1
|
Alsalloum A, Alrhmoun S, Perik-Zavosdkaia O, Fisher M, Volynets M, Lopatnikova J, Perik-Zavodskii R, Shevchenko J, Philippova J, Solovieva O, Zavjalov E, Kurilin V, Shiku H, Silkov A, Sennikov S. Decoding NY-ESO-1 TCR T cells: transcriptomic insights reveal dual mechanisms of tumor targeting in a melanoma murine xenograft model. Front Immunol 2024; 15:1507218. [PMID: 39660132 PMCID: PMC11628372 DOI: 10.3389/fimmu.2024.1507218] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 11/08/2024] [Indexed: 12/12/2024] Open
Abstract
The development of T cell receptor-engineered T cells (TCR-T) targeting intracellular antigens is a promising strategy for treating solid tumors; however, the mechanisms underlying their effectiveness remain poorly understood. In this study, we employed advanced techniques to investigate the functional state of T cells engineered with retroviral vectors to express a TCR specific for the NY-ESO-1 157-165 peptide in the HLA-A*02:01 context. Flow cytometry revealed a predominance of naïve T cells. Gene expression profiling using NanoString technology revealed upregulation of genes encoding chemokine receptors CCR2 and CCR5, indicating enhanced migration towards tumor sites. In the SK-Mel-37 xenograft model, these transduced T cells achieved complete tumor eradication. Furthermore, single-cell RNA sequencing (scRNA-seq) conducted 14 days post-TCR T cell infusion provided a comprehensive analysis of the in vivo adaptation of these cells, identifying a distinct subset of CD8+ effector T cells with an NK cell-like gene expression profile. Our findings indicate that NY-ESO-1 TCR-transduced T cells have the potential to mediate dual antitumor effects through both antigen-independent NK-like and antigen-specific CTL-like responses. This study underscores the potential of NY-ESO-1 TCR-T cells as potent tumor-eradicating agents, highlighting the importance of harnessing their versatile functional capabilities to refine and enhance therapeutic strategies.
Collapse
MESH Headings
- Animals
- Humans
- Mice
- Antigens, Neoplasm/immunology
- Antigens, Neoplasm/genetics
- Transcriptome
- Immunotherapy, Adoptive/methods
- Cell Line, Tumor
- Melanoma/therapy
- Melanoma/immunology
- Melanoma/genetics
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
- Xenograft Model Antitumor Assays
- HLA-A2 Antigen/genetics
- HLA-A2 Antigen/immunology
- CD8-Positive T-Lymphocytes/immunology
- Membrane Proteins/genetics
- Receptors, Chimeric Antigen/genetics
- Receptors, Chimeric Antigen/immunology
- Gene Expression Profiling
- Neoplasm Proteins
- Peptide Fragments
Collapse
Affiliation(s)
- Alaa Alsalloum
- Laboratory of molecular immunology, Federal State Budgetary Scientific Institution Research Institute of Fundamental and Clinical Immunology, Novosibirsk, Russia
- Department of Natural Sciences, Novosibirsk State University, Novosibirsk, Russia
| | - Saleh Alrhmoun
- Laboratory of molecular immunology, Federal State Budgetary Scientific Institution Research Institute of Fundamental and Clinical Immunology, Novosibirsk, Russia
- Department of Natural Sciences, Novosibirsk State University, Novosibirsk, Russia
| | - Olga Perik-Zavosdkaia
- Laboratory of molecular immunology, Federal State Budgetary Scientific Institution Research Institute of Fundamental and Clinical Immunology, Novosibirsk, Russia
| | - Marina Fisher
- Laboratory of molecular immunology, Federal State Budgetary Scientific Institution Research Institute of Fundamental and Clinical Immunology, Novosibirsk, Russia
| | - Marina Volynets
- Laboratory of molecular immunology, Federal State Budgetary Scientific Institution Research Institute of Fundamental and Clinical Immunology, Novosibirsk, Russia
| | - Julia Lopatnikova
- Laboratory of molecular immunology, Federal State Budgetary Scientific Institution Research Institute of Fundamental and Clinical Immunology, Novosibirsk, Russia
| | - Roman Perik-Zavodskii
- Laboratory of molecular immunology, Federal State Budgetary Scientific Institution Research Institute of Fundamental and Clinical Immunology, Novosibirsk, Russia
| | - Julia Shevchenko
- Laboratory of molecular immunology, Federal State Budgetary Scientific Institution Research Institute of Fundamental and Clinical Immunology, Novosibirsk, Russia
| | - Julia Philippova
- Laboratory of molecular immunology, Federal State Budgetary Scientific Institution Research Institute of Fundamental and Clinical Immunology, Novosibirsk, Russia
| | - Olga Solovieva
- Center for Collective Use SPF-vivarium ICG SB RAS, Ministry of Science and High Education of Russian Federation, Novosibirsk, Russia
| | - Evgenii Zavjalov
- Center for Collective Use SPF-vivarium ICG SB RAS, Ministry of Science and High Education of Russian Federation, Novosibirsk, Russia
| | - Vasily Kurilin
- Laboratory of molecular immunology, Federal State Budgetary Scientific Institution Research Institute of Fundamental and Clinical Immunology, Novosibirsk, Russia
| | - Hiroshi Shiku
- Department of Personalized Cancer Immunotherapy, Mie University Graduate School of Medicine, Tsu, Japan
| | - Alexander Silkov
- Laboratory of molecular immunology, Federal State Budgetary Scientific Institution Research Institute of Fundamental and Clinical Immunology, Novosibirsk, Russia
| | - Sergey Sennikov
- Laboratory of molecular immunology, Federal State Budgetary Scientific Institution Research Institute of Fundamental and Clinical Immunology, Novosibirsk, Russia
- Department of Immunology, V. Zelman Institute for Medicine and Psychology, Novosibirsk State University, Novosibirsk, Russia
| |
Collapse
|
2
|
Alsalloum A, Shevchenko JA, Sennikov S. NY-ESO-1 antigen: A promising frontier in cancer immunotherapy. Clin Transl Med 2024; 14:e70020. [PMID: 39275923 PMCID: PMC11399778 DOI: 10.1002/ctm2.70020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 08/22/2024] [Accepted: 09/01/2024] [Indexed: 09/16/2024] Open
Abstract
Significant strides have been made in identifying tumour-associated antigens over the past decade, revealing unique epitopes crucial for targeted cancer therapy. Among these, the New York esophageal squamous cell carcinoma (NY-ESO-1) protein, a cancer/testis antigen, stands out. This protein is presented on the cell surface by major histocompatibility complex class I molecules and exhibits restricted expression in germline cells and various cancers, marking it as an immune-privileged site. Remarkably, NY-ESO-1 serves a dual role as both a tumour-associated antigen and its own adjuvant, implying a potential function as a damage-associated molecular pattern. It elicits strong humoural immune responses, with specific antibody frequencies significantly correlating with disease progression. These characteristics make NY-ESO-1 an appealing candidate for developing effective and specific immunotherapy, particularly for advanced stages of disease. In this review, we provide a comprehensive overview of NY-ESO-1 as an immunogenic tumour antigen. We then explore the diverse strategies for targeting NY-ESO-1, including cancer vaccination with peptides, proteins, DNA, mRNA, bacterial vectors, viral vectors, dendritic cells and artificial adjuvant vector cells, while considering the benefits and drawbacks of each strategy. Additionally, we offer an in-depth analysis of adoptive T-cell therapies, highlighting innovative techniques such as next-generation NY-ESO-1 T-cell products and the integration with lymph node-targeted vaccines to address challenges and enhance therapeutic efficacy. Overall, this comprehensive review sheds light on the evolving landscape of NY-ESO-1 targeting and its potential implications for cancer treatment, opening avenues for future tailored directions in NY-ESO-1-specific immunotherapy. HIGHLIGHTS: Endogenous immune response: NY-ESO-1 exhibited high immunogenicity, activating endogenous dendritic cells, T cells and B cells. NY-ESO-1-based cancer vaccines: NY-ESO-1 vaccines using protein/peptide, RNA/DNA, microbial vectors and artificial adjuvant vector cells have shown promise in enhancing immune responses against tumours. NY-ESO-1-specific T-cell receptor-engineered cells: NY-ESO-1-targeted T cells, along with ongoing innovations in engineered natural killer cells and other cell therapies, have improved the efficacy of immunotherapy.
Collapse
Affiliation(s)
- Alaa Alsalloum
- Laboratory of Molecular ImmunologyFederal State Budgetary Scientific Institution Research Institute of Fundamental and Clinical ImmunologyNovosibirskRussia
- Faculty of Natural SciencesNovosibirsk State UniversityNovosibirskRussia
| | - Julia A. Shevchenko
- Laboratory of Molecular ImmunologyFederal State Budgetary Scientific Institution Research Institute of Fundamental and Clinical ImmunologyNovosibirskRussia
| | - Sergey Sennikov
- Laboratory of Molecular ImmunologyFederal State Budgetary Scientific Institution Research Institute of Fundamental and Clinical ImmunologyNovosibirskRussia
- Department of ImmunologyV. Zelman Institute for Medicine and PsychologyNovosibirsk State UniversityNovosibirskRussia
| |
Collapse
|
3
|
Li X, Li X, Zhang K, Guan Y, Fan M, Wu Q, Li Y, Holmdahl R, Lu S, Zhu W, Wang X, Meng L. Autoantibodies against Endophilin A2 as a novel biomarker are beneficial to early diagnosis of breast cancer. Clin Chim Acta 2024; 560:119748. [PMID: 38796051 DOI: 10.1016/j.cca.2024.119748] [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: 11/29/2023] [Revised: 03/24/2024] [Accepted: 05/22/2024] [Indexed: 05/28/2024]
Abstract
BACKGROUND Due to the lack of early symptoms, breast cancer is frequently overlooked, leading to distant metastases and multi-organ lesions that directly threaten patients' lives. We have identified a novel tumor marker, antibodies to endophilin A2 (EA2), to improve early diagnosis of breast cancer. METHODS Antibody levels of EA2 were analyzed in sera of patients with cancers of different origins and stages by indirect enzyme-linked immunosorbent assay (ELISA). Diagnostic accuracy and reference range were determined by the area under the receiver operating curve and distribution curve. The levels of EA2 antigen in sera were determined by sandwich ELISA. RESULTS The levels of antibodies against EA2 were higher in sera of patients with breast cancer (P < 0.0001), liver cancer (P = 0.0005), gastric cancer (P = 0.0026), and colon cancer (P = 0.0349) than those in healthy controls, but not in patients with rectal cancer (P = 0.1151), leukemia (P = 0.7508), or lung cancer (P = 0.2247). The highest diagnostic value was for breast cancer, particularly in early cases (AUC = 0.8014) and those with distant metastases (AUC = 0.7885). The titers of EA2 antibodies in sera were correlated with levels of EA2 antigen in breast cancer patients. CONCLUSION Antibodies to EA2 are novel blood biomarkers for early diagnosis of breast cancer that warrants further study in larger-scale cohort studies.
Collapse
Affiliation(s)
- Xiaomeng Li
- National-Local Joint Engineering Research Center of Biodiagnostics and Biotherapy, Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China; Institute of Molecular and Translational Medicine (IMTM), and Department of Biochemistry and Molecular Biology, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China
| | - Xiaowei Li
- National-Local Joint Engineering Research Center of Biodiagnostics and Biotherapy, Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Kaige Zhang
- School of Medical Technology, Xinxiang Medical University, Xinxiang, Henan 453003, China; Department of Clinical Laboratory, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Yanglong Guan
- Institute of Molecular and Translational Medicine (IMTM), and Department of Biochemistry and Molecular Biology, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China
| | - Meiyang Fan
- Institute of Molecular and Translational Medicine (IMTM), and Department of Biochemistry and Molecular Biology, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China
| | - Qian Wu
- Department of Clinical Laboratory, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Yue Li
- Institute of Molecular and Translational Medicine (IMTM), and Department of Biochemistry and Molecular Biology, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China
| | - Rikard Holmdahl
- National-Local Joint Engineering Research Center of Biodiagnostics and Biotherapy, Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China; Section for Medical Inflammation Research, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm 17177, Sweden
| | - Shemin Lu
- National-Local Joint Engineering Research Center of Biodiagnostics and Biotherapy, Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China; Institute of Molecular and Translational Medicine (IMTM), and Department of Biochemistry and Molecular Biology, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China; Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education, Xi'an, Shaanxi 710061, China
| | - Wenhua Zhu
- National-Local Joint Engineering Research Center of Biodiagnostics and Biotherapy, Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China; Institute of Molecular and Translational Medicine (IMTM), and Department of Biochemistry and Molecular Biology, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China.
| | - Xiaoqin Wang
- Department of Clinical Laboratory, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China.
| | - Liesu Meng
- National-Local Joint Engineering Research Center of Biodiagnostics and Biotherapy, Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China; Institute of Molecular and Translational Medicine (IMTM), and Department of Biochemistry and Molecular Biology, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China; Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education, Xi'an, Shaanxi 710061, China
| |
Collapse
|
4
|
Chen P, Chu Y, Liu R. Tumour-reactive plasma cells in antitumour immunity: current insights and future prospects. IMMUNOTHERAPY ADVANCES 2024; 4:ltae003. [PMID: 38736973 PMCID: PMC11088280 DOI: 10.1093/immadv/ltae003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 04/24/2024] [Indexed: 05/14/2024] Open
Abstract
Tumour-reactive plasma cells (TRPCs) have been reported to be positively associated with the long-term survival of patients with various cancers. However, unlike tumour-specific antigen (TSA)-induced T cells which have precise effects against tumours, plasma cells require TSA to obtain specific responses. Therefore, the search for a TSA suitable for B-cell recognition is urgent. In this review, we discuss the functions of tumour-reactive plasma cells. Further, this review also explores the concept of screening for neoantigen-reactive plasma cells, drawing inspiration from T-cell screening methods. While challenges exist, such as epitope prediction and efficient screening, the development of novel techniques may lead to the discovery of highly specific plasma cells for adoptive cell therapy. In conclusion, tumour-reactive plasma cells are emerging as powerful players in cancer immunotherapy. Their ability to produce antibodies against a variety of antigens, especially neoantigens, opens new avenues for personalised treatments. Overcoming challenges in epitope prediction and screening will be crucial in harnessing the full potential of these plasma cells for the benefit of cancer patients.
Collapse
Affiliation(s)
- Peng Chen
- Shanghai Fifth People’s Hospital and Key Laboratory of Medical Epigenetics and Metabolism, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Yiwei Chu
- Department of Immunology, School of Basic Medical Sciences, and MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan University, Shanghai, China
| | - Ronghua Liu
- Shanghai Fifth People’s Hospital and Key Laboratory of Medical Epigenetics and Metabolism, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| |
Collapse
|
5
|
Saito T, Kurokawa Y, Fujitani K, Kawabata R, Takeno A, Mikami J, Endo S, Matsuyama J, Akamaru Y, Hirota M, Kishi K, Urakawa S, Yamamoto K, Tanaka K, Takahashi T, Oka M, Wada H, Eguchi H, Doki Y. Serum NY-ESO-1 antibody as a predictive biomarker for postoperative recurrence of gastric cancer: a multicenter prospective observational study. Br J Cancer 2024; 130:1157-1165. [PMID: 38326601 PMCID: PMC10991393 DOI: 10.1038/s41416-023-02540-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 11/18/2023] [Accepted: 12/01/2023] [Indexed: 02/09/2024] Open
Abstract
BACKGROUND No reliable marker has been identified to predict postoperative recurrence of gastric cancer. We designed a clinical trial to investigate the utility of serum NY-ESO-1 antibody responses as a predictive marker for postoperative recurrence in gastric cancer. METHODS A multicenter prospective study was conducted between 2012 and 2021. Patients with resectable cT3-4 gastric cancer were included. Postoperative NY-ESO-1 and p53 antibody responses were serially evaluated every 3 months for 1 year in patients with positive preoperative antibody responses. The recurrence rate was assessed by the positivity of antibody responses at 3 and 12 months postoperatively. RESULTS Among 1001 patients, preoperative NY-ESO-1 and p53 antibody responses were positive in 12.6% and 18.1% of patients, respectively. NY-ESO-1 antibody responses became negative postoperatively in non-recurrent patients (negativity rates; 45% and 78% at 3 and 12 months, respectively), but remained positive in recurrent patients (negativity rates; 9% and 8%, respectively). p53 antibody responses remained positive in non-recurrent patients. In multivariate analysis, NY-ESO-1 antibody positivity at 3 months (P < 0.03) and 12 months (P < 0.001) were independent prognostic factors for a shorter recurrence-free interval. CONCLUSIONS Serum NY-ESO-1 antibodies may be a useful predictive marker for postoperative recurrence in gastric cancer. CLINICAL TRIAL REGISTRATION UMIN000007925.
Collapse
Affiliation(s)
- Takuro Saito
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Suita, Japan.
| | - Yukinori Kurokawa
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Suita, Japan
| | - Kazumasa Fujitani
- Department of Gastroenterological Surgery, Osaka General Medical Center, Osaka, Japan
| | | | - Atsushi Takeno
- Department of Surgery, Kansai Rosai Hospital, Amagasaki, Japan
| | - Jota Mikami
- Department of Surgery, Sakai City Medical Center, Sakai, Japan
| | - Shunji Endo
- Department of Surgery, Higashi-Osaka Medical Center, Higashi-Osaka, Japan
| | - Jin Matsuyama
- Department of Surgery, Yao Municipal Hospital, Yao, Japan
| | - Yusuke Akamaru
- Department of Surgery, Ikeda City Hospital, Osaka, Japan
| | - Masashi Hirota
- Department of Surgery, Toyonaka Municipal Hospital, Toyonaka, Japan
| | - Kentaro Kishi
- Department of Surgery, Osaka Police Hospital, Osaka, Japan
| | - Shinya Urakawa
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Suita, Japan
| | - Kei Yamamoto
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Suita, Japan
| | - Koji Tanaka
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Suita, Japan
| | - Tsuyoshi Takahashi
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Suita, Japan
| | - Mikio Oka
- Department of Immuno-Oncology, Kawasaki Medical School, Okayama, Japan
| | - Hisashi Wada
- Department of Clinical Research in Tumor Immunology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Hidetoshi Eguchi
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Suita, Japan
| | - Yuichiro Doki
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Suita, Japan
| |
Collapse
|
6
|
Khalid S, Guo J, Muhammad SA, Bai B. Designing, cloning and simulation studies of cancer/testis antigens based multi-epitope vaccine candidates against cutaneous melanoma: An immunoinformatics approach. Biochem Biophys Rep 2024; 37:101651. [PMID: 38371523 PMCID: PMC10873875 DOI: 10.1016/j.bbrep.2024.101651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 01/18/2024] [Accepted: 01/18/2024] [Indexed: 02/20/2024] Open
Abstract
Background Melanoma is the most fatal kind of skin cancer. Among its various types, cutaneous melanoma is the most prevalent one. Melanoma cells are thought to be highly immunogenic due to the presence of distinct tumor-associated antigens (TAAs), which includes carcinoembryonic antigen (CEA), cancer/testis antigens (CTAs) and neo-antigens. The CTA family is a group of antigens that are only expressed in malignancies and testicular germ cells. Methods We used integrative framework and systems-level analysis to predict potential vaccine candidates for cutaneous melanoma involving epitopes prediction, molecular modeling and molecular docking to cross-validate the binding affinity and interaction between potential vaccine agents and major histocompatibility molecules (MHCs) followed by molecular dynamics simulation, immune simulation and in silico cloning. Results In this study, three cancer/testis antigens were targeted for immunotherapy of cutaneous melanoma. Among many CTAs that were studied for their expression in primary and malignant melanoma, NY-ESO-1, MAGE1 and SSX2 antigens are most prevalent in cutaneous melanoma. Cytotoxic and Helper epitopes were predicted, and the finest epitopes were shortlisted based on binding score. The vaccine construct was composed of the four epitope-rich domains of antigenic proteins, an appropriate adjuvant, His tag and linkers. This potential multi-epitope vaccine was further evaluated in terms of antigenicity, allergencity, toxicity and other physicochemical properties. Molecular interaction estimated through protein-protein docking unveiled good interactions characterized by favorable binding energies. Molecular dynamics simulation ensured the stability of docked complex and the predicted immune response through immune simulation revealed elevated levels of antibodies titer, cytokines, interleukins and immune cells (NK, DC and MA) population. Conclusion The findings indicate that the potential vaccine candidates could be effective immunotherapeutic agents that modify the treatment strategies of cutaneous melanoma.
Collapse
Affiliation(s)
- Sana Khalid
- Institute of Molecular Biology and Biotechnology, Bahauddin Zakariya University Multan, Pakistan
| | - Jinlei Guo
- School of Intelligent Medical Engineering, Sanquan College of Xinxiang Medical University, Xinxiang, China
| | - Syed Aun Muhammad
- Institute of Molecular Biology and Biotechnology, Bahauddin Zakariya University Multan, Pakistan
| | - Baogang Bai
- School of Information and Technology, Wenzhou Business College, Wenzhou, China
- Zhejiang Province Engineering Research Center of Intelligent Medicine, Wenzhou, China
- The 1st School of Medical, School of Information and Engineering, The 1st Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
7
|
Kawada J, Saito T, Kurokawa Y, Kawabata R, Takeno A, Takeoka T, Nose Y, Wada H, Eguchi H, Doki Y, Osaka University Clinical Research Group for Gastroenterological Study. Serum NY-ESO-1 and p53 antibodies as useful tumor markers in gastric cancer. Ann Gastroenterol Surg 2024; 8:243-250. [PMID: 38455491 PMCID: PMC10914697 DOI: 10.1002/ags3.12757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 10/17/2023] [Accepted: 10/30/2023] [Indexed: 03/09/2024] Open
Abstract
Purpose The NY-ESO-1 antigen is highly immunogenic and often spontaneously induces an immune response in patients with cancer. We conducted a large-scale multicenter cohort study to investigate the utility of serum NY-ESO-1 and p53 antibodies as predictive markers for the postoperative recurrence of gastric cancer. Here, we examined the usefulness of pre-treatment NY-ESO-1 and p53 antibodies as tumor markers for the diagnosis of gastric cancer in combination with carcinoembryonic antigen (CEA) and carbohydrate antigen 19-9 (CA19-9). Methods A total of 1031 patients with cT3-4 gastric cancer were enrolled in the study. NY-ESO-1 and p53 antibodies were assessed prior to treatment. The positivity of NY-ESO-1 and p53 antibodies, CEA, and CA19-9 was evaluated before treatment. Results Serum NY-ESO-1 and p53 antibodies were positive in 12.6% and 18.1% of the patients, respectively. Positive NY-ESO-1 antibody response was correlated with male gender, higher cStage, and upper tumor location. However, a positive p53 antibody response was not associated with tumor factors. The combination of NY-ESO-1 or p53 antibody response with CEA and CA19-9, or the 4-factors, was positive in 45.1%, 49.6%, and 53.8% of patients, respectively. Moreover, the 4-factor combination was able to detect >60% of cStage III-IV diseases, which was 14% higher than that with the combination of CEA and CA19-9. Conclusion The combination of NY-ESO-1 and p53 antibody responses to CEA and CA19-9 increases the diagnostic accuracy of gastric cancer. Serum NY-ESO-1 and p53 antibodies may be useful tumor markers for gastric cancer.
Collapse
Affiliation(s)
- Junji Kawada
- Department of Gastroenterological SurgeryOsaka General Medical CenterOsakaJapan
- Department of SurgeryKaizuka City HospitalOsakaJapan
| | - Takuro Saito
- Department of Gastroenterological SurgeryOsaka University Graduate School of MedicineSuitaJapan
| | - Yukinori Kurokawa
- Department of Gastroenterological SurgeryOsaka University Graduate School of MedicineSuitaJapan
| | | | - Atsushi Takeno
- Department of SurgeryKansai Rosai HospitalAmagasakiJapan
| | - Tomohira Takeoka
- Department of SurgeryHyogo Prefectural Nishinomiya HospitalNishinomiyaJapan
| | - Yohei Nose
- Department of Gastroenterological SurgeryOsaka University Graduate School of MedicineSuitaJapan
| | - Hisashi Wada
- Department of Clinical Research in Tumor ImmunologyOsaka University Graduate School of MedicineOsakaJapan
| | - Hidetoshi Eguchi
- Department of Gastroenterological SurgeryOsaka University Graduate School of MedicineSuitaJapan
| | - Yuichiro Doki
- Department of Gastroenterological SurgeryOsaka University Graduate School of MedicineSuitaJapan
| | | |
Collapse
|
8
|
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
|
9
|
Okada S, Muraoka D, Yasui K, Tawara I, Kawamura A, Okamoto S, Mineno J, Seo N, Shiku H, Eguchi S, Ikeda H. T cell receptor gene-modified allogeneic T cells with siRNA for endogenous T cell receptor induce efficient tumor regression without graft-versus-host disease. Cancer Sci 2023; 114:4172-4183. [PMID: 37675556 PMCID: PMC10637063 DOI: 10.1111/cas.15954] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 08/04/2023] [Accepted: 08/23/2023] [Indexed: 09/08/2023] Open
Abstract
Adoptive immunotherapy using genetically engineered patient-derived lymphocytes to express tumor-reactive receptors is a promising treatment for malignancy. However, utilization of autologous T cells in this therapy limits the quality of gene-engineered T cells, thereby inhibiting the timely infusion of the cells into patients. In this study, we evaluated the anti-tumor efficacy and the potential to induce graft-versus-host disease (GVHD) in T cell receptor (TCR) gene-engineered allogeneic T cells that downregulate the endogenous TCR and HLA class I molecules with the aim of developing an "off-the-shelf" cell product with expanded application of genetically engineered T cells. We transduced human lymphocytes with a high-affinity TCR specific to the cancer/testis antigen NY-ESO-1 using a novel retrovirus vector with siRNAs specific to the endogenous TCR (siTCR vector). These T cells showed reduced expression of endogenous TCR and minimized reactivity to allogeneic cells in vitro. In non-obese diabetic/SCID/γcnull mice, TCR gene-transduced T cells induced tumor regression without development of GVHD. A lentivirus-based CRISPR/Cas9 system targeting β-2 microglobulin in TCR gene-modified T cells silenced the HLA class I expression and prevented allogeneic CD8+ T cell stimulation without disrupting their anti-tumor capacity. This report is the first demonstration that siTCR technology is effective in preventing GVHD. Adoptive cell therapy with allogeneic T cells engineered with siTCR vector may be useful in developing an "off-the-shelf" therapy for patients with malignancy.
Collapse
Affiliation(s)
- Satomi Okada
- Department of OncologyNagasaki University Graduate School of Biomedical SciencesNagasakiJapan
- Department of SurgeryNagasaki University Graduate School of Biomedical SciencesNagasakiJapan
| | - Daisuke Muraoka
- Department of OncologyNagasaki University Graduate School of Biomedical SciencesNagasakiJapan
- Division of Translational OncoimmunologyAichi Cancer Center Research InstituteNagoyaJapan
| | - Kiyoshi Yasui
- Department of OncologyNagasaki University Graduate School of Biomedical SciencesNagasakiJapan
| | - Isao Tawara
- Department of Hematology and OncologyMie University Graduate School of MedicineMieJapan
| | | | | | | | - Naohiro Seo
- Department of Personalized Cancer ImmunotherapyMie University Graduate School of MedicineMieJapan
- Department of Bioengineering, School of EngineeringThe University of TokyoTokyoJapan
| | - Hiroshi Shiku
- Department of Personalized Cancer ImmunotherapyMie University Graduate School of MedicineMieJapan
| | - Susumu Eguchi
- Department of SurgeryNagasaki University Graduate School of Biomedical SciencesNagasakiJapan
| | - Hiroaki Ikeda
- Department of OncologyNagasaki University Graduate School of Biomedical SciencesNagasakiJapan
- Leading Medical Research Core UnitNagasaki University Graduate School of Biomedical SciencesNagasakiJapan
| |
Collapse
|
10
|
Crescioli S, Correa I, Ng J, Willsmore ZN, Laddach R, Chenoweth A, Chauhan J, Di Meo A, Stewart A, Kalliolia E, Alberts E, Adams R, Harris RJ, Mele S, Pellizzari G, Black ABM, Bax HJ, Cheung A, Nakamura M, Hoffmann RM, Terranova-Barberio M, Ali N, Batruch I, Soosaipillai A, Prassas I, Ulndreaj A, Chatanaka MK, Nuamah R, Kannambath S, Dhami P, Geh JLC, MacKenzie Ross AD, Healy C, Grigoriadis A, Kipling D, Karagiannis P, Dunn-Walters DK, Diamandis EP, Tsoka S, Spicer J, Lacy KE, Fraternali F, Karagiannis SN. B cell profiles, antibody repertoire and reactivity reveal dysregulated responses with autoimmune features in melanoma. Nat Commun 2023; 14:3378. [PMID: 37291228 PMCID: PMC10249578 DOI: 10.1038/s41467-023-39042-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 05/23/2023] [Indexed: 06/10/2023] Open
Abstract
B cells are known to contribute to the anti-tumor immune response, especially in immunogenic tumors such as melanoma, yet humoral immunity has not been characterized in these cancers to detail. Here we show comprehensive phenotyping in samples of circulating and tumor-resident B cells as well as serum antibodies in melanoma patients. Memory B cells are enriched in tumors compared to blood in paired samples and feature distinct antibody repertoires, linked to specific isotypes. Tumor-associated B cells undergo clonal expansion, class switch recombination, somatic hypermutation and receptor revision. Compared with blood, tumor-associated B cells produce antibodies with proportionally higher levels of unproductive sequences and distinct complementarity determining region 3 properties. The observed features are signs of affinity maturation and polyreactivity and suggest an active and aberrant autoimmune-like reaction in the tumor microenvironment. Consistent with this, tumor-derived antibodies are polyreactive and characterized by autoantigen recognition. Serum antibodies show reactivity to antigens attributed to autoimmune diseases and cancer, and their levels are higher in patients with active disease compared to post-resection state. Our findings thus reveal B cell lineage dysregulation with distinct antibody repertoire and specificity, alongside clonally-expanded tumor-infiltrating B cells with autoimmune-like features, shaping the humoral immune response in melanoma.
Collapse
Affiliation(s)
- Silvia Crescioli
- St John's Institute of Dermatology, School of Basic & Medical Biosciences, King's College London, Guy's Hospital, London, UK
| | - Isabel Correa
- St John's Institute of Dermatology, School of Basic & Medical Biosciences, King's College London, Guy's Hospital, London, UK
| | - Joseph Ng
- Randall Centre for Cell and Molecular Biophysics, King's College London, London, UK
- Research Department of Structural and Molecular Biology, University College London, London, UK
| | - Zena N Willsmore
- St John's Institute of Dermatology, School of Basic & Medical Biosciences, King's College London, Guy's Hospital, London, UK
| | - Roman Laddach
- St John's Institute of Dermatology, School of Basic & Medical Biosciences, King's College London, Guy's Hospital, London, UK
- Department of Informatics, Faculty of Natural, Mathematical and Engineering Sciences, King's College London, London, UK
| | - Alicia Chenoweth
- St John's Institute of Dermatology, School of Basic & Medical Biosciences, King's College London, Guy's Hospital, London, UK
- Breast Cancer Now Research Unit, School of Cancer & Pharmaceutical Sciences, King's College London, Guy's Hospital, London, UK
| | - Jitesh Chauhan
- St John's Institute of Dermatology, School of Basic & Medical Biosciences, King's College London, Guy's Hospital, London, UK
| | - Ashley Di Meo
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
| | - Alexander Stewart
- School of Biosciences and Medicine, University of Surrey, Guildford, UK
| | - Eleni Kalliolia
- St John's Institute of Dermatology, School of Basic & Medical Biosciences, King's College London, Guy's Hospital, London, UK
| | - Elena Alberts
- Breast Cancer Now Research Unit, School of Cancer & Pharmaceutical Sciences, King's College London, Guy's Hospital, London, UK
| | - Rebecca Adams
- St John's Institute of Dermatology, School of Basic & Medical Biosciences, King's College London, Guy's Hospital, London, UK
| | - Robert J Harris
- St John's Institute of Dermatology, School of Basic & Medical Biosciences, King's College London, Guy's Hospital, London, UK
| | - Silvia Mele
- St John's Institute of Dermatology, School of Basic & Medical Biosciences, King's College London, Guy's Hospital, London, UK
| | - Giulia Pellizzari
- St John's Institute of Dermatology, School of Basic & Medical Biosciences, King's College London, Guy's Hospital, London, UK
| | - Anna B M Black
- St John's Institute of Dermatology, School of Basic & Medical Biosciences, King's College London, Guy's Hospital, London, UK
| | - Heather J Bax
- St John's Institute of Dermatology, School of Basic & Medical Biosciences, King's College London, Guy's Hospital, London, UK
| | - Anthony Cheung
- St John's Institute of Dermatology, School of Basic & Medical Biosciences, King's College London, Guy's Hospital, London, UK
- Breast Cancer Now Research Unit, School of Cancer & Pharmaceutical Sciences, King's College London, Guy's Hospital, London, UK
| | - Mano Nakamura
- St John's Institute of Dermatology, School of Basic & Medical Biosciences, King's College London, Guy's Hospital, London, UK
| | - Ricarda M Hoffmann
- St John's Institute of Dermatology, School of Basic & Medical Biosciences, King's College London, Guy's Hospital, London, UK
| | - Manuela Terranova-Barberio
- St John's Institute of Dermatology, School of Basic & Medical Biosciences, King's College London, Guy's Hospital, London, UK
| | - Niwa Ali
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, Faculty of Life Sciences and Medicine, King's College London, London, UK
- Centre for Gene Therapy and Regenerative Medicine, School of Basic and Medical Biosciences, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Ihor Batruch
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
| | | | - Ioannis Prassas
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
| | - Antigona Ulndreaj
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
| | - Miyo K Chatanaka
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Rosamund Nuamah
- Biomedical Research Centre, Guy's and St. Thomas' NHS Foundation Trust, London, UK
| | - Shichina Kannambath
- Biomedical Research Centre, Guy's and St. Thomas' NHS Foundation Trust, London, UK
- Genomics Facility, Institute of Cancer Research, London, UK
| | - Pawan Dhami
- Biomedical Research Centre, Guy's and St. Thomas' NHS Foundation Trust, London, UK
| | - Jenny L C Geh
- St John's Institute of Dermatology, Guy's, King's, and St. Thomas' Hospitals NHS Foundation Trust, London, UK
- Department of Plastic Surgery at Guy's and St. Thomas' NHS Foundation Trust, London, UK
| | | | - Ciaran Healy
- Department of Plastic Surgery at Guy's and St. Thomas' NHS Foundation Trust, London, UK
| | - Anita Grigoriadis
- Breast Cancer Now Research Unit, School of Cancer & Pharmaceutical Sciences, King's College London, Guy's Hospital, London, UK
| | - David Kipling
- School of Biosciences and Medicine, University of Surrey, Guildford, UK
| | - Panagiotis Karagiannis
- St John's Institute of Dermatology, School of Basic & Medical Biosciences, King's College London, Guy's Hospital, London, UK
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | | | - Eleftherios P Diamandis
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, ON, Canada
- Department of Clinical Biochemistry, University Health Network, Toronto, ON, Canada
| | - Sophia Tsoka
- Department of Informatics, Faculty of Natural, Mathematical and Engineering Sciences, King's College London, London, UK
| | - James Spicer
- School of Cancer & Pharmaceutical Sciences, King's College London, Guy's Hospital, London, UK
| | - Katie E Lacy
- St John's Institute of Dermatology, School of Basic & Medical Biosciences, King's College London, Guy's Hospital, London, UK
| | - Franca Fraternali
- Randall Centre for Cell and Molecular Biophysics, King's College London, London, UK
- Research Department of Structural and Molecular Biology, University College London, London, UK
| | - Sophia N Karagiannis
- St John's Institute of Dermatology, School of Basic & Medical Biosciences, King's College London, Guy's Hospital, London, UK.
- Breast Cancer Now Research Unit, School of Cancer & Pharmaceutical Sciences, King's College London, Guy's Hospital, London, UK.
| |
Collapse
|
11
|
Rao Bommi J, Kummari S, Lakavath K, Sukumaran RA, Panicker LR, Marty JL, Yugender Goud K. Recent Trends in Biosensing and Diagnostic Methods for Novel Cancer Biomarkers. BIOSENSORS 2023; 13:398. [PMID: 36979610 PMCID: PMC10046866 DOI: 10.3390/bios13030398] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 03/13/2023] [Accepted: 03/15/2023] [Indexed: 06/18/2023]
Abstract
Cancer is one of the major public health issues in the world. It has become the second leading cause of death, with approximately 75% of cancer deaths transpiring in low- or middle-income countries. It causes a heavy global economic cost estimated at more than a trillion dollars per year. The most common cancers are breast, colon, rectum, prostate, and lung cancers. Many of these cancers can be treated effectively and cured if detected at the primary stage. Nowadays, around 50% of cancers are detected at late stages, leading to serious health complications and death. Early diagnosis of cancer diseases substantially increases the efficient treatment and high chances of survival. Biosensors are one of the potential screening methodologies useful in the early screening of cancer biomarkers. This review summarizes the recent findings about novel cancer biomarkers and their advantages over traditional biomarkers, and novel biosensing and diagnostic methods for them; thus, this review may be helpful in the early recognition and monitoring of treatment response of various human cancers.
Collapse
Affiliation(s)
| | - Shekher Kummari
- Department of Chemistry, Indian Institute of Technology Palakkad, Palakkad 678 557, Kerala, India
| | - Kavitha Lakavath
- Department of Chemistry, Indian Institute of Technology Palakkad, Palakkad 678 557, Kerala, India
| | - Reshmi A. Sukumaran
- Department of Chemistry, Indian Institute of Technology Palakkad, Palakkad 678 557, Kerala, India
| | - Lakshmi R. Panicker
- Department of Chemistry, Indian Institute of Technology Palakkad, Palakkad 678 557, Kerala, India
| | - Jean Louis Marty
- Université de Perpignan Via Domitia, 52 Avenue Paul Alduy, 66860 Perpignan, France
| | - Kotagiri Yugender Goud
- Department of Chemistry, Indian Institute of Technology Palakkad, Palakkad 678 557, Kerala, India
| |
Collapse
|
12
|
Nin DS, Deng LW. Biology of Cancer-Testis Antigens and Their Therapeutic Implications in Cancer. Cells 2023; 12:cells12060926. [PMID: 36980267 PMCID: PMC10047177 DOI: 10.3390/cells12060926] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 03/14/2023] [Accepted: 03/15/2023] [Indexed: 03/30/2023] Open
Abstract
Tumour-specific antigens have been an area of interest in cancer therapy since their discovery in the middle of the 20th century. In the era of immune-based cancer therapeutics, redirecting our immune cells to target these tumour-specific antigens has become even more relevant. Cancer-testis antigens (CTAs) are a class of antigens with an expression specific to the testis and cancer cells. CTAs have also been demonstrated to be expressed in a wide variety of cancers. Due to their frequency and specificity of expression in a multitude of cancers, CTAs have been particularly attractive as cancer-specific therapeutic targets. There is now a rapid expansion of CTAs being identified and many studies have been conducted to correlate CTA expression with cancer and therapy-resistant phenotypes. Furthermore, there is an increasing number of clinical trials involving using some of these CTAs as molecular targets in pharmacological and immune-targeted therapeutics for various cancers. This review will summarise the current knowledge of the biology of known CTAs in tumorigenesis and the regulation of CTA genes. CTAs as molecular targets and the therapeutic implications of these CTA-targeted anticancer strategies will also be discussed.
Collapse
Affiliation(s)
- Dawn Sijin Nin
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, MD 7, 8 Medical Drive, Singapore 117596, Singapore
- NUS Center for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, 14 Medical Drive, Singapore 117599, Singapore
| | - Lih-Wen Deng
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, MD 7, 8 Medical Drive, Singapore 117596, Singapore
- NUS Center for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, 14 Medical Drive, Singapore 117599, Singapore
- National University Cancer Institute, National University Health System, 5 Lower Kent Ridge Road, Singapore 119074, Singapore
| |
Collapse
|
13
|
de Joode K, Veenbergen S, Kransse C, Kortleve D, Debets R, Mathijssen RHJ, Joosse A, Schreurs MWJ, Van der Veldt AAM. Suitability of tumor-associated antibodies as predictive biomarker for response to immune checkpoint inhibitors in patients with melanoma: a short report. J Immunother Cancer 2023; 11:jitc-2022-006467. [PMID: 36750254 PMCID: PMC9906380 DOI: 10.1136/jitc-2022-006467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/13/2023] [Indexed: 02/09/2023] Open
Abstract
In 2019, Fässler et al showed in this journal that the presence of tumor-associated antibodies correlated with response to immune checkpoint inhibitor treatment in patients with metastatic melanoma. The results of this study suggested that tumor-associated antibodies directed against melanocyte-differentiation antigens and the cancer-germline antigen NY-ESO-1 should be further investigated as candidate biomarkers for response to immune checkpoint inhibitors. The aim of the current study was to validate and extend these previous findings. Therefore, we examined the correlation between serum levels of tumor-associated antibodies and tumor response after treatment with immune checkpoint inhibitors in patients with metastatic melanoma.All patients included in this prospective study were diagnosed with advanced stage melanoma and treated with nivolumab or pembrolizumab monotherapy. Blood samples were collected before and during treatment. Serum levels of tumor-associated antibodies against the melanocyte differentiation antigen Melan-A and the cancer germline antigens NY-ESO-1, MAGE-C2, MAGE-A6 and ROPN1B were measured at baseline and during treatment. Differences between responders and non-responders were assessed using the Mann-Whitney U-test, and differences between different overall survival categories with the Kruskal-Wallis test. P values ≤0.05 were considered significant.Serum samples of 58 patients with advanced melanoma with long-term follow-up (>3 years) were collected. In contrast to the findings of Fässler et al, for all antibodies tested, we found no significant differences between serum levels of responders and non-responders before or during treatment with immune checkpoint inhibitors. In addition, no significant differences were found in serum levels of tumor-associated antibodies for different overall survival groups.Although our study included a larger and more mature cohort of patients with longer follow-up, we could not externally validate the findings of Fässler et al In addition, we were not able to identify other cancer germline antigens as predictive biomarkers of response to immune checkpoint inhibitors in patients advanced melanoma. Based on the results of the present study, clinical applicability of tumor-associated antibodies directed against tumor antigens as predictive biomarkers for immune checkpoint inhibitors in patients with advanced melanoma is not feasible.
Collapse
Affiliation(s)
- Karlijn de Joode
- Department of Medical Oncology, Erasmus MC Cancer Institute, University Medical Center, Rotterdam, The Netherlands
| | - Sharon Veenbergen
- Laboratory of Medical Immunology, Department of Immunology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Claudia Kransse
- Laboratory of Medical Immunology, Department of Immunology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Dian Kortleve
- Department of Medical Oncology, Erasmus MC Cancer Institute, University Medical Center, Rotterdam, The Netherlands
| | - Reno Debets
- Department of Medical Oncology, Erasmus MC Cancer Institute, University Medical Center, Rotterdam, The Netherlands
| | - Ron H J Mathijssen
- Department of Medical Oncology, Erasmus MC Cancer Institute, University Medical Center, Rotterdam, The Netherlands
| | - Arjen Joosse
- Department of Medical Oncology, Erasmus MC Cancer Institute, University Medical Center, Rotterdam, The Netherlands
| | - Marco W J Schreurs
- Laboratory of Medical Immunology, Department of Immunology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Astrid A M Van der Veldt
- Department of Medical Oncology, Erasmus MC Cancer Institute, University Medical Center, Rotterdam, The Netherlands .,Department of Radiology & Nuclear Medicine, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
14
|
Itoh M, Kawagoe S, Nakagawa H, Asahina A, Okano HJ. Generation of induced pluripotent stem cell (iPSC) from NY-ESO-I-specific cytotoxic T cells isolated from the melanoma patient with minor HLAs: The practical pilot study for the adoptive immunotherapy for melanoma using iPSC technology. Exp Dermatol 2023; 32:126-134. [PMID: 36222007 DOI: 10.1111/exd.14686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 09/23/2022] [Accepted: 10/10/2022] [Indexed: 11/29/2022]
Abstract
Melanoma is one of the most severe skin cancers, derived from melanocytes. Among various therapies for melanoma, adoptive immunotherapy using tumor-infiltrating lymphocytes/chimeric antigen receptor-T cells (TCs) is advanced in recent years; however, the efficacy is still limited, and major challenges remain in terms of safety and cell supply. To solve the issues of adoptive immunotherapy, we utilized induced pluripotent stem cells (iPSCs), which have an unlimited proliferative ability and various differentiation capability. First, we monoclonally isolated CD8+ TCs specifically reactive with NY-ESO-1, one of tumor antigens, from the melanoma patient's monocytes after stimulated with NY-ESO-1 peptide by manual procedure, and cultured NY-ESO-1-specific TCs until proliferated and formed colonies. iPSCs were consequently generated from colony-forming TCs by exogenous expression of reprogramming factors using Sendai virus vector. After the RAG2 gene in TC-derived iPSCs (T-iPSCs) was knocked out for preventing T-cell receptor (TCR) rearrangement, T-iPSCs were re-differentiated into rejuvenated cytotoxic TCs. We confirmed that TCR of T-iPSC-derived TC was maintained as the same of original TCs. In conclusion, T-iPSCs have a potential to be an unlimited cell source for providing cytotoxic TCs. Our study could be a "touchstone" to develop iPSC-based adoptive immunotherapy for the treatment of melanoma for the future clinical use.
Collapse
Affiliation(s)
- Munenari Itoh
- The Jikei University School of Medicine, Department of Dermatology, Tokyo, Japan
| | - Shiho Kawagoe
- The Jikei University School of Medicine, Department of Dermatology, Tokyo, Japan
| | - Hidemi Nakagawa
- The Jikei University School of Medicine, Department of Dermatology, Tokyo, Japan
| | - Akihiko Asahina
- The Jikei University School of Medicine, Department of Dermatology, Tokyo, Japan
| | - Hirotaka James Okano
- The Jikei University School of Medicine, Division of Regenerative Medicine, Tokyo, Japan
| |
Collapse
|
15
|
Noordam L, de Beijer MT, Mancham S, Vogler I, Boor PP, de Ruiter V, Luijten R, IJzermans JN, Sahin U, Bruno MJ, Sprengers D, Buschow SI, Kwekkeboom J. Systemic T-cell and humoral responses against cancer testis antigens in hepatocellular carcinoma patients. Oncoimmunology 2022; 11:2131096. [PMID: 36211805 PMCID: PMC9542711 DOI: 10.1080/2162402x.2022.2131096] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the fourth leading cause of cancer-related deaths worldwide due to high recurrence rates after curative treatment and being frequently diagnosed at an advanced stage. Immune-checkpoint inhibition (ICPI) has yielded impressive clinical successes in a variety of solid cancers, however results in treatment of HCC have been modest. Vaccination could be a promising treatment to synergize with ICPI and enhance response rates. Cancer testis antigens (CTAs) were recently discovered to be widely expressed in HCC and expression in macroscopically tumor-free tissues correlated with recurrence, implying the presence of micro-satellites. To determine whether CTAs are immunogenic in HCC patients, we analyzed systemic T-cell and humoral responses against seven CTAs in 38 HCC patients using a multitude of techniques; flowcytometry, ELISA and whole antigen and peptide stimulation assays. CTA-specific T-cells were detected in all (25/25) analyzed patients, of which most had a memory phenotype but did not exhibit unequivocal signs of chronic stimulation or recent antigen encounter. Proliferative CD4+ and CD8+ T-cell responses against these CTAs were found in 14/16 analyzed HCC patients. CTA-peptide stimulation-induced granzyme B, IL2, and TNFa in 8/8 analyzed patients, including two MAGEA1 peptides included based on in silico prediction. Finally, IgG responses were observed in 13/32 patients, albeit with low titers. The presence of CD4+ and CD8+ T-cells and IgG responses shows the immunogenicity of these CTAs in HCC-patients. We hypothesize that vaccines based on these tumor-specific antigens may boost preexisting CTA-specific immunity and could enhance therapeutic efficacy of ICPI in advanced HCC.
Collapse
Affiliation(s)
- Lisanne Noordam
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, The Netherlands
| | - Monique T.A. de Beijer
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, The Netherlands
| | - Shanta Mancham
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, The Netherlands
| | | | - Patrick P.C. Boor
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, The Netherlands
| | - Valeska de Ruiter
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, The Netherlands
| | - Robbie Luijten
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, The Netherlands
| | - Jan N.M. IJzermans
- Department of Surgery, Erasmus MC-University Medical Center, Rotterdam, The Netherlands
| | | | - Marco J. Bruno
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, The Netherlands
| | - Dave Sprengers
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, The Netherlands
| | - Sonja I. Buschow
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, The Netherlands
| | - Jaap Kwekkeboom
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
16
|
Thelen M, Keller D, Lehmann J, Wennhold K, Weitz H, Bauer E, Gathof B, Brüggemann M, Kotrova M, Quaas A, Mallmann C, Chon SH, Hillmer AM, Bruns C, von Bergwelt-Baildon M, Garcia-Marquez MA, Schlößer HA. Immune responses against shared antigens are common in esophago-gastric cancer and can be enhanced using CD40-activated B cells. J Immunother Cancer 2022; 10:jitc-2022-005200. [PMID: 36600602 PMCID: PMC9743382 DOI: 10.1136/jitc-2022-005200] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/19/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Specific immune response is a hallmark of cancer immunotherapy and shared tumor-associated antigens (TAAs) are important targets. Recent advances using combined cellular therapy against multiple TAAs renewed the interest in this class of antigens. Our study aims to determine the role of TAAs in esophago-gastric adenocarcinoma (EGA). METHODS RNA expression was assessed by NanoString in tumor samples of 41 treatment-naïve EGA patients. Endogenous T cell and antibody responses against the 10 most relevant TAAs were determined by FluoroSpot and protein-bound bead assays. Digital image analysis was used to evaluate the correlation of TAAs and T-cell abundance. T-cell receptor sequencing, in vitro expansion with autologous CD40-activated B cells (CD40Bs) and in vitro cytotoxicity assays were applied to determine specific expansion, clonality and cytotoxic activity of expanded T cells. RESULTS 68.3% of patients expressed ≥5 TAAs simultaneously with coregulated clusters, which were similar to data from The Cancer Genome Atlas (n=505). Endogenous cellular or humoral responses against ≥1 TAA were detectable in 75.0% and 53.7% of patients, respectively. We found a correlation of T-cell abundance and the expression of TAAs and genes related to antigen presentation. TAA-specific T-cell responses were polyclonal, could be induced or enhanced using autologous CD40Bs and were cytotoxic in vitro. Despite the frequent expression of TAAs co-occurrence with immune responses was rare. CONCLUSIONS We identified the most relevant TAAs in EGA for monitoring of clinical trials and as therapeutic targets. Antigen-escape rather than missing immune response should be considered as mechanism underlying immunotherapy resistance of EGA.
Collapse
Affiliation(s)
- Martin Thelen
- Center for Molecular Medicine Cologne, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Diandra Keller
- Center for Molecular Medicine Cologne, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Jonas Lehmann
- Center for Molecular Medicine Cologne, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Kerstin Wennhold
- Center for Molecular Medicine Cologne, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Hendrik Weitz
- Center for Molecular Medicine Cologne, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Eugen Bauer
- Institute of Transfusion Medicine, University of Cologne, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Birgit Gathof
- Institute of Transfusion Medicine, University of Cologne, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Monika Brüggemann
- Klinik für Innere Medizin II, Universitätsklinikum Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Michaela Kotrova
- Klinik für Innere Medizin II, Universitätsklinikum Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Alexander Quaas
- Institute of Pathology, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Christoph Mallmann
- Department of General, Visceral, Cancer and Transplantation Surgery, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Seung-Hun Chon
- Department of General, Visceral, Cancer and Transplantation Surgery, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Axel M Hillmer
- Institute of Pathology, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Christiane Bruns
- Department of General, Visceral, Cancer and Transplantation Surgery, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Michael von Bergwelt-Baildon
- Department of Internal Medicine III, University Hospital, Ludwig Maximilians University Munich, München, Germany,German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Maria Alejandra Garcia-Marquez
- Center for Molecular Medicine Cologne, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Hans Anton Schlößer
- Center for Molecular Medicine Cologne, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany,Department of General, Visceral, Cancer and Transplantation Surgery, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| |
Collapse
|
17
|
Yang R, Han Y, Yi W, Long Q. Autoantibodies as biomarkers for breast cancer diagnosis and prognosis. Front Immunol 2022; 13:1035402. [PMID: 36451832 PMCID: PMC9701846 DOI: 10.3389/fimmu.2022.1035402] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 10/28/2022] [Indexed: 10/07/2023] Open
Abstract
Breast cancer is the most common cancer in women worldwide and is a substantial public health problem. Screening for breast cancer mainly relies on mammography, which leads to false positives and missed diagnoses and is especially non-sensitive for patients with small tumors and dense breasts. The prognosis of breast cancer is mainly classified by tumor, node, and metastasis (TNM) staging, but this method does not consider the molecular characteristics of the tumor. As the product of the immune response to tumor-associated antigens, autoantibodies can be detected in peripheral blood and can be used as noninvasive, presymptomatic, and low-cost biomarkers. Therefore, autoantibodies can provide a possible supplementary method for breast cancer screening and prognosis classification. This article introduces the methods used to detect peripheral blood autoantibodies and the research progress in the screening and prognosis of breast cancer made in recent years to provide a potential direction for the examination and treatment of breast cancer.
Collapse
Affiliation(s)
| | | | | | - Qian Long
- Department of General Surgery, The Second Xiangya Hospital of Central South University, Changsha, China
| |
Collapse
|
18
|
Prognostic significance of NY-ESO-1 antigen and PIGR expression in esophageal tumors of CHP-NY-ESO-1-vaccinated patients as adjuvant therapy. Cancer Immunol Immunother 2022; 71:2743-2755. [DOI: 10.1007/s00262-022-03194-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 03/16/2022] [Indexed: 10/18/2022]
|
19
|
Papanikolaou NA, Hytiroglou P, Pantelidou P, Papavassiliou AG, Old LL. Systems-Level Mapping of Cancer Testis Antigen 1b/a to Sarcoma Pathways Identifies Activated Ran Binding-2 E3 SUMO-Protein Ligase and Transducin-Like Enhancer Protein 1. Front Genet 2022; 13:834445. [PMID: 35664317 PMCID: PMC9158115 DOI: 10.3389/fgene.2022.834445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 03/14/2022] [Indexed: 11/13/2022] Open
Abstract
Here we describe the identification of genes and their encoded proteins that are expressed in advanced grade tumors by reconstruction of a sarcoma cancer testis gene 1b/a (catg1b/a) network. CTAG1B/A is an ortholog of the yeast/Drosophila transcription factor Pcc1p, and a member of the KEOPS transcription complex. It has been implicated in telomere maintenance and transcriptional regulation through association with chromatin remodeling factors and is only expressed during adult testis germ cell differentiation. Ctag1b/a is re-activated in synovial sarcomas and myxoid liposarcomas but not in differentiated liposarcomas. We mapped CTAG1B/A protein to sarcoma transcription pathways with gene set expression analysis (GSEA) and using independent samples, we immunohistochemically identified expression of at least two network neighbors, RANBP2, and TLE1, thus validating our approach. This work demonstrates that mapping unknown genes to functional pathways by network re-construction is a powerful tool that can be used to identify candidate oncoproteins.
Collapse
Affiliation(s)
- Nikolaos A. Papanikolaou
- Laboratory of Biological Chemistry, Department of Medicine, Section of Biological Sciences and Preventive Medicine, School of Medicine, Aristotle University of Thessaloniki, Macedonia, Greece
- *Correspondence: Nikolaos A. Papanikolaou,
| | - Prodromos Hytiroglou
- Department of Medicine, Laboratory of Pathology, School of Medicine, Aristotle University of Thessaloniki, Macedonia, Greece
| | - Pavlina Pantelidou
- Department of Medicine, Laboratory of Pathology, School of Medicine, Aristotle University of Thessaloniki, Macedonia, Greece
| | - Athanasios G. Papavassiliou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Lloyd L. Old
- Ludwig Institute for Cancer Research, Memorial Sloan Kettering Institute for Cancer Research, New York City, NY, United States
| |
Collapse
|
20
|
Zappala F, Higbee-Dempsey E, Jang B, Miller J, Yan L, Minutolo NG, Rosado González GT, Tsourkas A, Ozdemir BA. Rapid, site-specific labeling of "off-the-shelf" and native serum autoantibodies with T cell-redirecting domains. SCIENCE ADVANCES 2022; 8:eabn4613. [PMID: 35522741 PMCID: PMC9075798 DOI: 10.1126/sciadv.abn4613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Accepted: 03/22/2022] [Indexed: 06/14/2023]
Abstract
Extensive antibody engineering and cloning is typically required to generate new bispecific antibodies. Made-to-order genes, advanced expression systems, and high-efficiency cloning can simplify and accelerate this process, but it still can take months before a functional product is realized. We developed a simple method to site-specifically and covalently attach a T cell-redirecting domain to any off-the-shelf, human immunoglobulin G (IgG) or native IgG isolated from serum. No antibody engineering, cloning, or knowledge of the antibody sequence is required. Bispecific antibodies are generated in just hours. By labeling antibodies isolated from tumor-bearing mice, including two syngeneic models, we generated T cell-redirecting autoantibodies (TRAAbs) that act as an effective therapeutic. TRAAbs preferentially bind tumor tissue over healthy tissue, indicating a previously unexplored therapeutic window. The use of autoantibodies to direct the tumor targeting of bispecific antibodies represents a new paradigm in personalized medicine that eliminates the need to identify tumor biomarkers.
Collapse
Affiliation(s)
- Fabiana Zappala
- Department of Bioengineering, University of Pennsylvania, 210 S. 33rd Street, 240 Skirkanich Hall, Philadelphia, PA 19104, USA
| | - Elizabeth Higbee-Dempsey
- Department of Bioengineering, University of Pennsylvania, 210 S. 33rd Street, 240 Skirkanich Hall, Philadelphia, PA 19104, USA
| | - Bian Jang
- Department of Bioengineering, University of Pennsylvania, 210 S. 33rd Street, 240 Skirkanich Hall, Philadelphia, PA 19104, USA
| | - Joann Miller
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd, Philadelphia, PA 19104, USA
| | - Lesan Yan
- Department of Bioengineering, University of Pennsylvania, 210 S. 33rd Street, 240 Skirkanich Hall, Philadelphia, PA 19104, USA
| | - Nicholas G. Minutolo
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd, Philadelphia, PA 19104, USA
| | - Gabriela T. Rosado González
- Gabriela T. Rosado González, Department of Chemistry, University of Puerto Rico, 14, 2534 Av. Universidad Ste. 1401, San Juan, 00925 Puerto Rico
| | - Andrew Tsourkas
- Department of Bioengineering, University of Pennsylvania, 210 S. 33rd Street, 240 Skirkanich Hall, Philadelphia, PA 19104, USA
| | - Burcin Altun Ozdemir
- Department of Bioengineering, University of Pennsylvania, 210 S. 33rd Street, 240 Skirkanich Hall, Philadelphia, PA 19104, USA
| |
Collapse
|
21
|
Abstract
The tumor microenvironment (TME) is a heterogeneous, complex organization composed of tumor, stroma, and endothelial cells that is characterized by cross talk between tumor and innate and adaptive immune cells. Over the last decade, it has become increasingly clear that the immune cells in the TME play a critical role in controlling or promoting tumor growth. The function of T lymphocytes in this process has been well characterized. On the other hand, the function of B lymphocytes is less clear, although recent data from our group and others have strongly indicated a critical role for B cells in antitumor immunity. There are, however, a multitude of populations of B cells found within the TME, ranging from naive B cells all the way to terminally differentiated plasma cells and memory B cells. Here, we characterize the role of B cells in the TME in both animal models and patients, with an emphasis on dissecting how B cell heterogeneity contributes to the immune response to cancer.
Collapse
Affiliation(s)
- Stephanie M Downs-Canner
- Department of Surgery, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA.,Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| | - Jeremy Meier
- Department of Medicine, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA;
| | - Benjamin G Vincent
- Department of Medicine, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA; .,Bioinformatics and Computational Biology Program, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA.,Department of Microbiology and Immunology, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| | - Jonathan S Serody
- Department of Medicine, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA; .,Department of Microbiology and Immunology, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| |
Collapse
|
22
|
Fridman WH, Meylan M, Petitprez F, Sun CM, Italiano A, Sautès-Fridman C. B cells and tertiary lymphoid structures as determinants of tumour immune contexture and clinical outcome. Nat Rev Clin Oncol 2022; 19:441-457. [PMID: 35365796 DOI: 10.1038/s41571-022-00619-z] [Citation(s) in RCA: 342] [Impact Index Per Article: 114.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/04/2022] [Indexed: 02/08/2023]
Abstract
B cells are a major component of the tumour microenvironment, where they are predominantly associated with tertiary lymphoid structures (TLS). In germinal centres within mature TLS, B cell clones are selectively activated and amplified, and undergo antibody class switching and somatic hypermutation. Subsequently, these B cell clones differentiate into plasma cells that can produce IgG or IgA antibodies targeting tumour-associated antigens. In tumours without mature TLS, B cells are either scarce or differentiate into regulatory cells that produce immunosuppressive cytokines. Indeed, different tumours vary considerably in their TLS and B cell content. Notably, tumours with mature TLS, a high density of B cells and plasma cells, as well as the presence of antibodies to tumour-associated antigens are typically associated with favourable clinical outcomes and responses to immunotherapy compared with those lacking these characteristics. However, polyclonal B cell activation can also result in the formation of immune complexes that trigger the production of pro-inflammatory cytokines by macrophages and neutrophils. In complement-rich tumours, IgG antibodies can also activate the complement cascade, resulting in the production of anaphylatoxins that sustain tumour-promoting inflammation and angiogenesis. Herein, we review the phenotypic heterogeneity of intratumoural B cells and the importance of TLS in their generation as well as the potential of B cells and TLS as prognostic and predictive biomarkers. We also discuss novel therapeutic approaches that are being explored with the aim of increasing mature TLS formation, B cell differentiation and anti-tumour antibody production within tumours.
Collapse
Affiliation(s)
- Wolf H Fridman
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université Paris-Cité, Equipe inflammation, complément et cancer, Paris, France. .,Equipe labellisée Ligue contre le Cancer, Paris, France.
| | - Maxime Meylan
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université Paris-Cité, Equipe inflammation, complément et cancer, Paris, France.,Equipe labellisée Ligue contre le Cancer, Paris, France
| | - Florent Petitprez
- MRC Centre for Reproductive Health, Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, UK
| | - Cheng-Ming Sun
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université Paris-Cité, Equipe inflammation, complément et cancer, Paris, France.,Equipe labellisée Ligue contre le Cancer, Paris, France
| | - Antoine Italiano
- Faculty of Medicine, University of Bordeaux, Bordeaux, France.,Department of Medicine, Institute Bergonié, Bordeaux, France
| | - Catherine Sautès-Fridman
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université Paris-Cité, Equipe inflammation, complément et cancer, Paris, France.,Equipe labellisée Ligue contre le Cancer, Paris, France
| |
Collapse
|
23
|
Liu X, Xu Y, Xiong W, Yin B, Huang Y, Chu J, Xing C, Qian C, Du Y, Duan T, Wang HY, Zhang N, Yu JS, An Z, Wang R. Development of a TCR-like antibody and chimeric antigen receptor against NY-ESO-1/HLA-A2 for cancer immunotherapy. J Immunother Cancer 2022; 10:jitc-2021-004035. [PMID: 35338087 PMCID: PMC8961179 DOI: 10.1136/jitc-2021-004035] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/27/2022] [Indexed: 01/09/2023] Open
Abstract
Background The current therapeutic antibodies and chimeric antigen receptor (CAR) T cells are capable of recognizing surface antigens, but not of intracellular proteins, thus limiting the target coverage for drug development. To mimic the feature of T-cell receptor (TCR) that recognizes the complex of major histocompatibility class I and peptide on the cell surface derived from the processed intracellular antigen, we used NY-ESO-1, a cancer-testis antigen, to develop a TCR-like fully human IgG1 antibody and its derivative, CAR-T cells, for cancer immunotherapy. Methods Human single-chain variable antibody fragment (scFv) phage library (~10∧11) was screened against HLA-A2/NY-ESO-1 (peptide 157–165) complex to obtain target-specific antibodies. The specificity and affinity of those antibodies were characterized by flow cytometry, ELISA, biolayer interferometry, and confocal imaging. The biological functions of CAR-T cells were evaluated against target tumor cells in vitro. In vivo antitumor activity was investigated in a triple-negative breast cancer (TNBC) model and primary melanoma tumor model in immunocompromised mice. Results Monoclonal antibody 2D2 identified from phage-displayed library specifically bound to NY-ESO-1157-165 in the context of human leukocyte antigen HLA-A*02:01 but not to non-A2 or NY-ESO-1 negative cells. The second-generation CAR-T cells engineered from 2D2 specifically recognized and eliminated A2+/NY-ESO-1+tumor cells in vitro, inhibited tumor growth, and prolonged the overall survival of mice in TNBC and primary melanoma tumor model in vivo. Conclusions This study showed the specificity of the antibody identified from human scFv phage library and demonstrated the potential antitumor activity by TCR-like CAR-T cells both in vitro and in vivo, warranting further preclinical and clinical evaluation of the TCR-like antibody in patients. The generation of TCR-like antibody and its CAR-T cells provides the state-of-the-art platform and proof-of-concept validation to broaden the scope of target antigen recognition and sheds light on the development of novel therapeutics for cancer immunotherapy.
Collapse
Affiliation(s)
- Xin Liu
- Keck School of Medicine, University of Southern California, Los Angeles, California, USA.,Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, Texas, USA.,Institute of Biosciences and Technology, College of Medicine, Texas A&M University, Houston, Texas, USA
| | - Yixiang Xu
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Wei Xiong
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Bingnan Yin
- Keck School of Medicine, University of Southern California, Los Angeles, California, USA.,Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, Texas, USA
| | - Yuqian Huang
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, Texas, USA.,Xiangya School of Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| | - Junjun Chu
- Keck School of Medicine, University of Southern California, Los Angeles, California, USA.,Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, Texas, USA
| | - Changsheng Xing
- Keck School of Medicine, University of Southern California, Los Angeles, California, USA.,Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, Texas, USA
| | - Chen Qian
- Keck School of Medicine, University of Southern California, Los Angeles, California, USA.,Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, Texas, USA
| | - Yang Du
- Keck School of Medicine, University of Southern California, Los Angeles, California, USA.,Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, Texas, USA
| | - Tianhao Duan
- Keck School of Medicine, University of Southern California, Los Angeles, California, USA.,Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, Texas, USA
| | - Helen Y Wang
- Keck School of Medicine, University of Southern California, Los Angeles, California, USA.,Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, Texas, USA
| | - Ningyan Zhang
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - John S Yu
- Neurosurgical Oncology in the Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Zhiqiang An
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Rongfu Wang
- Keck School of Medicine, University of Southern California, Los Angeles, California, USA .,Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, Texas, USA.,Institute of Biosciences and Technology, College of Medicine, Texas A&M University, Houston, Texas, USA
| |
Collapse
|
24
|
Zhao J, Xu Z, Liu Y, Wang X, Liu X, Gao Y, Jin Y. The expression of cancer-testis antigen in ovarian cancer and the development of immunotherapy. Am J Cancer Res 2022; 12:681-694. [PMID: 35261795 PMCID: PMC8899981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 01/26/2022] [Indexed: 06/14/2023] Open
Abstract
Ovarian cancer is a relatively common tumor in women with the highest mortality among female reproductive system tumors. The lack of apparent early symptoms and effective screening strategies often leads to ovarian cancer being diagnosed at an advanced stage. Immunotherapy relying on tumor-associated antigens might improve the treatment of ovarian cancer. Cancer-testis antigens (CTAs) are ideal tumor-associated antigens, and MAGE-A, NY-ESO-1, CT45, and Sp17 are classic CTAs highly expressed in ovarian cancer. Here, we review the research on CTAs in ovarian cancer, including prognostic value and advances in immunotherapy, all of which are essential for developing a theoretical basis for targeted therapy strategies.
Collapse
Affiliation(s)
- Jianhang Zhao
- Department of Pharmacology, School of Pharmacy, China Medical UniversityShenyang, Liaoning, China
| | - Zhaoxu Xu
- Department of Pharmacology, School of Pharmacy, China Medical UniversityShenyang, Liaoning, China
| | - Yan Liu
- Liaoning Research Institute of Family Planning (Reproductive Hospital Affiliated to China Medical University), Key Laboratory of Reproductive Health and Genetic Medicine, National Health Commission of ChinaShenyang, Liaoning, China
| | - Xiaobin Wang
- Center of Reproductive Medicine, Shengjing Hospital of China Medical UniversityShenyang, Liaoning, China
| | - Xinli Liu
- Medical Oncology Department of Gastrointestinal Cancer, Liaoning Cancer Hospital & Institute, Cancer Hospital of China Medical UniversityShenyang, Liaoning, China
| | - Yanan Gao
- Department of Pharmacology, School of Pharmacy, China Medical UniversityShenyang, Liaoning, China
| | - Ying Jin
- Liaoning Research Institute of Family Planning (Reproductive Hospital Affiliated to China Medical University), Key Laboratory of Reproductive Health and Genetic Medicine, National Health Commission of ChinaShenyang, Liaoning, China
| |
Collapse
|
25
|
Lue JK, Downs-Canner S, Chaudhuri J. The role of B cells in the development, progression, and treatment of lymphomas and solid tumors. Adv Immunol 2022; 154:71-117. [PMID: 36038195 DOI: 10.1016/bs.ai.2022.07.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
B cells are integral components of the mammalian immune response as they have the ability to generate antibodies against an almost infinite array of antigens. Over the past several decades, significant scientific progress has been made in understanding that this enormous B cell diversity contributes to pathogen clearance. However, our understanding of the humoral response to solid tumors and to tumor-specific antigens is unclear. In this review, we first discuss how B cells interact with other cells in the tumor microenvironment and influence the development and progression of various solid tumors. The ability of B lymphocytes to generate antibodies against a diverse repertoire of antigens and subsequently tailor the humoral immune response to specific pathogens relies on their ability to undergo genomic alterations during their development and differentiation. We will discuss key transforming events that lead to the development of B cell lymphomas. Overall, this review provides a foundation for innovative therapeutic interventions for both lymphoma and solid tumor malignancies.
Collapse
Affiliation(s)
- Jennifer K Lue
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, United States.
| | - Stephanie Downs-Canner
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, United States.
| | - Jayanta Chaudhuri
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, United States.
| |
Collapse
|
26
|
Immunomodulation: An immune regulatory mechanism in carcinoma therapeutics. Int Immunopharmacol 2021; 99:107984. [PMID: 34303999 DOI: 10.1016/j.intimp.2021.107984] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 06/29/2021] [Accepted: 07/11/2021] [Indexed: 01/01/2023]
Abstract
Cancer has been generally related to the possession of numerous mutations which interrupt important signaling pathways. Nevertheless, deregulated immunological signaling is considered as one of the key factors associated with the development and progression of cancer. The signaling pathways operate as modular network with different components interacting in a switch-like fashion with two proteins interplaying between each other leading to direct or indirect inhibition or stimulation of down-stream factors. Genetic, epigenetic, and transcriptomic alterations maintain the pathological conduit of different signaling pathways via affecting diverse mechanisms including cell destiny. At present, immunotherapy is one of the best therapies opted for cancer treatment. The cancer immunotherapy strategy includes harnessing the specificity and killing mechanisms of the immunological system to target and eradicate malignant cells. Targeted therapies utilizing several little molecules including Galunisertib, Astragaloside-IV, Melatonin, and Jervine capable of regulating key signaling pathways can effectively help in the management of different carcinomas.
Collapse
|
27
|
Zhang M, Hong JA, Kunst TF, Bond CD, Kenney CM, Warga CL, Yeray J, Lee MJ, Yuno A, Lee S, Miettinen M, Ripley RT, Hoang CD, Gnjatic S, Trepel JB, Schrump DS. Randomized phase II trial of a first-in-human cancer cell lysate vaccine in patients with thoracic malignancies. Transl Lung Cancer Res 2021; 10:3079-3092. [PMID: 34430349 PMCID: PMC8350099 DOI: 10.21037/tlcr-21-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2021] [Accepted: 05/21/2021] [Indexed: 01/10/2023]
Abstract
BACKGROUND Although most malignancies express cancer-testis antigens (CTA), immune responses to these proteins are limited in thoracic oncology patients. This trial was undertaken to examine if a cancer cell lysate vaccine could induce immunity to CTA, and to ascertain if metronomic cyclophosphamide and celecoxib enhances vaccine-induced immune responses. METHODS Eleven patients with primary thoracic malignancies and 10 patients with extrathoracic neoplasms metastatic to the chest rendered NED by conventional therapies were randomized to receive H1299 lung cancer cell lysates (10 mg protein/vaccine) with Iscomatrix™ adjuvant via deep intradermal injection q 4 weeks ×6 with or without daily oral metronomic cyclophosphamide/celecoxib. The primary endpoint was serologic response to purified CTA assessed 1 month after the 6th vaccination. Secondary endpoints included assessment of the effects of cyclophosphamide and celecoxib on frequency and magnitude of vaccine-induced immune responses to CTA. Exploratory endpoints included evaluation of the effects of the vaccine regimens on peripheral immune subsets. Standard of care imaging studies were obtained at baseline and 1 month after the 3rd and 6th vaccinations. RESULTS All patients exhibited local and systemic inflammatory responses lasting 72-96 hours following vaccinations. There were no dose limiting treatment related toxicities. Fourteen patients (67%) completed all six vaccinations. Eight of 14 patients (57%) exhibited serologic responses to NY-ESO-1. One patient developed antibodies to GAGE7; several patients exhibited reactivity to XAGE and MAGE-C2. Vaccine therapy decreased the percent of Tregs (P=0.0068), PD-1 expression on Tregs (P=0.0027), PD-L1 expression on CD14+ monocytes (P=0.0089), PD-L1 expression on classical monocytes (P=0.016), and PD-L1 expression on intermediate monocytes (P=0.0031). Cyclophosphamide/celecoxib did not appear to increase immune responses or enhance vaccine-induced alterations in peripheral immune subsets. CONCLUSIONS H1299 lysate vaccines with Iscomatrix™ induce immune responses to CTA and modulate peripheral immune subsets in a manner that may enhance antitumor immunity in patients with thoracic malignancies.
Collapse
Affiliation(s)
- Mary Zhang
- Thoracic Epigenetics Section, Thoracic Surgery Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Julie A. Hong
- Thoracic Epigenetics Section, Thoracic Surgery Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Tricia F. Kunst
- Thoracic Epigenetics Section, Thoracic Surgery Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Colleen D. Bond
- Thoracic Epigenetics Section, Thoracic Surgery Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Cara M. Kenney
- Thoracic Epigenetics Section, Thoracic Surgery Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Cheryl L. Warga
- Thoracic Epigenetics Section, Thoracic Surgery Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Javier Yeray
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Min-Jung Lee
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Akira Yuno
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Sunmin Lee
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Markku Miettinen
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - R. Taylor Ripley
- Thoracic Epigenetics Section, Thoracic Surgery Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Chuong D. Hoang
- Thoracic Epigenetics Section, Thoracic Surgery Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Sacha Gnjatic
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jane B. Trepel
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - David S. Schrump
- Thoracic Epigenetics Section, Thoracic Surgery Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| |
Collapse
|
28
|
Lan Y, Liu W, Zhang W, Hu J, Zhu X, Wan L, A S, Ping Y, Xiao Y. Transcriptomic heterogeneity of driver gene mutations reveals novel mutual exclusivity and improves exploration of functional associations. Cancer Med 2021; 10:4977-4993. [PMID: 34076361 PMCID: PMC8290236 DOI: 10.1002/cam4.4039] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 04/26/2021] [Accepted: 05/10/2021] [Indexed: 12/11/2022] Open
Abstract
Background Lung adenocarcinoma (LUAD), as the most common subtype of lung cancer, is the leading cause of cancer deaths in the world. The accumulation of driver gene mutations enables cancer cells to gradually acquire growth advantage. Therefore, it is important to understand the functions and interactions of driver gene mutations in cancer progression. Methods We obtained gene mutation data and gene expression profile of 506 LUAD tumors from The Cancer Genome Atlas (TCGA). The subtypes of tumors with driver gene mutations were identified by consensus cluster analysis. Results We found 21 significantly mutually exclusive pairs consisting of 20 genes among 506 LUAD patients. Because of the increased transcriptomic heterogeneity of mutations, we identified subtypes among tumors with non‐silent mutations in driver genes. There were 494 mutually exclusive pairs found among driver gene mutations within different subtypes. Furthermore, we identified functions of mutually exclusive pairs based on the hypothesis of functional redundancy of mutual exclusivity. These mutually exclusive pairs were significantly enriched in nuclear division and humoral immune response, which played crucial roles in cancer initiation and progression. We also found 79 mutually exclusive triples among subtypes of tumors with driver gene mutations, which were key roles in cell motility and cellular chemical homeostasis. In addition, two mutually exclusive triples and one mutually exclusive triple were associated with the overall survival and disease‐specific survival of LUAD patients, respectively. Conclusions We revealed novel mutual exclusivity and generated a comprehensive functional landscape of driver gene mutations, which could offer a new perspective to understand the mechanisms of cancer development and identify potential biomarkers for LUAD therapy.
Collapse
Affiliation(s)
- Yujia Lan
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Wei Liu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Wanmei Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Jing Hu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Xiaojing Zhu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Linyun Wan
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Suru A
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Yanyan Ping
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Yun Xiao
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| |
Collapse
|
29
|
Li J, Qin B, Huang M, Ma Y, Li D, Li W, Guo Z. Tumor-Associated Antigens (TAAs) for the Serological Diagnosis of Osteosarcoma. Front Immunol 2021; 12:665106. [PMID: 33995397 PMCID: PMC8119874 DOI: 10.3389/fimmu.2021.665106] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Accepted: 04/08/2021] [Indexed: 01/07/2023] Open
Abstract
Osteosarcoma (OS) is the most common form of malignant bone tumor found in childhood and adolescence. Although its incidence rate is low among cancers, the prognosis of OS is usually poor. Although some biomarkers, such as p53, have been identified in OS, the association between the biomarkers and clinical outcome is not well understood. Thus, it is necessary to establish a method to identify patients diagnosed with OS at an early stage. It is becoming obvious that anti-tumor-associated antigens (TAAs) autoantibodies (TAAbs) in sera could be used as serological biomarkers in the detection of many different types of cancers. This notion indicates that TAAbs are considered as immunological “sentinels” associated with tumorigenesis underlying molecular events. It provides new insights into the molecular and cellular biology of the differential diagnosis of cancers. What’s more, it is reported that a customized TAA array could significantly increase the sensitivity/specificity. TAA arrays also have great application prospects in detecting cancer at an early stage, monitoring cancer progression, discovering new therapeutic targets, and designing personalized treatment. In this review, we provide an overview of the TAAs identified in OS as well as the possibility that TAAs and TAAbs system be used as biomarkers in the immunodiagnosis and prognosis of OS.
Collapse
Affiliation(s)
- Jitian Li
- Henan Luoyang Orthopedic Hospital (Henan Provincial Orthopedic Hospital)/Henan Institute of Orthopedic and Traumatology, Luoyang, China
| | - Bo Qin
- Transitional Medical Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Manyu Huang
- Henan Luoyang Orthopedic Hospital (Henan Provincial Orthopedic Hospital)/Henan Institute of Orthopedic and Traumatology, Luoyang, China
| | - Yan Ma
- Henan Luoyang Orthopedic Hospital (Henan Provincial Orthopedic Hospital)/Henan Institute of Orthopedic and Traumatology, Luoyang, China
| | - Dongsheng Li
- Henan Luoyang Orthopedic Hospital (Henan Provincial Orthopedic Hospital)/Henan Institute of Orthopedic and Traumatology, Luoyang, China
| | - Wuyin Li
- Henan Luoyang Orthopedic Hospital (Henan Provincial Orthopedic Hospital)/Henan Institute of Orthopedic and Traumatology, Luoyang, China
| | - Zhiping Guo
- Henan Luoyang Orthopedic Hospital (Henan Provincial Orthopedic Hospital)/Henan Institute of Orthopedic and Traumatology, Luoyang, China
| |
Collapse
|
30
|
Clemente O, Ottaiano A, Di Lorenzo G, Bracigliano A, Lamia S, Cannella L, Pizzolorusso A, Di Marzo M, Santorsola M, De Chiara A, Fazioli F, Tafuto S. Is immunotherapy in the future of therapeutic management of sarcomas? J Transl Med 2021; 19:173. [PMID: 33902630 PMCID: PMC8077947 DOI: 10.1186/s12967-021-02829-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 04/12/2021] [Indexed: 12/19/2022] Open
Abstract
Sarcomas are rare, ubiquitous and heterogeneous tumors usually treated with surgery, chemotherapy, target therapy, and radiotherapy. However, 25-50% of patients experience local relapses and/or distant metastases after chemotherapy with an overall survival about 12-18 months. Recently, immuno-therapy has revolutionized the cancer treatments with initial indications for non-small cell lung cancer (NSCLC) and melanoma (immune-checkpoint inhibitors).Here, we provide a narrative review on the topic as well as a critical description of the currently available trials on immunotherapy treatments in patients with sarcoma. Given the promising results obtained with anti-PD-1 monoclonal antibodies (pembrolizumab and nivolumab) and CAR-T cells, we strongly believe that these new immunotherapeutic approaches, along with an innovative characterization of tumor genetics, will provide an exciting opportunity to ameliorate the therapeutic management of sarcomas.
Collapse
Affiliation(s)
- Ottavia Clemente
- Sarcomas and Rare Tumors Unit, Istituto Nazionale Tumori, IRCCS - Fondazione "G. Pascale", 80131, Naples, Italy
| | - Alessandro Ottaiano
- Division of Innovative Therapies, Istituto Nazionale Tumori, IRCCS - Fondazione "G. Pascale", 80131, Naples, Italy
| | - Giuseppe Di Lorenzo
- Sarcomas and Rare Tumors Unit, Istituto Nazionale Tumori, IRCCS - Fondazione "G. Pascale", 80131, Naples, Italy
| | - Alessandra Bracigliano
- Nuclear Medicine Unit, Istituto Nazionale Tumori, IRCCS - Fondazione "G. Pascale, 80131, Naples, Italy
| | - Sabrina Lamia
- Sarcomas and Rare Tumors Unit, Istituto Nazionale Tumori, IRCCS - Fondazione "G. Pascale", 80131, Naples, Italy
| | - Lucia Cannella
- Sarcomas and Rare Tumors Unit, Istituto Nazionale Tumori, IRCCS - Fondazione "G. Pascale", 80131, Naples, Italy
| | - Antonio Pizzolorusso
- Sarcomas and Rare Tumors Unit, Istituto Nazionale Tumori, IRCCS - Fondazione "G. Pascale", 80131, Naples, Italy
| | - Massimiliano Di Marzo
- Department of Abdominal Oncology, Istituto Nazionale Tumori, IRCCS - Fondazione "G. Pascale", 80131, Naples, Italy
| | - Mariachiara Santorsola
- Division of Innovative Therapies, Istituto Nazionale Tumori, IRCCS - Fondazione "G. Pascale", 80131, Naples, Italy
| | - Annarosaria De Chiara
- Histopathology of Lymphomas and Sarcomas SSD, Istituto Nazionale Tumori, IRCCS - Fondazione "G. Pascale", 80131, Naples, Italy
| | - Flavio Fazioli
- Orthopedic Oncology Unit, Istituto Nazionale Tumori, IRCCS - Fondazione "G. Pascale", 80131, Naples, Italy
| | - Salvatore Tafuto
- Sarcomas and Rare Tumors Unit, Istituto Nazionale Tumori, IRCCS - Fondazione "G. Pascale", 80131, Naples, Italy.
| |
Collapse
|
31
|
Da Gama Duarte J, Woods K, Quigley LT, Deceneux C, Tutuka C, Witkowski T, Ostrouska S, Hudson C, Tsao SCH, Pasam A, Dobrovic A, Blackburn JM, Cebon J, Behren A. Ropporin-1 and 1B Are Widely Expressed in Human Melanoma and Evoke Strong Humoral Immune Responses. Cancers (Basel) 2021; 13:1805. [PMID: 33918976 PMCID: PMC8069442 DOI: 10.3390/cancers13081805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 04/06/2021] [Accepted: 04/07/2021] [Indexed: 11/17/2022] Open
Abstract
Antibodies that block immune regulatory checkpoints (programmed cell death 1, PD-1 and cytotoxic T-lymphocyte-associated antigen 4, CTLA-4) to mobilise immunity have shown unprecedented clinical efficacy against cancer, demonstrating the importance of antigen-specific tumour recognition. Despite this, many patients still fail to benefit from these treatments and additional approaches are being sought. These include mechanisms that boost antigen-specific immunity either by vaccination or adoptive transfer of effector cells. Other than neoantigens, epigenetically regulated and shared antigens such as NY-ESO-1 are attractive targets; however, tissue expression is often heterogeneous and weak. Therefore, peptide-specific therapies combining multiple antigens rationally selected to give additive anti-cancer benefits are necessary to achieve optimal outcomes. Here, we show that Ropporin-1 (ROPN1) and 1B (ROPN1B), cancer restricted antigens, are highly expressed and immunogenic, inducing humoral immunity in patients with advanced metastatic melanoma. By multispectral immunohistochemistry, 88.5% of melanoma patients tested (n = 54/61) showed ROPN1B expression in at least 1 of 2/3 tumour cores in tissue microarrays. Antibody responses against ROPN1A and ROPN1B were detected in 71.2% of melanoma patients tested (n = 74/104), with increased reactivity seen with more advanced disease stages. Thus, ROPN1A and ROPN1B may indeed be viable targets for cancer immunotherapy, alone or in combination with other cancer antigens, and could be combined with additional therapies such as immune checkpoint blockade.
Collapse
Affiliation(s)
- Jessica Da Gama Duarte
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC 3084, Australia; (J.D.G.D.); (K.W.); (L.T.Q.); (C.D.); (C.T.); (T.W.); (S.O.); (C.H.); (S.C.-H.T.); (A.P.); (A.D.); (J.C.)
- School of Cancer Medicine, La Trobe University, Bundoora, VIC 3086, Australia
| | - Katherine Woods
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC 3084, Australia; (J.D.G.D.); (K.W.); (L.T.Q.); (C.D.); (C.T.); (T.W.); (S.O.); (C.H.); (S.C.-H.T.); (A.P.); (A.D.); (J.C.)
- School of Cancer Medicine, La Trobe University, Bundoora, VIC 3086, Australia
| | - Luke T. Quigley
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC 3084, Australia; (J.D.G.D.); (K.W.); (L.T.Q.); (C.D.); (C.T.); (T.W.); (S.O.); (C.H.); (S.C.-H.T.); (A.P.); (A.D.); (J.C.)
- School of Cancer Medicine, La Trobe University, Bundoora, VIC 3086, Australia
| | - Cyril Deceneux
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC 3084, Australia; (J.D.G.D.); (K.W.); (L.T.Q.); (C.D.); (C.T.); (T.W.); (S.O.); (C.H.); (S.C.-H.T.); (A.P.); (A.D.); (J.C.)
- School of Cancer Medicine, La Trobe University, Bundoora, VIC 3086, Australia
| | - Candani Tutuka
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC 3084, Australia; (J.D.G.D.); (K.W.); (L.T.Q.); (C.D.); (C.T.); (T.W.); (S.O.); (C.H.); (S.C.-H.T.); (A.P.); (A.D.); (J.C.)
- School of Cancer Medicine, La Trobe University, Bundoora, VIC 3086, Australia
| | - Tom Witkowski
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC 3084, Australia; (J.D.G.D.); (K.W.); (L.T.Q.); (C.D.); (C.T.); (T.W.); (S.O.); (C.H.); (S.C.-H.T.); (A.P.); (A.D.); (J.C.)
- School of Cancer Medicine, La Trobe University, Bundoora, VIC 3086, Australia
| | - Simone Ostrouska
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC 3084, Australia; (J.D.G.D.); (K.W.); (L.T.Q.); (C.D.); (C.T.); (T.W.); (S.O.); (C.H.); (S.C.-H.T.); (A.P.); (A.D.); (J.C.)
- School of Cancer Medicine, La Trobe University, Bundoora, VIC 3086, Australia
| | - Chris Hudson
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC 3084, Australia; (J.D.G.D.); (K.W.); (L.T.Q.); (C.D.); (C.T.); (T.W.); (S.O.); (C.H.); (S.C.-H.T.); (A.P.); (A.D.); (J.C.)
- School of Cancer Medicine, La Trobe University, Bundoora, VIC 3086, Australia
| | - Simon Chang-Hao Tsao
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC 3084, Australia; (J.D.G.D.); (K.W.); (L.T.Q.); (C.D.); (C.T.); (T.W.); (S.O.); (C.H.); (S.C.-H.T.); (A.P.); (A.D.); (J.C.)
- School of Cancer Medicine, La Trobe University, Bundoora, VIC 3086, Australia
| | - Anupama Pasam
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC 3084, Australia; (J.D.G.D.); (K.W.); (L.T.Q.); (C.D.); (C.T.); (T.W.); (S.O.); (C.H.); (S.C.-H.T.); (A.P.); (A.D.); (J.C.)
- School of Cancer Medicine, La Trobe University, Bundoora, VIC 3086, Australia
| | - Alexander Dobrovic
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC 3084, Australia; (J.D.G.D.); (K.W.); (L.T.Q.); (C.D.); (C.T.); (T.W.); (S.O.); (C.H.); (S.C.-H.T.); (A.P.); (A.D.); (J.C.)
- School of Cancer Medicine, La Trobe University, Bundoora, VIC 3086, Australia
- Department of Clinical Pathology, Melbourne Medical School, University of Melbourne, Parkville, VIC 3010, Australia
| | - Jonathan M. Blackburn
- Department of Integrative Biomedical Sciences, Faculty of Health Sciences, University of Cape Town, Cape Town 7925, South Africa;
- Institute for Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town 7925, South Africa
| | - Jonathan Cebon
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC 3084, Australia; (J.D.G.D.); (K.W.); (L.T.Q.); (C.D.); (C.T.); (T.W.); (S.O.); (C.H.); (S.C.-H.T.); (A.P.); (A.D.); (J.C.)
- School of Cancer Medicine, La Trobe University, Bundoora, VIC 3086, Australia
- Medical Oncology Unit, Austin Health, Heidelberg, VIC 3084, Australia
| | - Andreas Behren
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC 3084, Australia; (J.D.G.D.); (K.W.); (L.T.Q.); (C.D.); (C.T.); (T.W.); (S.O.); (C.H.); (S.C.-H.T.); (A.P.); (A.D.); (J.C.)
- School of Cancer Medicine, La Trobe University, Bundoora, VIC 3086, Australia
- Department of Medicine—Austin, Melbourne Medical School, University of Melbourne, Parkville, VIC 3010, Australia
| |
Collapse
|
32
|
Ward MP, E Kane L, A Norris L, Mohamed BM, Kelly T, Bates M, Clarke A, Brady N, Martin CM, Brooks RD, Brooks DA, Selemidis S, Hanniffy S, Dixon EP, A O'Toole S, J O'Leary J. Platelets, immune cells and the coagulation cascade; friend or foe of the circulating tumour cell? Mol Cancer 2021; 20:59. [PMID: 33789677 PMCID: PMC8011144 DOI: 10.1186/s12943-021-01347-1] [Citation(s) in RCA: 91] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 03/15/2021] [Indexed: 02/07/2023] Open
Abstract
Cancer cells that transit from primary tumours into the circulatory system are known as circulating tumour cells (CTCs). These cancer cells have unique phenotypic and genotypic characteristics which allow them to survive within the circulation, subsequently extravasate and metastasise. CTCs have emerged as a useful diagnostic tool using "liquid biopsies" to report on the metastatic potential of cancers. However, CTCs by their nature interact with components of the blood circulatory system on a constant basis, influencing both their physical and morphological characteristics as well as metastatic capabilities. These properties and the associated molecular profile may provide critical diagnostic and prognostic capabilities in the clinic. Platelets interact with CTCs within minutes of their dissemination and are crucial in the formation of the initial metastatic niche. Platelets and coagulation proteins also alter the fate of a CTC by influencing EMT, promoting pro-survival signalling and aiding in evading immune cell destruction. CTCs have the capacity to directly hijack immune cells and utilise them to aid in CTC metastatic seeding processes. The disruption of CTC clusters may also offer a strategy for the treatment of advance staged cancers. Therapeutic disruption of these heterotypical interactions as well as direct CTC targeting hold great promise, especially with the advent of new immunotherapies and personalised medicines. Understanding the molecular role that platelets, immune cells and the coagulation cascade play in CTC biology will allow us to identify and characterise the most clinically relevant CTCs from patients. This will subsequently advance the clinical utility of CTCs in cancer diagnosis/prognosis.
Collapse
Affiliation(s)
- Mark P Ward
- Department of Histopathology and Morbid Anatomy, Trinity College Dublin, Dublin 8, Ireland.
- Emer Casey Molecular Pathology Research Laboratory, Coombe Women and Infants University Hospital, Dublin 8, Ireland.
- Trinity St. James's Cancer Institute, St James's Hospital, Dublin 8, Ireland.
| | - Laura E Kane
- Department of Histopathology and Morbid Anatomy, Trinity College Dublin, Dublin 8, Ireland
- Emer Casey Molecular Pathology Research Laboratory, Coombe Women and Infants University Hospital, Dublin 8, Ireland
- Trinity St. James's Cancer Institute, St James's Hospital, Dublin 8, Ireland
| | - Lucy A Norris
- Trinity St. James's Cancer Institute, St James's Hospital, Dublin 8, Ireland
- Department of Obstetrics and Gynaecology, Trinity College Dublin, Dublin 8, Ireland
| | - Bashir M Mohamed
- Department of Histopathology and Morbid Anatomy, Trinity College Dublin, Dublin 8, Ireland
- Emer Casey Molecular Pathology Research Laboratory, Coombe Women and Infants University Hospital, Dublin 8, Ireland
- Trinity St. James's Cancer Institute, St James's Hospital, Dublin 8, Ireland
| | - Tanya Kelly
- Department of Histopathology and Morbid Anatomy, Trinity College Dublin, Dublin 8, Ireland
- Emer Casey Molecular Pathology Research Laboratory, Coombe Women and Infants University Hospital, Dublin 8, Ireland
- Trinity St. James's Cancer Institute, St James's Hospital, Dublin 8, Ireland
| | - Mark Bates
- Department of Histopathology and Morbid Anatomy, Trinity College Dublin, Dublin 8, Ireland
- Emer Casey Molecular Pathology Research Laboratory, Coombe Women and Infants University Hospital, Dublin 8, Ireland
- Trinity St. James's Cancer Institute, St James's Hospital, Dublin 8, Ireland
| | - Andres Clarke
- Department of Histopathology and Morbid Anatomy, Trinity College Dublin, Dublin 8, Ireland
- Emer Casey Molecular Pathology Research Laboratory, Coombe Women and Infants University Hospital, Dublin 8, Ireland
- Trinity St. James's Cancer Institute, St James's Hospital, Dublin 8, Ireland
| | - Nathan Brady
- Department of Histopathology and Morbid Anatomy, Trinity College Dublin, Dublin 8, Ireland
- Emer Casey Molecular Pathology Research Laboratory, Coombe Women and Infants University Hospital, Dublin 8, Ireland
- Trinity St. James's Cancer Institute, St James's Hospital, Dublin 8, Ireland
| | - Cara M Martin
- Department of Histopathology and Morbid Anatomy, Trinity College Dublin, Dublin 8, Ireland
- Emer Casey Molecular Pathology Research Laboratory, Coombe Women and Infants University Hospital, Dublin 8, Ireland
- Trinity St. James's Cancer Institute, St James's Hospital, Dublin 8, Ireland
| | - Robert D Brooks
- Cancer Research Institute, University of South Australia, 5001, Adelaide, Australia
| | - Doug A Brooks
- Cancer Research Institute, University of South Australia, 5001, Adelaide, Australia
| | - Stavros Selemidis
- School of Health and Biomedical Sciences, RMIT University, Victoria, 3083, Bundoora, Australia
| | | | - Eric P Dixon
- BD Technologies and Innovation, Research Triangle Park, NC, USA
| | - Sharon A O'Toole
- Department of Histopathology and Morbid Anatomy, Trinity College Dublin, Dublin 8, Ireland
- Emer Casey Molecular Pathology Research Laboratory, Coombe Women and Infants University Hospital, Dublin 8, Ireland
- Trinity St. James's Cancer Institute, St James's Hospital, Dublin 8, Ireland
- Department of Obstetrics and Gynaecology, Trinity College Dublin, Dublin 8, Ireland
| | - John J O'Leary
- Department of Histopathology and Morbid Anatomy, Trinity College Dublin, Dublin 8, Ireland
- Emer Casey Molecular Pathology Research Laboratory, Coombe Women and Infants University Hospital, Dublin 8, Ireland
- Trinity St. James's Cancer Institute, St James's Hospital, Dublin 8, Ireland
| |
Collapse
|
33
|
Slingluff CL, Zarour HM, Tawbi HAH, Kirkwood JM, Postow MA, Friedlander P, Devoe CE, Gaughan EM, Mauldin IS, Olson WC, Smith KT, Macri MJ, Ricciardi T, Ryan A, Venhaus R, Wolchok JD. A phase 1 study of NY-ESO-1 vaccine + anti-CTLA4 antibody Ipilimumab (IPI) in patients with unresectable or metastatic melanoma. Oncoimmunology 2021; 10:1898105. [PMID: 33796406 PMCID: PMC8007150 DOI: 10.1080/2162402x.2021.1898105] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Ipilimumab (IPI) can enhance immunity to the cancer-testis antigen NY-ESO-1. A clinical trial was designed to assess safety, immunogenicity, and clinical responses with IPI + NY-ESO-1 vaccines and effects on the tumor microenvironment (TME). Patients with measurable NY-ESO-1+ tumors were enrolled among three arms: A) IPI + NY-ESO-1 protein + poly-ICLC (pICLC) + incomplete Freund’s adjuvant (IFA); B) IPI + NY-ESO-1 overlapping long peptides (OLP) + pICLC + IFA; and C) IPI + NY-ESO-1 OLP + pICLC. Clinical responses were assessed by irRC. T cell and Ab responses were assessed by ex vivo IFN-gamma ELIspot and ELISA. Tumor biopsies pre- and post-treatment were evaluated for immune infiltrates. Eight patients were enrolled: 5, 2, and 1 in Arms A-C, respectively. There were no DLTs. Best clinical responses were SD (4) and PD (4). T-cell and antibody (Ab) responses to NY-ESO-1 were detected in 6 (75%) and 7 (88%) patients, respectively, and were associated with SD. The breadth of Ab responses was greater for patients with SD than PD (p = .036). For five patients evaluable in the TME, treatment was associated with increases in proliferating (Ki67+) CD8+ T cells and decreases in RORγt+ CD4+ T cells. T cell densities increased for those with SD. Detection of T cell responses to NY-ESO-1 ex vivo in most patients suggests that IPI may have enhanced those responses. Proliferating intratumoral CD8+ T cells increased after vaccination plus IPI suggesting favorable impact of IPI plus NY-ESO-1 vaccines on the TME. List of Abbreviations: Ab = antibody; CTCAE = NCI Common Terminology Criteria for Adverse Events; DHFR/DHRP = dihydrofolate reductase; DLT = Dose-limiting toxicity; ELISA = enzyme-linked immunosorbent assay; IFA = incomplete Freund’s adjuvant (Montanide ISA-51); IFNγ = Interferon gamma; IPI = Ipilimumab; irRC = immune-related response criteria; mIFH = multispectral immunofluorescence histology; OLP = NY-ESO-1 overlapping long peptides; PBMC = peripheral blood mononuclear cells; PD = Progressive disease; pICLC = poly-ICLC (Hiltonol), a TLR3/MDA-5 agonist; RLT = Regimen-limiting Toxicity; ROI = regions of interest; RT = room temperature; SAE = serious adverse event; SD = stable disease; TEAE = treatment-emergent adverse events; TLR = toll-like receptor; TME = tumor microenvironment; TRAE = treatment-related adverse events.
Collapse
Affiliation(s)
- Craig L Slingluff
- Department of Surgery/Division of Surgical Oncology, University of Virginia, Charlottesville, VA, USA
| | - Hassane M Zarour
- Division of Medical Oncology, Dept of Medicine and Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, USA
| | - Hussein Abdul-Hassan Tawbi
- Division of Medical Oncology, Dept of Medicine and Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, USA.,Department of Melanoma Medical Oncology, MD Anderson Cancer Center, Houston, TX
| | - John M Kirkwood
- Division of Medical Oncology, Dept of Medicine and Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, USA
| | - Michael A Postow
- Department of Medicine, Memorial Sloan-Kettering Cancer Center, Weill Cornell Medical College, New York, NY, USA
| | - Philip Friedlander
- Department of Medicine, Hematology, and Medical Oncology, Mount Sinai Medical Center, New York, NY, USA
| | - Craig E Devoe
- Northwell Health Cancer Institute, Lake Success, NY, USA
| | - Elizabeth M Gaughan
- Department of Medicine/Division of Hematology Oncology, University of Virginia, Charlottesville, VA, USA
| | - Ileana S Mauldin
- Department of Surgery/Division of Surgical Oncology, University of Virginia, Charlottesville, VA, USA
| | - Walter C Olson
- Department of Surgery/Division of Surgical Oncology, University of Virginia, Charlottesville, VA, USA
| | - Kelly T Smith
- Department of Surgery/Division of Surgical Oncology, University of Virginia, Charlottesville, VA, USA
| | - Mary J Macri
- Ludwig Institute for Cancer Research, New York, NY, USA
| | | | - Aileen Ryan
- Ludwig Institute for Cancer Research, New York, NY, USA
| | - Ralph Venhaus
- Ludwig Institute for Cancer Research, New York, NY, USA
| | - Jedd D Wolchok
- Department of Medicine, Memorial Sloan-Kettering Cancer Center, Weill Cornell Medical College, New York, NY, USA.,Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center.,Parker Institute for Cancer Immunotherapy, San Francisco, California, USA
| |
Collapse
|
34
|
Safety and antibody immune response of CHP-NY-ESO-1 vaccine combined with poly-ICLC in advanced or recurrent esophageal cancer patients. Cancer Immunol Immunother 2021; 70:3081-3091. [PMID: 33751208 DOI: 10.1007/s00262-021-02892-w] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 02/13/2021] [Indexed: 10/21/2022]
Abstract
The nanoparticle complex of cholesteryl pullulan (CHP) and NY-ESO-1 antigen protein (CHP-NY-ESO-1) presents multiple epitope peptides to MHC class I and II pathways, leading to CD8+ and CD4+ T cell responses. Poly-ICLC is a synthetic, double-stranded RNA, an agonist of toll-like receptor (TLR)-3, and a cytoplasmic receptor of melanoma differentiation-associated gene (MDA)-5. It should be a suitable immune adjuvant of cancer vaccine to overcome the inhibitory tumor microenvironment. We conducted a phase 1 clinical trial of CHP-NY-ESO-1 with poly-ICLC in patients with advanced or recurrent esophageal cancer. CHP-NY-ESO-1/poly-ICLC (μg/mg) was administered at a dose of 200/0.5 or 200/1.0 (cohorts 1 and 2, respectively) every 2 weeks for a total of six doses. The primary endpoints were safety and immune response. The secondary endpoint was tumor response. In total, 16 patients were enrolled, and six patients in each cohort completed the trial. The most common adverse event (AE) was injection site skin reaction (86.7%). No grade 3 or higher drug-related AEs were observed. No tumor responses were observed, and three patients (30%) had stable disease. The immune response was comparable between the two cohorts, and all patients (100%) achieved antibody responses with a median of 2.5 vaccinations. Comparing CHP-NY-ESO-1 alone to the poly-ICLC combination, all patients in both groups exhibited antibody responses, but the titers were higher in the combination group. In a mouse model, adding anti-PD-1 antibody to the combination of CHP-NY-ESO-1/poly-ICLC suppressed the growth of NY-ESO-1-expressing tumors. Combining the vaccine with PD-1 blockade holds promise in human trials.
Collapse
|
35
|
Abstract
The opposing roles of innate and adaptive immune cells in suppressing or supporting cancer initiation, progression, metastasis and response to therapy has been long debated. The mechanisms by which different monocyte and T cell subtypes affect and modulate cancer have been extensively studied. However, the role of B cells and their subtypes have remained elusive, perhaps partially due to their heterogeneity and range of actions. B cells can produce a variety of cytokines and present tumor-derived antigens to T cells in combination with co-stimulatory or inhibitory ligands based on their phenotype. Unlike most T cells, B cells can be activated by innate immune stimuli, such as endotoxin. Furthermore, the isotype and specificity of the antibodies produced by plasma cells regulate distinct immune responses, including opsonization, antibody-mediated cellular cytotoxicity (ADCC) and complement activation. B cells are shaped by the tumor environment (TME), with the capability to regulate the TME in return. In this review, we will describe the mechanisms of B cell action, including cytokine production, antigen presentation, ADCC, opsonization, complement activation and how they affect tumor development and response to immunotherapy. We will also discuss how B cell fate within the TME is affected by tumor stroma, microbiome and metabolism.
Collapse
Affiliation(s)
- Shabnam Shalapour
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA.
| | - Michael Karin
- Department of Pharmacology, School of Medicine, University of California San Diego, CA 92093, USA; Laboratory of Gene Regulation and Signal Transduction, Department of Pharmacology, School of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| |
Collapse
|
36
|
Kandalaft LE, Odunsi K, Coukos G. Immune Therapy Opportunities in Ovarian Cancer. Am Soc Clin Oncol Educ Book 2021; 40:1-13. [PMID: 32412818 DOI: 10.1200/edbk_280539] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Immunotherapy has emerged as a highly promising approach in the treatment of epithelial ovarian cancer (EOC). Immune checkpoint blockade (ICB) therapy, PARP inhibitors (PARPis), neoantigen vaccines, and personalized T-cell therapy have been associated with encouraging clinical activity in a small subset of patients. To increase the proportion of patients who are likely to derive benefit, it will be important not only to generate sufficient numbers of antitumor T cells but also to overcome multiple inhibitory networks in the ovarian tumor microenvironment (TME). Therefore, a major direction is to develop biomarkers that would predict responsiveness to different types of immunotherapies and allow treatment selection based on the results. Moreover, such biomarkers would allow rational combination of immunotherapies while minimizing toxicities. In this review, we provide progress on immune therapies and future directions for maximally exploiting immune-based strategies for the treatment of ovarian cancer.
Collapse
Affiliation(s)
- Lana E Kandalaft
- Ludwig Institute for Cancer Research, University of Lausanne, and Department of Oncology, Lausanne University Hospital, Lausanne, Switzerland
| | - Kunle Odunsi
- Center for Immunotherapy, Roswell Park Comprehensive Cancer Center, Buffalo, NY.,Department of Gynecologic Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY
| | - George Coukos
- Ludwig Institute for Cancer Research, University of Lausanne, and Department of Oncology, Lausanne University Hospital, Lausanne, Switzerland
| |
Collapse
|
37
|
Feng Y, Xia W, He G, Ke R, Liu L, Xie M, Tang A, Yi X. Accuracy Evaluation and Comparison of 14 Diagnostic Markers for Nasopharyngeal Carcinoma: A Meta-Analysis. Front Oncol 2020; 10:1779. [PMID: 33072558 PMCID: PMC7531263 DOI: 10.3389/fonc.2020.01779] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 08/11/2020] [Indexed: 12/14/2022] Open
Abstract
The aim of the present study was to collect published studies and compare the diagnostic accuracy of different markers for nasopharyngeal carcinoma (NPC). We systematically searched PubMed/MEDLINE, EMBASE, Cochrane Library, CNKI, and Wanfang for relevant studies until April 29, 2020. The revised Quality Assessment of Diagnostic Accuracy Studies (QUADAS-2) tool was used to evaluate the methodological quality of the studies. The sensitivity, specificity, positive likelihood ratio (PLR), negative likelihood ratio (NLR), diagnostic odds ratio (DOR), and area under the curve (AUC) values of the diagnostic markers were combined by a bivariate mixed effect model to compare their diagnostic accuracy. We explored heterogeneity through meta-regression. In total, 244 records from 101 articles were included, with 49,432 total study subjects (13,109 cases and 36,323 controls). EA-IgG, Zta-IgG, and Epstein-Barr virus (EBV) DNA load in non-invasive nasopharyngeal brushings (EBV-DNA brushings) have both high sensitivity and specificity, EBNA1-IgG and VCA-IgG have only high sensitivity, and EBNA1-IgA, VCA-IgA, Rta-IgG, Zta-IgA, HSP70, and serum sialic acid (SA) have only high specificity. The bivariate mixed effect model of EA-IgA had a significant threshold effect. Meta-regression analysis showed that ethnicity affected EBNA1-IgA, EBNA1-IgG, VCA-IgA, and EBV DNA load in plasma, test methods affected EBNA1-IgG, publication year affected VCA-IgA, and sample size affected Rta-IgG. There was significant publication bias for VCA-IgA and Rta-IgG (P < 0.05). EA-IgG, Zta-IgG, and EBV-DNA brushings are good diagnostic markers for NPC. The diagnostic accuracy was influenced by publication year, sample size, test methods, and ethnicity.
Collapse
Affiliation(s)
- Yiwei Feng
- Department of Otolaryngology-Head & Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, China
- First Clinical Medical College, Guangxi Medical University, Nanning, China
| | - Wei Xia
- Department of Otolaryngology-Head & Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, China
- First Clinical Medical College, Guangxi Medical University, Nanning, China
| | - Guangyao He
- Department of Otolaryngology-Head & Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Rongdan Ke
- Department of Otolaryngology-Head & Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Lei Liu
- Department of Otolaryngology-Head & Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Mao Xie
- Department of Otolaryngology-Head & Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Anzhou Tang
- Department of Otolaryngology-Head & Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Xiang Yi
- Department of Otolaryngology-Head & Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Nanning, China
| |
Collapse
|
38
|
Music M, Iafolla M, Soosaipillai A, Batruch I, Prassas I, Pintilie M, Hansen AR, Bedard PL, Lheureux S, Spreafico A, Razak AA, Siu LL, Diamandis EP. Predicting response and toxicity to PD-1 inhibition using serum autoantibodies identified from immuno-mass spectrometry. F1000Res 2020; 9:337. [PMID: 33299547 PMCID: PMC7707117 DOI: 10.12688/f1000research.22715.1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/22/2020] [Indexed: 12/15/2022] Open
Abstract
Background: Validated biomarkers are needed to identify patients at increased risk of immune-related adverse events (irAEs) to immune checkpoint blockade (ICB). Antibodies directed against endogenous antigens can change after exposure to ICB. Methods: Patients with different solid tumors stratified into cohorts received pembrolizumab every 3 weeks in a Phase II trial (INSPIRE study). Blood samples were collected prior to first pembrolizumab exposure (baseline) and approximately 7 weeks (pre-cycle 3) into treatment. In a discovery analysis, autoantibody target immuno-mass spectrometry was performed in baseline and pre-cycle 3 pooled sera of 24 INSPIRE patients based on clinical benefit (CBR) and irAEs. Results: Thyroglobulin (Tg) and thyroid peroxidase (TPO) were identified as the candidate autoantibody targets. In the overall cohort of 78 patients, the frequency of CBR and irAEs from pembrolizumab was 31% and 24%, respectively. Patients with an anti-Tg titer increase ≥1.5x from baseline to pre-cycle 3 were more likely to have irAEs relative to patients without this increase in unadjusted, cohort adjusted, and multivariable models (OR=17.4, 95% CI 1.8-173.8, p=0.015). Similarly, patients with an anti-TPO titer ≥ 1.5x from baseline to pre-cycle 3 were more likely to have irAEs relative to patients without the increase in unadjusted and cohort adjusted (OR=6.1, 95% CI 1.1-32.7, p=0.035) models. Further, the cohort adjusted analysis showed patients with anti-Tg titer greater than median (10.0 IU/mL) at pre-cycle 3 were more likely to have irAEs (OR=4.7, 95% CI 1.2-17.8, p=0.024). Patients with pre-cycle 3 anti-TPO titers greater than median (10.0 IU/mL) had a significant difference in overall survival (23.8 vs 11.5 months; HR=1.8, 95% CI 1.0-3.2, p=0.05). Conclusions: Patient increase ≥1.5x of anti-Tg and anti-TPO titers from baseline to pre-cycle 3 were associated with irAEs from pembrolizumab, and patients with elevated pre-cycle 3 anti-TPO titers had an improvement in overall survival.
Collapse
Affiliation(s)
- Milena Music
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Marco Iafolla
- Division of Medical Oncology and Hematology, University Health Network, Canada, Toronto, ON, Canada
| | - Antoninus Soosaipillai
- Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, Toronto, ON, Canada
| | - Ihor Batruch
- Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, Toronto, ON, Canada
| | - Ioannis Prassas
- Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, Toronto, ON, Canada
| | - Melania Pintilie
- Department of Biostatistics, Princess Margaret Cancer Centre, University Health Network, Canada, Toronto, ON, Canada
| | - Aaron R. Hansen
- Division of Medical Oncology and Hematology, University Health Network, Canada, Toronto, ON, Canada
| | - Philippe L. Bedard
- Division of Medical Oncology and Hematology, University Health Network, Canada, Toronto, ON, Canada
| | - Stephanie Lheureux
- Division of Medical Oncology and Hematology, University Health Network, Canada, Toronto, ON, Canada
| | - Anna Spreafico
- Division of Medical Oncology and Hematology, University Health Network, Canada, Toronto, ON, Canada
| | - Albiruni Abdul Razak
- Division of Medical Oncology and Hematology, University Health Network, Canada, Toronto, ON, Canada
| | - Lillian L. Siu
- Division of Medical Oncology and Hematology, University Health Network, Canada, Toronto, ON, Canada
| | - Eleftherios P. Diamandis
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, Toronto, ON, Canada
- Department of Clinical Biochemistry, University Health Network, Canada, Toronto, ON, Canada
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Toronto, ON, Canada
| |
Collapse
|
39
|
Cebon JS, Gore M, Thompson JF, Davis ID, McArthur GA, Walpole E, Smithers M, Cerundolo V, Dunbar PR, MacGregor D, Fisher C, Millward M, Nathan P, Findlay MPN, Hersey P, Evans TRJ, Ottensmeier CH, Marsden J, Dalgleish AG, Corrie PG, Maria M, Brimble M, Williams G, Winkler S, Jackson HM, Endo-Munoz L, Tutuka CSA, Venhaus R, Old LJ, Haack D, Maraskovsky E, Behren A, Chen W. Results of a randomized, double-blind phase II clinical trial of NY-ESO-1 vaccine with ISCOMATRIX adjuvant versus ISCOMATRIX alone in participants with high-risk resected melanoma. J Immunother Cancer 2020; 8:e000410. [PMID: 32317292 PMCID: PMC7204806 DOI: 10.1136/jitc-2019-000410] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/03/2020] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND To compare the clinical efficacy of New York Esophageal squamous cell carcinoma-1 (NY-ESO-1) vaccine with ISCOMATRIX adjuvant versus ISCOMATRIX alone in a randomized, double-blind phase II study in participants with fully resected melanoma at high risk of recurrence. METHODS Participants with resected stage IIc, IIIb, IIIc and IV melanoma expressing NY-ESO-1 were randomized to treatment with three doses of NY-ESO-1/ISCOMATRIX or ISCOMATRIX adjuvant administered intramuscularly at 4-week intervals, followed by a further dose at 6 months. Primary endpoint was the proportion free of relapse at 18 months in the intention-to-treat (ITT) population and two per-protocol populations. Secondary endpoints included relapse-free survival (RFS) and overall survival (OS), safety and NY-ESO-1 immunity. RESULTS The ITT population comprised 110 participants, with 56 randomized to NY-ESO-1/ISCOMATRIX and 54 to ISCOMATRIX alone. No significant toxicities were observed. There were no differences between the study arms in relapses at 18 months or for median time to relapse; 139 vs 176 days (p=0.296), or relapse rate, 27 (48.2%) vs 26 (48.1%) (HR 0.913; 95% CI 0.402 to 2.231), respectively. RFS and OS were similar between the study arms. Vaccine recipients developed strong positive antibody responses to NY-ESO-1 (p≤0.0001) and NY-ESO-1-specific CD4+ and CD8+ responses. Biopsies following relapse did not demonstrate differences in NY-ESO-1 expression between the study populations although an exploratory study demonstrated reduced (NY-ESO-1)+/Human Leukocyte Antigen (HLA) class I+ double-positive cells in biopsies from vaccine recipients performed on relapse in 19 participants. CONCLUSIONS The vaccine was well tolerated, however, despite inducing antigen-specific immunity, it did not affect survival endpoints. Immune escape through the downregulation of NY-ESO-1 and/or HLA class I molecules on tumor may have contributed to relapse.
Collapse
MESH Headings
- Adjuvants, Immunologic/administration & dosage
- Adjuvants, Immunologic/adverse effects
- Antigens, Neoplasm/genetics
- Antigens, Neoplasm/immunology
- Biopsy
- Cancer Vaccines/administration & dosage
- Cancer Vaccines/adverse effects
- Cancer Vaccines/genetics
- Cancer Vaccines/immunology
- Chemotherapy, Adjuvant/adverse effects
- Chemotherapy, Adjuvant/methods
- Cholesterol/administration & dosage
- Cholesterol/adverse effects
- Dermatologic Surgical Procedures
- Disease-Free Survival
- Double-Blind Method
- Drug Combinations
- Female
- Follow-Up Studies
- Humans
- Immunogenicity, Vaccine
- Male
- Melanoma/diagnosis
- Melanoma/immunology
- Melanoma/mortality
- Melanoma/therapy
- Membrane Proteins/genetics
- Membrane Proteins/immunology
- Middle Aged
- Neoplasm Recurrence, Local/diagnosis
- Neoplasm Recurrence, Local/epidemiology
- Neoplasm Recurrence, Local/prevention & control
- Neoplasm Staging
- Phospholipids/administration & dosage
- Phospholipids/adverse effects
- Saponins/administration & dosage
- Saponins/adverse effects
- Skin/pathology
- Skin Neoplasms/diagnosis
- Skin Neoplasms/immunology
- Skin Neoplasms/mortality
- Skin Neoplasms/therapy
Collapse
Affiliation(s)
- Jonathan S Cebon
- Cancer Immunobiology Programme, Olivia Newton-John Cancer Research Institute, School of Cancer Medicine, La Trobe University at Austin Health, Heidelberg, Victoria, Australia
- Ludwig Institute for Cancer Research Austin Branch, Heidelberg, Victoria, Australia
| | - Martin Gore
- Oncology, Royal Marsden Hospital NHS Trust, London, UK
| | - John F Thompson
- Melanoma Institute Australia, North Sydney, New South Wales, Australia
| | - Ian D Davis
- Ludwig Institute for Cancer Research Austin Branch, Heidelberg, Victoria, Australia
- Monash University Eastern Health Clinical School, Box Hill, Victoria, Australia
| | - Grant A McArthur
- Melanona and Skin Service, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Euan Walpole
- Cancer Services Division, Princess Alexandra Hospital Health Service District, Woolloongabba, Queensland, Australia
| | - Mark Smithers
- Oncology Services Unit, Princess Alexandra Hospital Health Service District, Woolloongabba, Queensland, Australia
| | - Vincenzo Cerundolo
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, Oxford, Oxfordshire, UK
| | - P Rod Dunbar
- School of Biological Sciences and Maurice Wilkins Centre, The University of Auckland, Auckland, New Zealand
| | - Duncan MacGregor
- Department of Anatomical Pathology, Austin Health, Heidelberg, Victoria, Australia
| | - Cyril Fisher
- Oncology, Royal Marsden Hospital NHS Trust, London, UK
| | - Michael Millward
- School of Medicine and Pharmacology, Sir Charles Gairdner Hospital, Nedlands, Western Australia, Australia
| | - Paul Nathan
- Mount Vernon Cancer Centre, Mount Vernon Hospital, Northwood, London, UK
| | - Michael P N Findlay
- School of Medicine and Health Science, The University of Auckland, Auckland, New Zealand
| | - Peter Hersey
- Melanoma Immunology and Oncology Group, Centenary Institute, Newtown, New South Wales, Australia
| | - T R Jeffry Evans
- Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| | | | - Jeremy Marsden
- University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Angus G Dalgleish
- Cell and Molecular Sciences, Division of Oncology, St Georges Hospital Medical School, London, UK
| | - Pippa G Corrie
- West Anglia Cancer Research Network Oncology Centre, Addenbrooke's Hospital, Cambridge, Cambridgeshire, UK
| | - Marples Maria
- The Cancer Research Centre, Weston Park Hospital, Sheffield, UK
| | - Margaret Brimble
- School of Biological Sciences and Maurice Wilkins Centre, The University of Auckland, Auckland, New Zealand
| | - Geoff Williams
- School of Biological Sciences and Maurice Wilkins Centre, The University of Auckland, Auckland, New Zealand
| | - Sintia Winkler
- School of Biological Sciences and Maurice Wilkins Centre, The University of Auckland, Auckland, New Zealand
| | - Heather M Jackson
- Ludwig Institute for Cancer Research Austin Branch, Heidelberg, Victoria, Australia
| | - Liliana Endo-Munoz
- Cancer Immunobiology Programme, Olivia Newton-John Cancer Research Institute, School of Cancer Medicine, La Trobe University at Austin Health, Heidelberg, Victoria, Australia
| | - Candani S A Tutuka
- Cancer Immunobiology Programme, Olivia Newton-John Cancer Research Institute, School of Cancer Medicine, La Trobe University at Austin Health, Heidelberg, Victoria, Australia
- Ludwig Institute for Cancer Research Austin Branch, Heidelberg, Victoria, Australia
| | - Ralph Venhaus
- Ludwig Institute for Cancer Research, New York, New York, USA
| | - Lloyd J Old
- Ludwig Institute for Cancer Research, New York, New York, USA
| | - Dennis Haack
- Versagenics Inc, Morrisville, North Carolina, USA
| | | | - Andreas Behren
- Cancer Immunobiology Programme, Olivia Newton-John Cancer Research Institute, School of Cancer Medicine, La Trobe University at Austin Health, Heidelberg, Victoria, Australia
- Ludwig Institute for Cancer Research Austin Branch, Heidelberg, Victoria, Australia
| | - Weisan Chen
- Ludwig Institute for Cancer Research Austin Branch, Heidelberg, Victoria, Australia
- Biochemistry and Genetics, La Trobe University, Melbourne, Victoria, Australia
| |
Collapse
|
40
|
Hou Z, Liang X, Wang X, Zhou Z, Shi G. Myeloid-derived suppressor cells infiltration in non-small-cell lung cancer tumor and MAGE-A4 and NY-ESO-1 expression. Oncol Lett 2020; 19:3982-3992. [PMID: 32382343 PMCID: PMC7202317 DOI: 10.3892/ol.2020.11497] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 01/14/2020] [Indexed: 12/22/2022] Open
Abstract
Cancer/testis antigens melanoma-associated antigen 4 (MAGE-A4) and New York esophageal squamous cell carcinoma-1 (NY-ESO-1) are of clinical interest as biomarkers and present valuable targets for immunotherapy; however, they are poor prognostic markers in non-small cell lung cancer (NSCLC). In addition, myeloid derived suppressor cells (MDSCs) are recognized as a key element in tumor escape and progression. The aim of the present study was to investigate the diagnostic and prognostic value of MAGE-A4 and NY-ESO-1, and their association with MDSCs in NSCLC samples. The expression levels of MAGE-A4 and NY-ESO-1, and the infiltration of MDSCs (CD33+), were analyzed by immunohistochemistry of 67 tissue samples from patients with NSCLC. Overall, 58.33% of the NSCLC squamous cell carcinoma tissues and 94.7% of adenocarcinoma tissues were positive for MAGE-A4. NY-ESO-1 expression was observed in 52.78% of the squamous cell carcinoma tissues and 80% of the adenocarcinoma tissues. In primary adenocarcinoma tumor tissues, MAGE-A4 and NY-ESO-1 demonstrated a higher intensity of expression compared with the squamous cell carcinoma tissues. A total of 33 (91.7%) squamous cell carcinoma and 19 (95.0%) adenocarcinoma specimens were positive for CD33. The expression of MAGE-A4 and NY-ESO-1 antigens and infiltration of MDSCs was associated with poor prognosis of patients with NSCLC. Further studies investigating the association between these findings and underlying molecular mechanisms are required.
Collapse
Affiliation(s)
- Zhenbo Hou
- Department of Pathology, Zibo Central Hospital, Zibo, Shandong 255000, P.R. China
| | - Xiao Liang
- Department of Thoracic Surgery, Zibo Central Hospital, Zibo, Shandong 255000, P.R. China
| | - Xinmei Wang
- Department of Pathology, Zibo Central Hospital, Zibo, Shandong 255000, P.R. China
| | - Ziqiang Zhou
- Department of Pathology, Zibo Central Hospital, Zibo, Shandong 255000, P.R. China
| | - Guilan Shi
- Department of Immunology, School of Nursing, Zibo Vocational Institute, Zibo, Shandong 255314, P.R. China.,Frank Reidy Research Center for Bioelectrics, Old Dominion University, Norfolk, VA 23508, USA
| |
Collapse
|
41
|
Pedersen JW, Nøstdal A, Wandall HH. Multiplexed Detection of Autoantibodies to Glycopeptides Using Microarray. Methods Mol Biol 2020; 2024:199-211. [PMID: 31364051 DOI: 10.1007/978-1-4939-9597-4_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2023]
Abstract
Protein microarray is a highly sensitive tool for antibody detection in serum. Monitoring of patients' antibody titers to specific antigens is increasingly employed in the diagnosis of several conditions, ranging from infectious diseases, allergies, autoimmune diseases, and cancer. In this protocol, we present a detailed method for enzymatic generation of disease-specific O-glycopeptides and how to monitor the antibody response to these in serum using microarray technology.
Collapse
Affiliation(s)
- Johannes W Pedersen
- Department of Cellular and Molecular Medicine, Copenhagen Center for Glycomics, University of Copenhagen, Copenhagen, Denmark
| | - Alexander Nøstdal
- Department of Cellular and Molecular Medicine, Copenhagen Center for Glycomics, University of Copenhagen, Copenhagen, Denmark
| | - Hans H Wandall
- Department of Cellular and Molecular Medicine, Copenhagen Center for Glycomics, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
42
|
Lynam S, Lugade AA, Odunsi K. Immunotherapy for Gynecologic Cancer: Current Applications and Future Directions. Clin Obstet Gynecol 2020; 63:48-63. [PMID: 31833846 PMCID: PMC7298668 DOI: 10.1097/grf.0000000000000513] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The role of the immune system in the development of cancer has been a subject of ongoing clinical investigation in recent years. Emerging data demonstrate that tumorigenesis resulting in ovarian, uterine, and cervical cancers is a consequence of impaired host immune responses to cancerous cells. Leveraging the immune system through the use of immune checkpoint inhibitors, therapeutic vaccine therapy, and adoptive cell transfer presents a profound opportunity to revolutionize cancer treatment. This review will encompass the role of the immune system in development of gynecologic cancers and highlight recent data regarding immunotherapy applications in ovarian, uterine, and cervical cancers.
Collapse
Affiliation(s)
| | - Amit A Lugade
- Center for Immunotherapy Roswell Park Cancer Institute, Buffalo, New York
| | - Kunle Odunsi
- Department of Gynecologic Oncology
- Center for Immunotherapy Roswell Park Cancer Institute, Buffalo, New York
| |
Collapse
|
43
|
Ren Y, Lv Q, Yue W, Liu B, Zou Z. The programmed cell death protein-1/programmed cell death ligand 1 expression, CD3+ T cell infiltration, NY-ESO-1 expression, and microsatellite instability phenotype in primary cutaneous melanoma and mucosal melanoma and their clinical significance and prognostic value: a study of 89 consecutive cases. Melanoma Res 2020; 30:85-101. [PMID: 31095042 DOI: 10.1097/cmr.0000000000000620] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
We evaluated the expression of programmed cell death protein-1 (PD-1), programmed cell death ligand 1 (PD-L1), and NY-ESO-1 antigen; the infiltration of CD3+ T cells; and the microsatellite instability (MSI) phenotype, as well as the relationship of each factor to survival in malignant melanoma patients. Malignant melanoma samples from 89 patients were stained by immunohistochemistry to evaluate PD-1, PD-L1, CD3+ tumor-infiltrating lymphocytes (TILs), NY-ESO-1, and MSI. PD-1 and PD-L1 were expressed in 19.1 and 32.6% of the 89 samples, respectively. There was a significant correlation between PD-1 and PD-L1 expression (r = 0.207, P = 0.046). High infiltration of CD3+ T cells was observed in 41.6% of the samples, and increased cell infiltration was associated with increased PD-1 expression (P = 0.001). NY-ESO-1 antigen was detected in 13.5% of all samples, and the expression of NY-ESO-1 was positively correlated with the expression of PD-1 (P < 0.001). In our research, MSI was detected in 18 samples (20.2%). Survival analysis showed that a high infiltration of CD3+ T cells was related to longer progression-free survival (PFS) [24.0 months, 95% confidence interval (CI): 7.4-40.6 vs. 11.0 months, 95% CI: 7.1-12.9, P = 0.031], similarly, the median overall survival (OS) of the CD3+ T cell high-infiltration patients was also longer (53.0 vs. 38.0 months), but with no statistical significance (P = 0.200). The results for the immune markers mentioned above provide a theoretical basis for the prognosis and immunotherapy selection of malignant melanoma patients.
Collapse
Affiliation(s)
- Yu Ren
- Department of the Comprehensive Cancer Center, Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School
| | - Qing Lv
- Yi Xing Tumor Hospital, Yixing, China
| | - Wuheng Yue
- Department of the Comprehensive Cancer Center, Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School
- The Comprehensive Cancer Center, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing
| | - Baorui Liu
- Department of the Comprehensive Cancer Center, Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School
| | - Zhengyun Zou
- Department of the Comprehensive Cancer Center, Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School
| |
Collapse
|
44
|
Sharonov GV, Serebrovskaya EO, Yuzhakova DV, Britanova OV, Chudakov DM. B cells, plasma cells and antibody repertoires in the tumour microenvironment. Nat Rev Immunol 2020; 20:294-307. [DOI: 10.1038/s41577-019-0257-x] [Citation(s) in RCA: 416] [Impact Index Per Article: 83.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/27/2019] [Indexed: 02/07/2023]
|
45
|
Wei R, Dean DC, Thanindratarn P, Hornicek FJ, Guo W, Duan Z. Cancer testis antigens in sarcoma: Expression, function and immunotherapeutic application. Cancer Lett 2019; 479:54-60. [PMID: 31634526 DOI: 10.1016/j.canlet.2019.10.024] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 09/23/2019] [Accepted: 10/15/2019] [Indexed: 02/06/2023]
Abstract
Sarcomas are a group of heterogeneous malignancies of mesenchymal origin. Patient outcomes remain especially grim for those with recurrent or metastatic disease, and current therapeutic strategies have not significantly improved outcomes over the past few decades. This has led to a number of studies assessing novel therapies. Cancer testis antigens (CTAs) are tumor-associated antigens with physiologic expression in the testis and various malignancies, including sarcomas. Genes encoding CTAs include MAGE, NY-ESO-1, PRAME, TRAG-3/CSAGE, and SSX. The importance and function of CTAs in tumorigenesis have gained recognition in recent years. They are also proving as robust diagnostic and prognostic biomarkers. Therapeutically, antigens derived from CTAs are highly recognizable by T lymphocytes and therefore capable of generating a potent antitumor immune response. CTAs are, therefore, promising targets for novel immunotherapies. Here we review the emerging works on expression, function, and immunotherapeutic application of CTAs in sarcoma therapy.
Collapse
Affiliation(s)
- Ran Wei
- Department of Orthopedic Surgery, Sarcoma Biology Laboratory, David Geffen School of Medicine at UCLA, 615 Charles E. Young Dr. South, Los Angeles, CA, 90095, USA; Musculoskeletal Tumor Center, Peking University People's Hospital, No. 11 Xizhimen South Street, Xicheng District, Beijing, 100044, China.
| | - Dylan C Dean
- Department of Orthopedic Surgery, Sarcoma Biology Laboratory, David Geffen School of Medicine at UCLA, 615 Charles E. Young Dr. South, Los Angeles, CA, 90095, USA.
| | - Pichaya Thanindratarn
- Department of Orthopedic Surgery, Sarcoma Biology Laboratory, David Geffen School of Medicine at UCLA, 615 Charles E. Young Dr. South, Los Angeles, CA, 90095, USA.
| | - Francis J Hornicek
- Department of Orthopedic Surgery, Sarcoma Biology Laboratory, David Geffen School of Medicine at UCLA, 615 Charles E. Young Dr. South, Los Angeles, CA, 90095, USA.
| | - Wei Guo
- Musculoskeletal Tumor Center, Peking University People's Hospital, No. 11 Xizhimen South Street, Xicheng District, Beijing, 100044, China.
| | - Zhenfeng Duan
- Department of Orthopedic Surgery, Sarcoma Biology Laboratory, David Geffen School of Medicine at UCLA, 615 Charles E. Young Dr. South, Los Angeles, CA, 90095, USA.
| |
Collapse
|
46
|
Abstract
New therapies that promote antitumour immunity have been recently developed. Most of these immunomodulatory approaches have focused on enhancing T-cell responses, either by targeting inhibitory pathways with immune checkpoint inhibitors, or by targeting activating pathways, as with chimeric antigen receptor T cells or bispecific antibodies. Although these therapies have led to unprecedented successes, only a minority of patients with cancer benefit from these treatments, highlighting the need to identify new cells and molecules that could be exploited in the next generation of immunotherapy. Given the crucial role of innate immune responses in immunity, harnessing these responses opens up new possibilities for long-lasting, multilayered tumour control.
Collapse
|
47
|
Gangkofner DS, Holzinger D, Schroeder L, Eichmüller SB, Zörnig I, Jäger D, Wichmann G, Dietz A, Broglie MA, Herold-Mende C, Dyckhoff G, Boscolo-Rizzo P, Ezic J, Marienfeld RB, Möller P, Völkel G, Kraus JM, Kestler HA, Brunner C, Schuler PJ, Wigand M, Theodoraki MN, Doescher J, Hoffmann TK, Pawlita M, Butt J, Waterboer T, Laban S. Patterns of antibody responses to nonviral cancer antigens in head and neck squamous cell carcinoma patients differ by human papillomavirus status. Int J Cancer 2019; 145:3436-3444. [PMID: 31407331 DOI: 10.1002/ijc.32623] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 07/20/2019] [Accepted: 07/22/2019] [Indexed: 12/24/2022]
Abstract
There have been hints that nonviral cancer antigens are differentially expressed in human papillomavirus (HPV)-positive and HPV-negative head and neck squamous cell carcinoma (HNSCC). Antibody responses (AR) to cancer antigens may be used to indirectly determine cancer antigen expression in the tumor using a noninvasive and tissue-saving liquid biopsy. Here, we set out to characterize AR to a panel of nonviral cancer antigens in HPV-positive and HPV-negative HNSCC patients. A fluorescent microbead multiplex serology to 29 cancer antigens (16 cancer-testis antigens, 5 cancer-retina antigens and 8 oncogenes) and 29 HPV-antigens was performed in 382 HNSCC patients from five independent cohorts (153 HPV-positive and 209 HPV-negative). AR to any of the cancer antigens were found in 272/382 patients (72%). The ten most frequent AR were CT47, cTAGE5a, c-myc, LAGE-1, MAGE-A1, -A3, -A4, NY-ESO-1, SpanX-a1 and p53. AR to MAGE-A3, MAGE-A9 and p53 were found at significantly different prevalences by HPV status. An analysis of AR mean fluorescent intensity values uncovered remarkably different AR clusters by HPV status. To identify optimal antigen selections covering a maximum of patients with ≤10 AR, multiobjective optimization revealed distinct antigen selections by HPV status. We identified that AR to nonviral antigens differ by HPV status indicating differential antigen expression. Multiplex serology may be used to characterize antigen expression using serum or plasma as a tissue-sparing liquid biopsy. Cancer antigen panels should address the distinct antigen repertoire of HPV-positive and HPV-negative HNSCC.
Collapse
Affiliation(s)
- Dominik S Gangkofner
- Department of Otorhinolaryngology and Head & Neck Surgery, University Medical Center Ulm, Head & Neck Cancer Center of the Comprehensive Cancer Center Ulm, Ulm, Germany
| | - Dana Holzinger
- Infections and Cancer Epidemiology (F022), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Lea Schroeder
- Infections and Cancer Epidemiology (F022), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Stefan B Eichmüller
- Research Group GMP & T Cell Therapy (D210), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Inka Zörnig
- National Center for Tumor Diseases (NCT) and Heidelberg University Hospital, Heidelberg, Germany.,National Center for Tumor Diseases (NCT) and German Cancer Research Center (DKFZ), Applied Tumor Immunity (D120), Heidelberg, Germany
| | - Dirk Jäger
- National Center for Tumor Diseases (NCT) and Heidelberg University Hospital, Heidelberg, Germany.,National Center for Tumor Diseases (NCT) and German Cancer Research Center (DKFZ), Applied Tumor Immunity (D120), Heidelberg, Germany
| | - Gunnar Wichmann
- Department of Otorhinolaryngology, University Hospital Leipzig, Leipzig, Germany
| | - Andreas Dietz
- Department of Otorhinolaryngology, University Hospital Leipzig, Leipzig, Germany
| | - Martina A Broglie
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Zurich, Zurich, Switzerland
| | - Christel Herold-Mende
- Department of Otorhinolaryngology, Head and Neck Surgery, Heidelberg University Hospital, Heidelberg, Germany.,Department of Neurosurgery, Division of Experimental Neurosurgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Gerhard Dyckhoff
- Department of Otorhinolaryngology, Head and Neck Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Paolo Boscolo-Rizzo
- Department of Neurosciences, ENT Clinic and Regional Center for Head and Neck Cancer, University of Padua, Treviso, Italy
| | - Jasmin Ezic
- Department of Otorhinolaryngology and Head & Neck Surgery, University Medical Center Ulm, Head & Neck Cancer Center of the Comprehensive Cancer Center Ulm, Ulm, Germany
| | | | - Peter Möller
- Institute of Pathology, University Medical Center Ulm, Ulm, Germany
| | - Gunnar Völkel
- Institute of Medical Systems Biology, Ulm University, Ulm, Germany
| | - Johann M Kraus
- Institute of Medical Systems Biology, Ulm University, Ulm, Germany
| | - Hans A Kestler
- Institute of Medical Systems Biology, Ulm University, Ulm, Germany
| | - Cornelia Brunner
- Department of Otorhinolaryngology and Head & Neck Surgery, University Medical Center Ulm, Head & Neck Cancer Center of the Comprehensive Cancer Center Ulm, Ulm, Germany
| | - Patrick J Schuler
- Department of Otorhinolaryngology and Head & Neck Surgery, University Medical Center Ulm, Head & Neck Cancer Center of the Comprehensive Cancer Center Ulm, Ulm, Germany
| | - Marlene Wigand
- Department of Otorhinolaryngology and Head & Neck Surgery, University Medical Center Ulm, Head & Neck Cancer Center of the Comprehensive Cancer Center Ulm, Ulm, Germany
| | - Marie N Theodoraki
- Department of Otorhinolaryngology and Head & Neck Surgery, University Medical Center Ulm, Head & Neck Cancer Center of the Comprehensive Cancer Center Ulm, Ulm, Germany
| | - Johannes Doescher
- Department of Otorhinolaryngology and Head & Neck Surgery, University Medical Center Ulm, Head & Neck Cancer Center of the Comprehensive Cancer Center Ulm, Ulm, Germany
| | - Thomas K Hoffmann
- Department of Otorhinolaryngology and Head & Neck Surgery, University Medical Center Ulm, Head & Neck Cancer Center of the Comprehensive Cancer Center Ulm, Ulm, Germany
| | - Michael Pawlita
- Infections and Cancer Epidemiology (F022), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Julia Butt
- Infections and Cancer Epidemiology (F022), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Tim Waterboer
- Infections and Cancer Epidemiology (F022), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Simon Laban
- Department of Otorhinolaryngology and Head & Neck Surgery, University Medical Center Ulm, Head & Neck Cancer Center of the Comprehensive Cancer Center Ulm, Ulm, Germany
| |
Collapse
|
48
|
Antibody Responses to Cancer Antigens Identify Patients with a Poor Prognosis among HPV-Positive and HPV-Negative Head and Neck Squamous Cell Carcinoma Patients. Clin Cancer Res 2019; 25:7405-7412. [DOI: 10.1158/1078-0432.ccr-19-1490] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 06/27/2019] [Accepted: 08/20/2019] [Indexed: 12/24/2022]
|
49
|
Kandalaft LE, Odunsi K, Coukos G. Immunotherapy in Ovarian Cancer: Are We There Yet? J Clin Oncol 2019; 37:2460-2471. [PMID: 31403857 DOI: 10.1200/jco.19.00508] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Affiliation(s)
- Lana E Kandalaft
- Ludwig Institute for Cancer Research and University of Lausanne, Lausanne, Switzerland
| | - Kunle Odunsi
- Roswell Park Comprehensive Cancer Center, Buffalo, NY
| | - George Coukos
- Ludwig Institute for Cancer Research and University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
50
|
MAGE-A4 and MAGE-A1 Immunohistochemical Expression in High-grade Endometrial Cancer. Int J Gynecol Pathol 2019; 38:59-65. [PMID: 29140883 DOI: 10.1097/pgp.0000000000000470] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The aim was to investigate MAGE-A4 and MAGE-A1 protein expression in high-grade endometrial cancer and determine its correlation with histologic subtype, FIGO stage, presence of vascular invasion, disease free, and overall survival. Immunohistochemical staining was performed by using 77B (MAGE-A1) and 57B (MAGE-A4) monoclonal antibodies on paraffin-embedded sections from high-grade endometrial cancers diagnosed in University Hospital Split between 1998 and 2011 (n=77). Median follow-up time for survivors was 48 mo. MAGE-A4 was found to be expressed in 33% of endometrioid type endometrial cancers grade 3 and in 27% of serous and clear cell carcinomas. MAGE-A1 was found to be expressed in 93% endometrioid endometrial cancer grade 3 and 86% of serous and clear cell carcinomas. Univariate analysis showed that positive immunohistochemical staining for MAGE-A4 was associated with decreased disease free and overall survival in patients with high-grade endometrial cancer. Multivariate analysis showed an association between MAGE-A4 overexpression and decreased disease free but not overall survival in high-grade endometrial cancer. No correlation was found between MAGE-A1 immunohistochemical expression and patient survival. There was no significant correlation between MAGE-A4 and MAGE-A1 expression and histologic subtype, FIGO stage, lymph node metastasis, muscular infiltration, and lymphovascular invasion. MAGE-A4 immunohistochemical expression is associated with decreased disease free and overall survival in patients with high-grade endometrial cancer. Our findings suggest that MAGE-A1 may be expressed in the epithelial cells of the normal endometrium. MAGE-A1 is highly expressed in high-grade endometrial cancer, with no impact on survival.
Collapse
|