metabolism
- Homozygote
- Humans
- Kruppel-Like Transcription Factors/genetics
- Kruppel-Like Transcription Factors/metabolism
- Male
- Mutation Rate
- Osteosarcoma/genetics
- Osteosarcoma/metabolism
- Osteosarcoma/mortality
- Osteosarcoma/pathology
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Signal Transduction
- Survival Analysis
- Transcription Factor HES-1
Collapse
Affiliation(s)
- Tale Barøy
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, Norway
| | - Stine H Kresse
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, Norway
| | - Magne Skårn
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, Norway
| | - Marianne Stabell
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, Norway
| | - Russell Castro
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, Norway
| | - Silje Lauvrak
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, Norway
| | | | - Ola Myklebost
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, Norway
- Department of Molecular Biosciences, University of Oslo, Oslo, Norway
| | - Leonardo A Meza-Zepeda
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, Norway
- Genomics Core Facility, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
38
|
Skorodumova LO, Lukashina MI, Sal'nikova LE, Tikhonova OA, Ivanov SY, Larin SS. Study of expression of testicular cancer genes in breast cancer samples. DOKL BIOCHEM BIOPHYS 2014; 453:316-8. [PMID: 24385105 DOI: 10.1134/s1607672913060112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2013] [Indexed: 11/23/2022]
Affiliation(s)
- L O Skorodumova
- Institute of Gene Biology, Russian Academy of Sciences, ul. Vavilova 34/5, Moscow, 119334, Russia
| | | | | | | | | | | |
Collapse
|
39
|
Hou SY, Sang MX, Geng CZ, Liu WH, Lü WH, Xu YY, Shan BE. Expressions of MAGE-A9 and MAGE-A11 in breast cancer and their expression mechanism. Arch Med Res 2013; 45:44-51. [PMID: 24316396 DOI: 10.1016/j.arcmed.2013.10.005] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2013] [Accepted: 09/26/2013] [Indexed: 11/26/2022]
Abstract
BACKGROUND AND AIMS The MAGE gene encodes cancer/testis antigens that are selectively expressed in various types of human neoplasms but not in normal tissues other than testis and placenta. However, the expression pattern of MAGE-A9 and MAGE-A11 in breast cancer patients is still unclear. The purpose of our study is to investigate the expression pattern and mechanism of MAGE-A9 and MAGE-A11 in breast cancer patients. METHODS The expression of MAGE-A9 and MAGE-A11 was investigated in 60 breast benign diseases specimens, 60 tumor-free breast specimens and 60 breast cancer specimens by RT-PCR, and their correlation with clinicopathological parameters was elucidated. We examined the influence of the DNA methylase inhibitor 5-aza-2'-deoxycytidine (5-aza-CdR) together with the histone deacetylase inhibitor trichostatin A (TSA) on the expression of MAGE-A9 and MAGE-A11 genes in two breast cancer cell lines. RESULTS The expression rates of MAGE-A9 and MAGE-A11 in breast cancer specimens were 45 and 66.7%, respectively. MAGE-A9 and MAGE-A11 expression was positively associated with estrogen-receptor (ER) and HER-2 expression (p <0.05). 5-Aza-CdR treatment alone could induce the expression of MAGE-A9 and MAGE-A11 in cell lines that did not express this antigen. TSA treatment alone had no influence on MAGE-A9 and MAGE-A11 gene expression. However, TSA was able synergistically to enhance 5-aza-CdR-mediated MAGE-A transcription (p <0.05). CONCLUSIONS Our data show that MAGE-A9 and MAGE-A11 are tumor-specific antigens and not only DNA hypermethylation but also histone deacetylation is responsible for the mechanism underlying MAGE-A9 and MAGE-A11 gene silencing.
Collapse
Affiliation(s)
- Shu-Yun Hou
- Department of Oncology, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, PR China; Research Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, PR China
| | - Mei-Xiang Sang
- Research Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, PR China
| | - Cui-Zhi Geng
- Department of Breast Cancer Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, PR China
| | - Wei-Hua Liu
- Research Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, PR China; Center for Disease Control and Prevention of Shijiazhuang, Shijiazhuang, Hebei, PR China
| | - Wei-Hua Lü
- Research Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, PR China
| | - Ying-Ying Xu
- Research Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, PR China
| | - Bao-En Shan
- Research Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, PR China.
| |
Collapse
|
40
|
Komatsu N, Jackson HM, Chan KF, Oveissi S, Cebon J, Itoh K, Chen W. Fine-mapping naturally occurring NY-ESO-1 antibody epitopes in melanoma patients’ sera using short overlapping peptides and full-length recombinant protein. Mol Immunol 2013; 54:465-71. [DOI: 10.1016/j.molimm.2013.01.014] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2013] [Accepted: 01/30/2013] [Indexed: 11/25/2022]
|
41
|
NY-ESO-1 cancer testis antigen demonstrates high immunogenicity in triple negative breast cancer. PLoS One 2012; 7:e38783. [PMID: 22761704 PMCID: PMC3386262 DOI: 10.1371/journal.pone.0038783] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2012] [Accepted: 05/10/2012] [Indexed: 12/31/2022] Open
Abstract
PURPOSE NY-ESO-1 cancer testis (CT) antigen is an attractive candidate for immunotherapy as a result of its high immunogenicity. The aim of this study was to explore the potential for NY-ESO-1 antigen directed immunotherapy in triple negative breast cancer (TNBC) by determining the frequency of expression by immunohistochemistry (IHC) and the degree of inherent immunogenicity to NY-ESO-1. EXPERIMENTAL DESIGN 168 TNBC and 47 ER+/HER2- primary breast cancer specimens were used to determine NY-ESO-1 frequency by IHC. As previous studies have shown that patients with a robust innate humoral immune response to CT antigens are more likely to develop CD8 T-cell responses to NY-ESO-1 peptides, we evaluated the degree to which patients with NY-ESO-1 expression had inherent immunogenicity by measuring antibodies. The relationship between NY-ESO-1 expression and CD8+ T lymphocytes was also examined. RESULTS The frequency of NY-ESO-1 expression in the TNBC cohort was 16% versus 2% in ER+/HER2- patients. A higher NY-ESO-1 score was associated with a younger age at diagnosis in the TNBC patients with NY-ESO-1 expression (p = 0.026). No differences in OS (p = 0.278) or PFS (p = 0.238) by NY-ESO-1 expression status were detected. Antibody responses to NY-ESO-1 were found in 73% of TNBC patients whose tumors were NY-ESO-1 positive. NY-ESO-1 positive patients had higher CD8 counts than negative patients (p = 0.018). CONCLUSION NY-ESO-1 is expressed in a substantial subset of TNBC patients and leads to a high humoral immune response in a large proportion of these individuals. Given these observations, patients with TNBC may benefit from targeted therapies directed against NY-ESO-1.
Collapse
|
42
|
NY-ESO-1 expression in synovial sarcoma and other mesenchymal tumors: significance for NY-ESO-1-based targeted therapy and differential diagnosis. Mod Pathol 2012; 25:854-8. [PMID: 22388761 PMCID: PMC6309776 DOI: 10.1038/modpathol.2012.31] [Citation(s) in RCA: 77] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
A promising targeted therapy against NY-ESO-1 (CTAG 1B) using genetically modified T-cells in synovial sarcomas was recently demonstrated in a clinical trial at the NCI. To investigate the role of NY-ESO-1 immunohistochemistry in patient selection and gain better insight into the incidence of NY-ESO-1 expression in synovial sarcomas and other mesenchymal tumors, we evaluated NY-ESO-1 expression by immunohistochemistry in 417 tumors. This collection of samples included: 50 SS18/SSX1/2 fusion positive synovial sarcomas, 155 gastrointestinal stromal tumors (GIST), 135 other spindle cell sarcomas as well as 77 other sarcomas (chondrosarcoma, osteosarcoma, dedifferentiated liposarcoma, alveolar soft part sarcoma, rhabdomyosarcoma, angiosarcoma, malignant mesothelioma, and Ewing's sarcoma). We report that 76% of synovial sarcomas expressed NY-ESO-1 in a strong and diffuse pattern (2-3+, >50-70% of tumor cells). In contrast, only rare cases of other spindle cell mesenchymal tumor expressed NY-ESO-1 (GIST (2/155), malignant peripheral nerve sheath tumors (1/34), and dermatofibrosarcoma protuberans (2/20)). Individual cases of other sarcomas (angiosarcoma, malignant mesothelioma, chondrosarcoma, osteosarcoma, dedifferentiated liposarcoma, alveolar soft part sarcoma, and Ewing's sarcoma) were positive for NY-ESO-1. However, no positive cases were identified amongst our cohort of leiomyosarcomas (0/24), hemangiopericytoma/solitary fibrous tumors (0/40), and cellular schwannomas (0/17). In summary, we find that NY-ESO-1 is strongly and diffusely expressed in a majority of synovial sarcomas, but only rarely in other mesenchymal lesions. Beyond its role in patient selection for targeted therapy, immunohistochemistry for NY-ESO-1 may be diagnostically useful for the distinction of synovial sarcoma from other spindle cell neoplasms.
Collapse
|
43
|
Shao C, Sun W, Tan M, Glazer CA, Bhan S, Zhong X, Fakhry C, Sharma R, Westra WH, Hoque MO, Moskaluk CA, Sidransky D, Califano JA, Ha PK. Integrated, genome-wide screening for hypomethylated oncogenes in salivary gland adenoid cystic carcinoma. Clin Cancer Res 2011; 17:4320-30. [PMID: 21551254 DOI: 10.1158/1078-0432.ccr-10-2992] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
PURPOSE Salivary gland adenoid cystic carcinoma (ACC) is a rare malignancy that is poorly understood. To look for relevant oncogene candidates under the control of promoter methylation, an integrated, genome-wide screen was conducted. EXPERIMENTAL DESIGN Global demethylation of normal salivary gland cell strains using 5-aza-2'-deoxycytidine (5-aza-dC) and trichostatin A (TSA), followed by expression array analysis was conducted. ACC-specific expression profiling was generated using expression microarray analysis of primary ACC and normal samples. Next, the two profiles were integrated to identify a subset of genes for further validation of promoter demethylation in ACC versus normal. Finally, promising candidates were further validated for mRNA, protein, and promoter methylation levels in larger ACC cohorts. Functional validation was then conducted in cancer cell lines. RESULTS We found 159 genes that were significantly re-expressed after 5-aza-dC/TSA treatment and overexpressed in ACC. After initial validation, eight candidates showed hypomethylation in ACC: AQP1, CECR1, C1QR1, CTAG2, P53AIP1, TDRD12, BEX1, and DYNLT3. Aquaporin 1 (AQP1) showed the most significant hypomethylation and was further validated. AQP1 hypomethylation in ACC was confirmed with two independent cohorts. Of note, there was significant overexpression of AQP1 in both mRNA and protein in the paraffin-embedded ACC cohort. Furthermore, AQP1 was upregulated in 5-aza-dC/TSA-treated SACC83. Finally, AQP1 promoted cell proliferation and colony formation in SACC83. CONCLUSIONS Our integrated, genome-wide screening method proved to be an effective strategy for detecting novel oncogenes in ACC. AQP1 is a promising oncogene candidate for ACC and is transcriptionally regulated by promoter hypomethylation.
Collapse
Affiliation(s)
- Chunbo Shao
- Department of Otolaryngology-Head and Neck Surgery and Pathology, Johns Hopkins Medical Institutions, Baltimore, Maryland, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Bujas T, Marusic Z, Peric Balja M, Mijic A, Kruslin B, Tomas D. MAGE-A3/4 and NY-ESO-1 antigens expression in metastatic esophageal squamous cell carcinoma. Eur J Histochem 2011; 55:e7. [PMID: 21556122 PMCID: PMC3167346 DOI: 10.4081/ejh.2011.e7] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2011] [Revised: 02/09/2011] [Accepted: 02/15/2011] [Indexed: 11/23/2022] Open
Abstract
In the present study we analyzed immunohistochemical expression of MAGE-A 3/4 and NY-ESO-1 in 55 samples of esophageal squamous cell carcinomas (ESCC) and their respective lymph node metastases. To our knowledge this is the first study to assess and compare the expression of these antigens in ESCC lymph node metastases. Fifty (90.9%) primary ESCC were positive for MAGE-A 3/4 and 53 (96.6%) were positive for NY-ESO-1. MAGE-A 3/4 was expressed in all lymph node metastases and the intensity of expression was high in a majority of cases. NY-ESO-1 was negative in 2 (7.1%) lymph nodes metastases, while the reaction was predominantly moderate in the positive group. In primary tumors MAGE-A 3/4 showed a significantly higher intensity of expression compared to NY-ESO-1 (P=0.047), while in lymph node metastases the intensity of expression was not significantly different (P=0.387). Primary tumors with and without lymph node metastases showed no significant differences in MAGE-A 3/4 (P=0.672) and NY-ESO-1 (P=0.444) expression. Intensity of MAGE-A 3/4 (P=0.461) and NY-ESO-1 (P=0.414) expression in primary tumors was not significantly different compared to the expression in their respective lymph nodes metastases. Expression of MAGE-A 3/4 in primary tumors showed significant positive correlation with primary tumor expression of NY-ESO-1 (P=0.021) but no significant correlation with the expression of MAGE-A 3/4 in lymph node metastases (P=0.056). Expression of NY-ESO-1 in primary tumors showed significant positive correlation with the expression of NY-ESO-1 in lymph node metastases (P=0.001) and significant negative correlation with patients' age (P<0.001). Expression of MAGE-A 3/4 and NY-ESO-1 in primary tumors and lymph node metastases showed no significant correlation with prognostic parameters such as tumor grade and TNM stage (P>0.05). We have shown different levels of MAGE-A 3/4 and NY-ESO-1 expression in almost all specimens of primary tumor and lymph node metastases, suggesting that ESCC may be possible target of immunotherapy and anti-tumor vaccination. High levels of expression in lymph node metastases indicate possible clinical benefit of postoperative vaccine with MAGE-A3 and NY-ESO-1 in advanced stage of disease.
Collapse
Affiliation(s)
- T Bujas
- Department of Pathology, GeneralHospital Karlovac, University Hospital, Zagreb, Croatia.
| | | | | | | | | | | |
Collapse
|
45
|
Fratta E, Coral S, Covre A, Parisi G, Colizzi F, Danielli R, Nicolay HJM, Sigalotti L, Maio M. The biology of cancer testis antigens: putative function, regulation and therapeutic potential. Mol Oncol 2011; 5:164-82. [PMID: 21376678 DOI: 10.1016/j.molonc.2011.02.001] [Citation(s) in RCA: 260] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2010] [Revised: 01/31/2011] [Accepted: 02/03/2011] [Indexed: 12/14/2022] Open
Abstract
Cancer testis antigens (CTA) are a large family of tumor-associated antigens expressed in human tumors of different histological origin, but not in normal tissues except for testis and placenta. This tumor-restricted pattern of expression, together with their strong in vivo immunogenicity, identified CTA as ideal targets for tumor-specific immunotherapeutic approaches, and prompted the development of several clinical trials of CTA-based vaccine therapy. Driven by this practical clinical interest, a more detailed characterization of CTA biology has been recently undertaken. So far, at least 70 families of CTA, globally accounting for about 140 members, have been identified. Most of these CTA are expressed during spermatogenesis, but their function is still largely unknown. Epigenetic events, particularly DNA methylation, appear to be the primary mechanism regulating CTA expression in both normal and transformed cells, as well as in cancer stem cells. In view of the growing interest in CTA biology, the aim of this review is to provide the most recent information on their expression, regulation and function, together with a brief summary of the major clinical trials involving CTA as therapeutic agents. The pharmacologic modulation of CTA expression profiles on neoplastic cells by DNA hypomethylating drugs will also be discussed as a feasible approach to design new combination therapies potentially able to improve the clinical efficacy of currently adopted CTA-based immunotherapeutic regimens in cancer patients.
Collapse
Affiliation(s)
- Elisabetta Fratta
- Cancer Bioimmunotherapy Unit, Centro di Riferimento Oncologico, Istituto di Ricovero e Cura a Carattere Scientifico, Via Franco Gallini 2, 33081 Aviano, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
The SSX family of cancer-testis antigens as target proteins for tumor therapy. Clin Dev Immunol 2010; 2010:150591. [PMID: 20981248 PMCID: PMC2963798 DOI: 10.1155/2010/150591] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2010] [Accepted: 09/01/2010] [Indexed: 01/01/2023]
Abstract
Cancer-testis antigens (CTAs) represent an expanding class of tumor-associated proteins defined on the basis of their tissue-restricted expression to testis or ovary germline cells and frequent ectopic expression in tumor tissue. The expression of CTA in MHC class I-deficient germline cells makes these proteins particularly attractive as immunotherapeutic targets because they serve as essentially tumor-specific antigens for MHC class I-restricted CD8+ T cells. Moreover, because CTAs are expressed in many types of cancer, any therapeutic developed to target these antigens might have efficacy for multiple cancer types. Of particular interest among CTAs is the synovial sarcoma X chromosome breakpoint (SSX) family of proteins, which includes ten highly homologous family members. Expression of SSX proteins in tumor tissues has been associated with advanced stages of disease and worse patient prognosis. Additionally, both humoral and cell-mediated immune responses to SSX proteins have been demonstrated in patients with tumors of varying histological origin, which indicates that natural immune responses can be spontaneously generated to these antigens in cancer patients. The current review will describe the history and identification of this family of proteins, as well as what is known of their function, expression in normal and malignant tissues, and immunogenicity.
Collapse
|
47
|
Gnjatic S, Cao Y, Reichelt U, Yekebas EF, Nölker C, Marx AH, Erbersdobler A, Nishikawa H, Hildebrandt Y, Bartels K, Horn C, Stahl T, Gout I, Filonenko V, Ling KL, Cerundolo V, Luetkens T, Ritter G, Friedrichs K, Leuwer R, Hegewisch-Becker S, Izbicki JR, Bokemeyer C, Old LJ, Atanackovic D. NY-CO-58/KIF2C is overexpressed in a variety of solid tumors and induces frequent T cell responses in patients with colorectal cancer. Int J Cancer 2010; 127:381-93. [PMID: 19937794 DOI: 10.1002/ijc.25058] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
NY-CO-58/KIF2C has been identified as a tumor antigen by screening antibody responses in patients with colorectal cancer. However, expression had not consequently been examined, and nothing was known about its ability to induce spontaneous T cell responses, which have been suggested to play a role in the development of colorectal cancer. We analyzed 5 colorectal cancer cell lines, and tumor samples and adjacent healthy tissues from 176 patients with epithelial cancers for the expression of NY-CO-58/KIF2C by RT-PCR and Western Blot. T cell responses of 43 colorectal cancer patients and 35 healthy donors were evaluated by ELISpot following stimulation with 30mer peptides or full-length protein. All cell lines and tumor samples from colorectal cancer patients expressed NY-CO-58/KIF2C on the protein and RNA level, and expression levels correlated strongly with Ki-67 expression (r = 0.69; p = 0.0003). Investigating NY-CO-58/KIF2C-specific T cell responses, CD8(+) T cells directed against 1 or more peptides were found in less than 10% of patients, whereas specific CD4(+) T cells were detected in close to 50% of patients. These T cells were of high avidity, recognized the naturally processed antigen and secreted IFN-gamma and TNF-alpha. Depletion of CD4(+)CD25(+) T cells before stimulation significantly increased the intensity of the preexisting response. NY-CO-58/KIF2C is significantly overexpressed in colorectal and other epithelial cancers and expression levels correlate with the proliferative activity of the tumor. Importantly, NY-CO-58/KIF2C was able to induce spontaneous CD4(+) T cell responses of the Th1-type, which were tightly controlled by peripheral T regulatory cells.
Collapse
Affiliation(s)
- Sacha Gnjatic
- Ludwig Institute for Cancer Research, Memorial Sloan-Kettering Cancer Center, New York, NY, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
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
|
49
|
mRNA expression of the XAGE-1 gene in human acute leukemia. Int J Hematol 2010; 91:209-12. [PMID: 20178013 DOI: 10.1007/s12185-010-0527-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2009] [Revised: 01/23/2010] [Accepted: 02/05/2010] [Indexed: 10/19/2022]
Abstract
XAGE-1 is a cancer-testis antigen, which was demonstrated to be expressed at a significant frequency and to be immunogenic in some solid tumors. We analyzed the expression of 4 XAGE-1 transcript variant gene in human acute leukemias by reverse-transcription polymerase chain reaction. Among the 114 acute leukemias, 14/63 (22.22%) of the acute myeloid leukemia samples were positive for XAGE-1b genes. XAGE-1b mRNA expression was detected in 10/51 (19.61%) of the acute lymphocyte leukemia samples. However, we did not find any important correlation between XAGE-1b mRNA expression and clinical characteristics, such as sex, leukemia type, response to therapy and the percentage of blast in the first diagnosed bone marrow. We concluded that the XAGE-1b gene was expressed at the mRNA level in a proportion of human acute leukemia.
Collapse
|
50
|
Aoki M, Ueda S, Nishikawa H, Kitano S, Hirayama M, Ikeda H, Toyoda H, Tanaka K, Kanai M, Takabayashi A, Imai H, Shiraishi T, Sato E, Wada H, Nakayama E, Takei Y, Katayama N, Shiku H, Kageyama S. Antibody responses against NY-ESO-1 and HER2 antigens in patients vaccinated with combinations of cholesteryl pullulan (CHP)-NY-ESO-1 and CHP-HER2 with OK-432. Vaccine 2009; 27:6854-61. [PMID: 19761832 DOI: 10.1016/j.vaccine.2009.09.018] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2009] [Revised: 09/01/2009] [Accepted: 09/02/2009] [Indexed: 10/20/2022]
Abstract
Combination vaccines of the NY-ESO-1 protein complexed with cholesteryl pullulan (CHP), CHP-NY-ESO-1, and the truncated 146HER2 protein with CHP, CHP-HER2, were subcutaneously administered with the immuno-adjuvant OK-432 to eight esophageal cancer patients. Vaccination was well-tolerated. NY-ESO-1- and HER2-specific antibody responses were analyzed using the patients' sera and samples from previous single CHP-NY-ESO-1 or CHP-HER2 vaccine trial. The responses to NY-ESO-1 in the combination vaccine study were comparable to the single vaccine. For responses to HER2, there were fewer antibody responses in the combination vaccines. Although there were marked individual variations in the antibody responses to the NY-ESO-1 and HER2 antigens, the reaction patterns to these antigens were comparable within each patient. Antibodies to OK-432 were not augmented. Protein cancer vaccines targeting multiple antigens are feasible.
Collapse
Affiliation(s)
- Masatoshi Aoki
- Department of Haematology and Oncology, Mie University Graduate School of Medicine, 2-174, Edobashi, Tsu, Mie 514-8507, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|