1
|
Ding YD, Shu LZ, He RS, Chen KY, Deng YJ, Zhou ZB, Xiong Y, Deng H. Listeria monocytogenes: a promising vector for tumor immunotherapy. Front Immunol 2023; 14:1278011. [PMID: 37868979 PMCID: PMC10587691 DOI: 10.3389/fimmu.2023.1278011] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 09/25/2023] [Indexed: 10/24/2023] Open
Abstract
Cancer receives enduring international attention due to its extremely high morbidity and mortality. Immunotherapy, which is generally expected to overcome the limits of traditional treatments, serves as a promising direction for patients with recurrent or metastatic malignancies. Bacteria-based vectors such as Listeria monocytogenes take advantage of their unique characteristics, including preferential infection of host antigen presenting cells, intracellular growth within immune cells, and intercellular dissemination, to further improve the efficacy and minimize off-target effects of tailed immune treatments. Listeria monocytogenes can reshape the tumor microenvironment to bolster the anti-tumor effects both through the enhancement of T cells activity and a decrease in the frequency and population of immunosuppressive cells. Modified Listeria monocytogenes has been employed as a tool to elicit immune responses against different tumor cells. Currently, Listeria monocytogenes vaccine alone is insufficient to treat all patients effectively, which can be addressed if combined with other treatments, such as immune checkpoint inhibitors, reactivated adoptive cell therapy, and radiotherapy. This review summarizes the recent advances in the molecular mechanisms underlying the involvement of Listeria monocytogenes vaccine in anti-tumor immunity, and discusses the most concerned issues for future research.
Collapse
Affiliation(s)
- Yi-Dan Ding
- Medical College, Nanchang University, Nanchang, China
| | - Lin-Zhen Shu
- Medical College, Nanchang University, Nanchang, China
| | - Rui-Shan He
- Medical College, Nanchang University, Nanchang, China
| | - Kai-Yun Chen
- Office of Clinical Trials Administration, The Fourth Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yan-Juan Deng
- Department of Pathology, The Fourth Affiliated Hospital of Nanchang University, Nanchang, China
- Tumor Immunology Institute, Nanchang University, Nanchang, China
| | - Zhi-Bin Zhou
- Department of Pathology, The Fourth Affiliated Hospital of Nanchang University, Nanchang, China
- Tumor Immunology Institute, Nanchang University, Nanchang, China
| | - Ying Xiong
- Department of General Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Huan Deng
- Department of Pathology, The Fourth Affiliated Hospital of Nanchang University, Nanchang, China
- Tumor Immunology Institute, Nanchang University, Nanchang, China
| |
Collapse
|
2
|
Kang S, Wang L, Xu L, Wang R, Kang Q, Gao X, Yu L. Decitabine enhances targeting of AML cells by NY-ESO-1-specific TCR-T cells and promotes the maintenance of effector function and the memory phenotype. Oncogene 2022; 41:4696-4708. [PMID: 36097193 PMCID: PMC9568428 DOI: 10.1038/s41388-022-02455-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 08/19/2022] [Accepted: 08/25/2022] [Indexed: 12/03/2022]
Abstract
NY-ESO-1 is a well-known cancer-testis antigen (CTA) with re-expression in numerous cancer types, but its expression is suppressed in myeloid leukemia cells. Patients with acute myeloid leukemia (AML) receiving decitabine (DAC) exhibit induced expression of NY-ESO-1 in blasts; thus, we investigated the effects of NY-ESO-1-specific TCR-engineered T (TCR-T) cells combined with DAC against AML. NY-ESO-1-specific TCR-T cells could efficiently eliminate AML cell lines (including U937, HL60, and Kasumi-1cells) and primary AML blasts in vitro by targeting the DAC-induced NY-ESO-1 expression. Moreover, the incubation of T cells with DAC during TCR transduction (designated as dTCR-T cells) could further enhance the anti-leukemia efficacy of TCR-T cells and increase the generation of memory-like phenotype. The combination of DAC with NY-ESO-1-specific dTCR-T cells showed a superior anti-tumor efficacy in vivo and prolonged the survival of an AML xenograft mouse model, with three out of five mice showing complete elimination of AML cells over 90 days. This outcome was correlated with enhanced expressions of IFN-γ and TNF-α, and an increased proportion of central memory T cells (CD45RO+CD62L+ and CD45RO+CCR7+). Taken together, these data provide preclinical evidence for the combined use of DAC and NY-ESO-1-specific dTCR-T cells for the treatment of AML.
Collapse
Affiliation(s)
- Synat Kang
- Department of Hematology and Oncology, International Cancer Center, Shenzhen Key Laboratory of Precision Medicine for Hematological Malignancies, Shenzhen University General Hospital, Shenzhen University Clinical Medical Academy, Shenzhen University Health Science Center, Shenzhen, 518000, Guangdong, China
| | - Lixin Wang
- Department of Hematology and Oncology, International Cancer Center, Shenzhen Key Laboratory of Precision Medicine for Hematological Malignancies, Shenzhen University General Hospital, Shenzhen University Clinical Medical Academy, Shenzhen University Health Science Center, Shenzhen, 518000, Guangdong, China
| | - Lu Xu
- Department of Hematology and Oncology, International Cancer Center, Shenzhen Key Laboratory of Precision Medicine for Hematological Malignancies, Shenzhen University General Hospital, Shenzhen University Clinical Medical Academy, Shenzhen University Health Science Center, Shenzhen, 518000, Guangdong, China
| | - Ruiqi Wang
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Qingzheng Kang
- Department of Hematology and Oncology, International Cancer Center, Shenzhen Key Laboratory of Precision Medicine for Hematological Malignancies, Shenzhen University General Hospital, Shenzhen University Clinical Medical Academy, Shenzhen University Health Science Center, Shenzhen, 518000, Guangdong, China
| | - Xuefeng Gao
- Department of Hematology and Oncology, International Cancer Center, Shenzhen Key Laboratory of Precision Medicine for Hematological Malignancies, Shenzhen University General Hospital, Shenzhen University Clinical Medical Academy, Shenzhen University Health Science Center, Shenzhen, 518000, Guangdong, China. .,Central Laboratory, Shenzhen Key Laboratory of Precision Medicine for Hematological Malignancies, Shenzhen University General Hospital, Shenzhen, 518000, Guangdong, China.
| | - Li Yu
- Department of Hematology and Oncology, International Cancer Center, Shenzhen Key Laboratory of Precision Medicine for Hematological Malignancies, Shenzhen University General Hospital, Shenzhen University Clinical Medical Academy, Shenzhen University Health Science Center, Shenzhen, 518000, Guangdong, China.
| |
Collapse
|
3
|
Safety and preliminary immunogenicity of JNJ-64041809, a live-attenuated, double-deleted Listeria monocytogenes-based immunotherapy, in metastatic castration-resistant prostate cancer. Prostate Cancer Prostatic Dis 2021; 25:219-228. [PMID: 34257408 PMCID: PMC9184270 DOI: 10.1038/s41391-021-00402-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 05/06/2021] [Accepted: 05/27/2021] [Indexed: 12/24/2022]
Abstract
Background The safety and immunogenicity of JNJ-64041809 (JNJ-809), a live-attenuated, double-deleted Listeria monocytogenes (LADD Lm)-based immunotherapy targeting 4 relevant prostate cancer antigens, was evaluated in a phase 1 study in patients with metastatic castration-resistant prostate cancer (mCRPC). Methods Men with progressive mCRPC who had received ≥2 prior approved therapies were enrolled. Primary study objectives were to determine the recommended phase 2 dose (RP2D) and to evaluate the safety and immunogenicity of JNJ-809. Results A total of 26 patients received JNJ-809 (1 × 108 CFU (n = 6); 1 × 109 CFU (n = 20)). No dose-limiting toxicities were reported, and 1 × 109 CFU was selected as the RP2D. The most common adverse events (AEs) reported were chills (92%), pyrexia (81%), and fatigue (62%). The most frequent grade ≥3 AEs were lymphopenia (27%) and hypertension (23%). Serious AEs were reported in 27% of patients including 1 patient with grade 3 intestinal obstruction. JNJ-809 transiently induced peripheral cytokines, including interferon-γ, interleukin-10, and tumor necrosis factor-α. Of the 7 patients evaluable for T cell responses at the 1 × 109 CFU dose, evidence of post-treatment antigenic responses were observed in 6 to the Listeria antigen listeriolysin O and in 5 to ≥1 of the 4 encoded tumor antigens. Best overall response was stable disease in 13/25 response-evaluable patients. The study was terminated early as data collected were considered sufficient to evaluate safety and immunogenicity. Conclusions JNJ-809 has manageable safety consistent with other LADD Lm-based therapies. Limited antigen-specific immune responses were observed, which did not translate into objective clinical responses.
Collapse
|
4
|
Wang P, Zhao H, Ren F, Zhao Q, Shi R, Liu X, Liu J, Li Y, Li Y, Liu H, Chen J. [Research Progress of Epigenetics in Pathogenesis and Treatment of Malignant Tumors]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2020; 23:91-100. [PMID: 32093453 PMCID: PMC7049791 DOI: 10.3779/j.issn.1009-3419.2020.02.04] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
表观遗传学修饰与肿瘤的发生发展密切相关,其主要通过DNA甲基化、组蛋白修饰、非编码RNA调控和染色质结构重构等方式对基因功能和表达水平进行调控,从而影响肿瘤的进展。目前针对表观遗传学的药物已经逐渐应用于恶性肿瘤的治疗,常见的药物类型包括DNA甲基转移酶抑制剂和组蛋白去乙酰化酶抑制剂,但此类药物仍存在诸多不足之处广泛的临床应用仍需要进一步的研究,令人鼓舞的是表观遗传药物与多种抗肿瘤药物联合应用已表现出巨大的应用潜力。本文就表观遗传学在恶性肿瘤的发生发展机制和相关药物的新进展进行了综述。
Collapse
Affiliation(s)
- Pan Wang
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin Lung Cancer Institute, Tianjin Key Laboratory of lung Cancer Metastasis and Tumor Microenvironment, Tianjin 300052, China
| | - Honglin Zhao
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin Lung Cancer Institute, Tianjin Key Laboratory of lung Cancer Metastasis and Tumor Microenvironment, Tianjin 300052, China
| | - Fan Ren
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin Lung Cancer Institute, Tianjin Key Laboratory of lung Cancer Metastasis and Tumor Microenvironment, Tianjin 300052, China
| | - Qingchun Zhao
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin Lung Cancer Institute, Tianjin Key Laboratory of lung Cancer Metastasis and Tumor Microenvironment, Tianjin 300052, China
| | - Ruifeng Shi
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin Lung Cancer Institute, Tianjin Key Laboratory of lung Cancer Metastasis and Tumor Microenvironment, Tianjin 300052, China
| | - Xingyu Liu
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin Lung Cancer Institute, Tianjin Key Laboratory of lung Cancer Metastasis and Tumor Microenvironment, Tianjin 300052, China
| | - Jinghao Liu
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin Lung Cancer Institute, Tianjin Key Laboratory of lung Cancer Metastasis and Tumor Microenvironment, Tianjin 300052, China
| | - Yongwen Li
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin Lung Cancer Institute, Tianjin Key Laboratory of lung Cancer Metastasis and Tumor Microenvironment, Tianjin 300052, China
| | - Ying Li
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin Lung Cancer Institute, Tianjin Key Laboratory of lung Cancer Metastasis and Tumor Microenvironment, Tianjin 300052, China
| | - Hongyu Liu
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin Lung Cancer Institute, Tianjin Key Laboratory of lung Cancer Metastasis and Tumor Microenvironment, Tianjin 300052, China
| | - Jun Chen
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin Lung Cancer Institute, Tianjin Key Laboratory of lung Cancer Metastasis and Tumor Microenvironment, Tianjin 300052, China
| |
Collapse
|
5
|
Bezu L, Wu Chuang A, Liu P, Kroemer G, Kepp O. Immunological Effects of Epigenetic Modifiers. Cancers (Basel) 2019; 11:cancers11121911. [PMID: 31805711 PMCID: PMC6966579 DOI: 10.3390/cancers11121911] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 11/27/2019] [Accepted: 11/28/2019] [Indexed: 12/23/2022] Open
Abstract
Epigenetic alterations are associated with major pathologies including cancer. Epigenetic dysregulation, such as aberrant histone acetylation, altered DNA methylation, or modified chromatin organization, contribute to oncogenesis by inactivating tumor suppressor genes and activating oncogenic pathways. Targeting epigenetic cancer hallmarks can be harnessed as an immunotherapeutic strategy, exemplified by the use of pharmacological inhibitors of DNA methyltransferases (DNMT) and histone deacetylases (HDAC) that can result in the release from the tumor of danger-associated molecular patterns (DAMPs) on one hand and can (re-)activate the expression of tumor-associated antigens on the other hand. This finding suggests that epigenetic modifiers and more specifically the DNA methylation status may change the interaction of chromatin with chaperon proteins including HMGB1, thereby contributing to the antitumor immune response. In this review, we detail how epigenetic modifiers can be used for stimulating therapeutically relevant anticancer immunity when used as stand-alone treatments or in combination with established immunotherapies.
Collapse
Affiliation(s)
- Lucillia Bezu
- Service anesthésie-réanimation, Hôpital européen Georges Pompidou, AP-HP, 75015 Paris, France;
- Faculty of Medicine, University of Paris Sud, 94270 Kremlin-Bicêtre, France;
- Equipe labellisée par la Ligue contre le cancer, 75000, Paris, France;
- Université de Paris, Sorbonne, INSERM U1138, Centre de Recherche des Cordeliers, 75006 Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, 94800 Villejuif, France
| | - Alejandra Wu Chuang
- Faculty of Medicine, University of Paris Sud, 94270 Kremlin-Bicêtre, France;
- Equipe labellisée par la Ligue contre le cancer, 75000, Paris, France;
- Université de Paris, Sorbonne, INSERM U1138, Centre de Recherche des Cordeliers, 75006 Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, 94800 Villejuif, France
| | - Peng Liu
- Equipe labellisée par la Ligue contre le cancer, 75000, Paris, France;
- Université de Paris, Sorbonne, INSERM U1138, Centre de Recherche des Cordeliers, 75006 Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, 94800 Villejuif, France
| | - Guido Kroemer
- Equipe labellisée par la Ligue contre le cancer, 75000, Paris, France;
- Université de Paris, Sorbonne, INSERM U1138, Centre de Recherche des Cordeliers, 75006 Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, 94800 Villejuif, France
- Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, 75015 Paris, France
- Suzhou Institute for Systems Medicine, Chinese Academy of Medical Sciences, 215123 Suzhou, China
- Department of Women’s and Children’s Health, Karolinska Institute, Karolinska University Hospital, 171 76 Stockholm, Sweden
- Correspondence: (G.K.); (O.K.)
| | - Oliver Kepp
- Equipe labellisée par la Ligue contre le cancer, 75000, Paris, France;
- Université de Paris, Sorbonne, INSERM U1138, Centre de Recherche des Cordeliers, 75006 Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, 94800 Villejuif, France
- Correspondence: (G.K.); (O.K.)
| |
Collapse
|
6
|
Fenton SE, Sosman JA, Chandra S. Resistance mechanisms in melanoma to immuneoncologic therapy with checkpoint inhibitors. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2019; 2:744-761. [PMID: 35582566 PMCID: PMC8992532 DOI: 10.20517/cdr.2019.28] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 05/16/2019] [Accepted: 05/22/2019] [Indexed: 11/14/2022]
Abstract
Checkpoint inhibitors act by blocking physiologic mechanisms coopted by tumor cells to evade immune surveillance, restoring the immune system's ability to identify and kill malignant cells. These therapies have dramatically improved outcomes in multiple tumor types with durable responses in many patients, leading to FDA approval first in advanced melanoma, then in many other malignancies. However, as experience with checkpoint inhibitors has grown, populations of patients who are primary nonresponders or develop secondary resistance have been the majority of cases, even in melanoma. Mechanisms of resistance include those inherent to the tumor microenvironment, the tumor cells themselves, and the function of the patient's native immune cells. This review will discuss resistance to checkpoint inhibitors in melanoma as well as possible methods to restore sensitivity.
Collapse
Affiliation(s)
- Sarah E. Fenton
- Division of Hematology Oncology, Northwestern University, Chicago, IL 60611, USA
| | - Jeffrey A. Sosman
- Division of Hematology Oncology, Northwestern University, Chicago, IL 60611, USA
| | - Sunandana Chandra
- Division of Hematology Oncology, Northwestern University, Chicago, IL 60611, USA
| |
Collapse
|
7
|
Arenas-Ramirez N, Sahin D, Boyman O. Epigenetic mechanisms of tumor resistance to immunotherapy. Cell Mol Life Sci 2018; 75:4163-4176. [PMID: 30140960 PMCID: PMC11105392 DOI: 10.1007/s00018-018-2908-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Revised: 08/10/2018] [Accepted: 08/16/2018] [Indexed: 12/14/2022]
Abstract
The recent impact of cancer immunotherapies has firmly established the ability and importance of the immune system to fight malignancies. However, the intimate interaction between the highly dynamic tumor and immune cells leads to a selection process driven by genetic and epigenetic processes. As the molecular pathways of cancer resistance mechanisms to immunotherapy become increasingly known, novel therapeutic targets are being tested in combination with immune-stimulating approaches. We here review recent insights into the molecular mechanisms of tumor resistance with particular emphasis on epigenetic processes and place these in the context of previous models.
Collapse
Affiliation(s)
| | - Dilara Sahin
- Department of Immunology, University Hospital Zurich, 8091, Zurich, Switzerland
| | - Onur Boyman
- Department of Immunology, University Hospital Zurich, 8091, Zurich, Switzerland.
- Faculty of Medicine, University of Zurich, 8006, Zurich, Switzerland.
| |
Collapse
|
8
|
Fan PW, Huang L, Chang XM, Feng YN, Yao X, Peng YC, Dong T, Wang RZ. Human Leukocyte Antigen-A Allele Distribution in Nasopharyngeal Carcinoma Patients Showing Anti-Melanoma-Associated Antigen A or Synovial Sarcoma X-2 T Cell Response in Blood. Chin Med J (Engl) 2018; 131:1289-1295. [PMID: 29786040 PMCID: PMC5987498 DOI: 10.4103/0366-6999.232791] [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] [Indexed: 12/05/2022] Open
Abstract
Background: Development of innovative immunotherapy is imperative to improve the poor survival of the nasopharyngeal carcinoma (NPC) patients. In this study, we evaluated the T cell response to melanoma-associated antigen (MAGE)-A1, MAGE-A3, or synovial sarcoma X-2 (SSX-2) in the peripheral blood of treatment-naive NPC patients. The relationship of responses among the three proteins and the human leukocyte antigen (HLA)-A types were analyzed to provide evidence of designing novel therapy. Methods: Sixty-one NPC patients admitted into the Tumor Hospital affiliated to the Xinjiang Medical University between March 2015 and July 2016 were enrolled. Mononuclear cells were isolated from the peripheral blood before any treatment. HLA-A alleles were typed with Sanger sequence-based typing technique. The T cell response to the MAGE-A1, MAGE-A3, or SSX-2 was evaluated with the Enzyme-Linked ImmunoSpot assay. Mann-Whitney U-test was used to compare the T cell responses from different groups. Spearman's rank correlation was used to analyze the relationship of T cell responses. Results: HLA-A*02:01, A*02:07, and A*24:02 were the three most frequent alleles (18.9%, 12.3%, and 11.5%, respectively) among the 22 detected alleles. 31.1%, 19.7%, and 16.4% of the patients displayed MAGE-A1, MAGE-A3, or SSX-2-specific T cell response, respectively. The magnitudes of response to the three proteins were 32.5, 38.0, and 28.7 SFC/106 peripheral blood mononuclear cells, respectively. The T cell response against the three proteins correlated with each other to different extent. The percentage of A*02:01 and A*24:02 carriers were significantly higher in patients responding to any of the three proteins compared to the nonresponders. Conclusion: MAGE-A1, MAGE-A3, or SSX-2-specific T cell responses were detectable in a subgroup of NPC patients, the frequency and magnitude of which were correlated.
Collapse
Affiliation(s)
- Pei-Wen Fan
- Xinjiang Key Laboratory of Oncology, The Affiliated Tumor Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830000, China
| | - Li Huang
- Department of Radiation Oncology, The Affiliated Tumor Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830000, China
| | - Xue-Mei Chang
- Xinjiang Key Laboratory of Oncology, The Affiliated Tumor Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830000, China
| | - Ya-Ning Feng
- Xinjiang Key Laboratory of Oncology, The Affiliated Tumor Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830000, China
| | - Xuan Yao
- CAMS Oxford Center for Translation Immunology, Chinese Academy of Medical Science Oxford Institute, Nuffield Department of Medicine; MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, Oxford University, Oxford OX3 9DS, UK
| | - Yan-Chun Peng
- CAMS Oxford Center for Translation Immunology, Chinese Academy of Medical Science Oxford Institute, Nuffield Department of Medicine; MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, Oxford University, Oxford OX3 9DS, UK
| | - Tao Dong
- CAMS Oxford Center for Translation Immunology, Chinese Academy of Medical Science Oxford Institute, Nuffield Department of Medicine; MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, Oxford University, Oxford OX3 9DS, UK
| | - Ruo-Zheng Wang
- Xinjiang Key Laboratory of Oncology, The Affiliated Tumor Hospital of Xinjiang Medical University; Department of Radiation Oncology, The Affiliated Tumor Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830000, China
| |
Collapse
|
9
|
Thomas R, Al-Khadairi G, Roelands J, Hendrickx W, Dermime S, Bedognetti D, Decock J. NY-ESO-1 Based Immunotherapy of Cancer: Current Perspectives. Front Immunol 2018; 9:947. [PMID: 29770138 PMCID: PMC5941317 DOI: 10.3389/fimmu.2018.00947] [Citation(s) in RCA: 280] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 04/16/2018] [Indexed: 12/12/2022] Open
Abstract
NY-ESO-1 or New York esophageal squamous cell carcinoma 1 is a well-known cancer-testis antigen (CTAs) with re-expression in numerous cancer types. Its ability to elicit spontaneous humoral and cellular immune responses, together with its restricted expression pattern, have rendered it a good candidate target for cancer immunotherapy. In this review, we provide background information on NY-ESO-1 expression and function in normal and cancerous tissues. Furthermore, NY-ESO-1-specific immune responses have been observed in various cancer types; however, their utility as biomarkers are not well determined. Finally, we describe the immune-based therapeutic options targeting NY-ESO-1 that are currently in clinical trial. We will highlight the recent advancements made in NY-ESO-1 cancer vaccines, adoptive T cell therapy, and combinatorial treatment with checkpoint inhibitors and will discuss the current trends for future NY-ESO-1 based immunotherapy. Cancer treatment has been revolutionized over the last few decades with immunotherapy emerging at the forefront. Immune-based interventions have shown promising results, providing a new treatment avenue for durable clinical responses in various cancer types. The majority of successful immunotherapy studies have been reported in liquid cancers, whereas these approaches have met many challenges in solid cancers. Effective immunotherapy in solid cancers is hampered by the complex, dynamic tumor microenvironment that modulates the extent and phenotype of the antitumor immune response. Furthermore, many solid tumor-associated antigens are not private but can be found in normal somatic tissues, resulting in minor to detrimental off-target toxicities. Therefore, there is an ongoing effort to identify tumor-specific antigens to target using various immune-based modalities. CTAs are considered good candidate targets for immunotherapy as they are characterized by a restricted expression in normal somatic tissues concomitant with a re-expression in solid epithelial cancers. Moreover, several CTAs have been found to induce a spontaneous immune response, NY-ESO-1 being the most immunogenic among the family members. Hence, this review will focus on NY-ESO-1 and discuss the past and current NY-ESO-1 targeted immunotherapeutic strategies.
Collapse
Affiliation(s)
- Remy Thomas
- Cancer Research Center, Qatar Biomedical Research Institute, Qatar Foundation, Hamad Bin Khalifa University, Doha, Qatar
| | - Ghaneya Al-Khadairi
- Cancer Research Center, Qatar Biomedical Research Institute, Qatar Foundation, Hamad Bin Khalifa University, Doha, Qatar
| | - Jessica Roelands
- Immunology, Inflammation, and Metabolism Department, Tumor Biology, Immunology, and Therapy Section, Division of Translational Medicine, Sidra Medicine, Doha, Qatar.,Department of Surgery, Leiden University Medical Center, Leiden, Netherlands
| | - Wouter Hendrickx
- Immunology, Inflammation, and Metabolism Department, Tumor Biology, Immunology, and Therapy Section, Division of Translational Medicine, Sidra Medicine, Doha, Qatar
| | - Said Dermime
- Translational Cancer Research Facility, National Center for Cancer Care and Research, Doha, Qatar
| | - Davide Bedognetti
- Immunology, Inflammation, and Metabolism Department, Tumor Biology, Immunology, and Therapy Section, Division of Translational Medicine, Sidra Medicine, Doha, Qatar
| | - Julie Decock
- Cancer Research Center, Qatar Biomedical Research Institute, Qatar Foundation, Hamad Bin Khalifa University, Doha, Qatar
| |
Collapse
|
10
|
Heninger E, Krueger TEG, Thiede SM, Sperger JM, Byers BL, Kircher MR, Kosoff D, Yang B, Jarrard DF, McNeel DG, Lang JM. Inducible expression of cancer-testis antigens in human prostate cancer. Oncotarget 2018; 7:84359-84374. [PMID: 27769045 PMCID: PMC5341296 DOI: 10.18632/oncotarget.12711] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Accepted: 10/11/2016] [Indexed: 12/23/2022] Open
Abstract
Immune tolerance to self-antigens can limit robust anti-tumor immune responses in the use of tumor vaccines. Expression of novel tumor associated antigens can improve immune recognition and lysis of tumor cells. The cancer-testis antigen (CTA) family of proteins has been hypothesized to be an ideal class of antigens due to tumor-restricted expression, a subset of which have been found to induce antibody responses in patients with prostate disease. We demonstrate that CTA expression is highly inducible in five different Prostate Cancer (PC) cell lines using a hypomethylating agent 5-Aza-2′-deoxycytidine (5AZA) and/or a histone deacetylase inhibitor LBH589. These CTAs include NY-ESO1, multiple members of the MAGE and SSX families and NY-SAR35. A subset of CTAs is synergistically induced by the combination of 5AZA and LBH589. We developed an ex vivo organ culture using human PC biopsies for ex vivo drug treatments to evaluate these agents in clinical samples. These assays found significant induction of SSX2 in 9/9 distinct patient samples and NY-SAR35 in 7/9 samples. Further, we identify expression of SSX2 in circulating tumor cells (CTC) from patients with advanced PC. These results indicate that epigenetic modifying agents can induce expression of a broad range of neoantigens in human PC and may serve as a useful adjunctive therapy with novel tumor vaccines and checkpoint inhibitors.
Collapse
Affiliation(s)
- Erika Heninger
- Department of Medicine, University of Wisconsin, Madison, Madison, WI 53705, USA.,University of Wisconsin Carbone Cancer Center, Madison, Madison, WI 53705, USA
| | - Timothy E G Krueger
- University of Wisconsin Carbone Cancer Center, Madison, Madison, WI 53705, USA
| | - Stephanie M Thiede
- Department of Medicine, University of Wisconsin, Madison, Madison, WI 53705, USA.,University of Wisconsin Carbone Cancer Center, Madison, Madison, WI 53705, USA
| | - Jamie M Sperger
- Department of Medicine, University of Wisconsin, Madison, Madison, WI 53705, USA.,University of Wisconsin Carbone Cancer Center, Madison, Madison, WI 53705, USA
| | - Brianna L Byers
- University of Wisconsin Carbone Cancer Center, Madison, Madison, WI 53705, USA
| | - Madison R Kircher
- University of Wisconsin Carbone Cancer Center, Madison, Madison, WI 53705, USA
| | - David Kosoff
- Department of Medicine, University of Wisconsin, Madison, Madison, WI 53705, USA.,University of Wisconsin Carbone Cancer Center, Madison, Madison, WI 53705, USA
| | - Bing Yang
- Department of Urology, University of Wisconsin, Madison, Madison, WI 53705, USA.,University of Wisconsin Carbone Cancer Center, Madison, Madison, WI 53705, USA
| | - David F Jarrard
- Department of Urology, University of Wisconsin, Madison, Madison, WI 53705, USA.,University of Wisconsin Carbone Cancer Center, Madison, Madison, WI 53705, USA
| | - Douglas G McNeel
- Department of Medicine, University of Wisconsin, Madison, Madison, WI 53705, USA.,University of Wisconsin Carbone Cancer Center, Madison, Madison, WI 53705, USA
| | - Joshua M Lang
- Department of Medicine, University of Wisconsin, Madison, Madison, WI 53705, USA.,University of Wisconsin Carbone Cancer Center, Madison, Madison, WI 53705, USA
| |
Collapse
|
11
|
Bloom JE, McNeel DG. SSX2 regulates focal adhesion but does not drive the epithelial to mesenchymal transition in prostate cancer. Oncotarget 2018; 7:50997-51011. [PMID: 27276714 PMCID: PMC5239454 DOI: 10.18632/oncotarget.9802] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Accepted: 05/26/2016] [Indexed: 12/19/2022] Open
Abstract
Prostate cancer is the most commonly diagnosed malignancy for men in the United States. Metastatic prostate cancer, the lethal form of the disease, has a life expectancy of approximately five years. Identification of factors associated with this transition to metastatic disease is crucial for future therapies. One such factor is the SSX gene family, a family of cancer/testis antigens (CTA) transcription factors which have been shown to be aberrantly expressed in other cancers and associated with the epithelial to mesenchymal transition (EMT). We have previously shown that SSX expression in prostate cancers was restricted to metastatic tissue and not primary tumors. In this study, we have identified SSX2 as the predominant SSX family member expressed in prostate cancer, and found its expression in the peripheral blood of 19 of 54 (35%) prostate cancer patients, with expression restricted to circulating tumor cells, and in 7 of 15 (47%) metastatic cDNA samples. Further, we examined SSX2 function in prostate cancer through knockdown and overexpression in prostate cancer cell lines. While overexpression had little effect on morphology or gene transcript changes, knockdown of SSX2 resulted in an epithelial morphology, increased cell proliferation, increased expression of genes involved in focal adhesion, decreased anchorage independent growth, increased invasion, and increased tumorigenicity in vivo. We conclude from these findings that SSX2 expression in prostate cancer is not a driver of EMT, but is involved in processes associated with EMT including loss of focal adhesion that may be related to tumor cell dissemination.
Collapse
Affiliation(s)
- Jordan E Bloom
- Department of Medicine, University of Wisconsin, Madison, WI, USA
| | - Douglas G McNeel
- Department of Medicine, University of Wisconsin, Madison, WI, USA.,University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, WI, USA
| |
Collapse
|
12
|
Sulek JE, Robinson SP, Petrossian AA, Zhou S, Goliadze E, Manjili MH, Toor A, Guruli G. Role of Epigenetic Modification and Immunomodulation in a Murine Prostate Cancer Model. Prostate 2017; 77:361-373. [PMID: 27862100 DOI: 10.1002/pros.23275] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Accepted: 10/21/2016] [Indexed: 11/07/2022]
Abstract
INTRODUCTION Decreased expression of highly immunogenic cancer-testis antigens (CTA) might help tumor to achieve low immunogenicity, escape immune surveillance and grow unimpeded. Our aim was to evaluate CTA expression in tumor and normal tissues and to investigate possible means of improving the immune response in a murine prostate cancer (CaP) model by using the combination of epigenetic modifier 5-azacitidine (5-AzaC) and immunomodulator lenalidomide. No study to date has examined the effect of this combination on the prostate cancer or its impact on antigen-presenting cells (APC). MATERIALS AND METHODS Gene microarrays were performed to compare expression of several CTA in murine prostate cancer (RM-1 cells) and normal prostate. RM-1 cells were treated with 5-AzaC and real-time PCR was performed to investigate the expression of several CTA. Western blotting was used to determine whether expression of CTA-specific mRNA induced by 5-AzaC resulted in increase in the corresponding protein. Effect of the epigenetic agents and immunomodulators was assessed on dendritic cells (DC) using flow cytometry, ELISA and T-cell proliferation assay. RESULTS Gene arrays demonstrated decreased expression of 35 CTA in CaP tissue compared to normal prostate. 5-AzaC treatment of RM-1 prostate cancer cells upregulated the expression of all 13 CTA tested in a dose-dependent fashion. DC were treated with 5-AzaC and lenalidomide and the expression of surface markers MHC Class I, MHC Class II, CD80, CD86, CD 205, and CD40 was increased. Combination of 5-AzaC and lenalidomide enhances the ability of DC to stimulate T-cell proliferation in mixed leukocyte reaction. Secretion of IL-12 and IL-15 by DC increased significantly with addition of 5-AzaC or 5-AzaC and lenalidomide. CONCLUSIONS Decreased expression of CTA by prostate cancer may be a means of escaping immune monitoring. Combination of epigenetic modifications and immunomodulation by 5-AzaC and lenalidomide increased tumor immunogenicity and enhanced DC function and may be used in the treatment of advanced prostate cancer. Prostate 77: 361-373, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Jay E Sulek
- Division of Urology, Department of Surgery, Virginia Commonwealth University Medical Center, Richmond, Virginia
| | - Samuel P Robinson
- Division of Urology, Department of Surgery, Virginia Commonwealth University Medical Center, Richmond, Virginia
| | - Albert A Petrossian
- Division of Urology, Department of Surgery, Virginia Commonwealth University Medical Center, Richmond, Virginia
| | - Shaoqing Zhou
- Division of Urology, Department of Surgery, Virginia Commonwealth University Medical Center, Richmond, Virginia
| | - Ekaterine Goliadze
- Division of Urology, Department of Surgery, Virginia Commonwealth University Medical Center, Richmond, Virginia
- Massey Cancer Center, Virginia Commonwealth University Medical Center, Richmond, Virginia
| | - Masoud H Manjili
- Massey Cancer Center, Virginia Commonwealth University Medical Center, Richmond, Virginia
- Department of Microbiology and Immunology, Virginia Commonwealth University School of Medicine, Richmond, Virginia
| | - Amir Toor
- Massey Cancer Center, Virginia Commonwealth University Medical Center, Richmond, Virginia
| | - Georgi Guruli
- Division of Urology, Department of Surgery, Virginia Commonwealth University Medical Center, Richmond, Virginia
- Massey Cancer Center, Virginia Commonwealth University Medical Center, Richmond, Virginia
| |
Collapse
|
13
|
Liu M, Zhou J, Chen Z, Cheng ASL. Understanding the epigenetic regulation of tumours and their microenvironments: opportunities and problems for epigenetic therapy. J Pathol 2016; 241:10-24. [PMID: 27770445 DOI: 10.1002/path.4832] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Revised: 10/06/2016] [Accepted: 10/18/2016] [Indexed: 12/13/2022]
Abstract
The tumour microenvironment plays an instrumental role in cancer development, progression and treatment response/resistance. Accumulating evidence is underscoring the fundamental importance of epigenetic regulation in tumour immune evasion. Following many pioneering discoveries demonstrating malignant transformation through epigenetic anomalies ('epimutations'), there is also a growing emphasis on elucidating aberrant epigenetic mechanisms that reprogramme the milieu of tumour-associated immune and stromal cells towards an immunosuppressive state. Pharmacological inhibition of DNA methylation and histone modifications can augment the efficiency of immune checkpoint blockage, and unleash anti-tumour T-cell responses. However, these non-specific agents also represent a 'double-edged sword', as they can also reactivate gene transcription of checkpoint molecules, interrupting immune surveillance programmes. By understanding the impact of epigenetic control on the tumour microenvironment, rational combinatorial epigenetic and checkpoint blockage therapies have the potential to harness the immune system for the treatment of cancer. Copyright © 2016 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Man Liu
- School of Biomedical Sciences and State Key Laboratory of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, SAR, PR China
| | - Jingying Zhou
- School of Biomedical Sciences and State Key Laboratory of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, SAR, PR China
| | - Zhiwei Chen
- AIDS Institute and Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, PR China
| | - Alfred Sze-Lok Cheng
- School of Biomedical Sciences and State Key Laboratory of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, SAR, PR China
| |
Collapse
|
14
|
Hassan HE, Keita JA, Narayan L, Brady SM, Frederick R, Carlson S, C Glass K, Natesan S, Buttolph T, Fandy TE. The combination of dimethoxycurcumin with DNA methylation inhibitor enhances gene re-expression of promoter-methylated genes and antagonizes their cytotoxic effect. Epigenetics 2016; 11:740-749. [PMID: 27588609 DOI: 10.1080/15592294.2016.1226452] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Curcumin and its analogs exhibited antileukemic activity either as single agent or in combination therapy. Dimethoxycurcumin (DMC) is a more metabolically stable curcumin analog that was shown to induce the expression of promoter-methylated genes without reversing DNA methylation. Accordingly, co-treatment with DMC and DNA methyltransferase (DNMT) inhibitors could hypothetically enhance the re-expression of promoter-methylated tumor suppressor genes. In this study, we investigated the cytotoxic effects and epigenetic changes associated with the combination of DMC and the DNMT inhibitor decitabine (DAC) in primary leukemia samples and cell lines. The combination demonstrated antagonistic cytotoxic effects and was minimally cytotoxic to primary leukemia cells. The combination did not affect the metabolic stability of DMC. Although the combination enhanced the downregulation of nuclear DNMT proteins, the hypomethylating activity of the combination was not increased significantly compared to DAC alone. On the other hand, the combination significantly increased H3K27 acetylation (H3K27Ac) compared to the single agents near the promoter region of promoter-methylated genes. Furthermore, sequential chromatin immunoprecipitation (ChIP) and DNA pyrosequencing of the chromatin-enriched H3K27Ac did not show any significant decrease in DNA methylation compared to other regions. Consequently, the enhanced induction of promoter-methylated genes by the combination compared to DAC alone is mediated by a mechanism that involves increased histone acetylation and not through potentiation of the DNA hypomethylating activity of DAC. Collectively, our results provide the mechanistic basis for further characterization of this combination in leukemia animal models and early phase clinical trials.
Collapse
Affiliation(s)
- Hazem E Hassan
- a Department of Pharmaceutical Sciences , University of Maryland , Baltimore , MD , USA.,b Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy , Helwan University , Cairo , Egypt
| | - Jean-Arnaud Keita
- c Department of Pharmaceutical Sciences , Albany College of Pharmacy (Vermont Campus) , Colchester , VT , USA
| | - Lawrence Narayan
- c Department of Pharmaceutical Sciences , Albany College of Pharmacy (Vermont Campus) , Colchester , VT , USA
| | - Sean M Brady
- c Department of Pharmaceutical Sciences , Albany College of Pharmacy (Vermont Campus) , Colchester , VT , USA
| | - Richard Frederick
- c Department of Pharmaceutical Sciences , Albany College of Pharmacy (Vermont Campus) , Colchester , VT , USA
| | - Samuel Carlson
- c Department of Pharmaceutical Sciences , Albany College of Pharmacy (Vermont Campus) , Colchester , VT , USA
| | - Karen C Glass
- c Department of Pharmaceutical Sciences , Albany College of Pharmacy (Vermont Campus) , Colchester , VT , USA
| | - Senthil Natesan
- d Department of Experimental and Systems Pharmacology , Washington State University , Spokane , WA , USA
| | - Thomm Buttolph
- e Department of Neurological Sciences , University of Vermont , Burlington , VT , USA
| | - Tamer E Fandy
- c Department of Pharmaceutical Sciences , Albany College of Pharmacy (Vermont Campus) , Colchester , VT , USA
| |
Collapse
|
15
|
Acevedo M, Vernier M, Mignacca L, Lessard F, Huot G, Moiseeva O, Bourdeau V, Ferbeyre G. A CDK4/6-Dependent Epigenetic Mechanism Protects Cancer Cells from PML-induced Senescence. Cancer Res 2016; 76:3252-64. [PMID: 27206849 DOI: 10.1158/0008-5472.can-15-2347] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Accepted: 03/10/2016] [Indexed: 12/17/2022]
Abstract
Promyelocytic leukemia (PML) plays a tumor suppressive role by inducing cellular senescence in response to oncogenic stress. However, tumor cell lines fail to engage in complete senescence upon PML activation. In this study, we investigated the mechanisms underlying resistance to PML-induced senescence. Here, we report that activation of the cyclin-dependent kinases CDK4 and CDK6 are essential and sufficient to impair senescence induced by PML expression. Disrupting CDK function by RNA interference or pharmacological inhibition restored senescence in tumor cells and diminished their tumorigenic potential in mouse xenograft models. Complete senescence correlated with an increase in autophagy, repression of E2F target genes, and an gene expression signature of blocked DNA methylation. Accordingly, treatment of tumor cells with inhibitors of DNA methylation reversed resistance to PML-induced senescence. Further, CDK inhibition with palbociclib promoted autophagy-dependent degradation of the DNA methyltransferase DNMT1. Lastly, we found that CDK4 interacted with and phosphorylated DNMT1 in vitro, suggesting that CDK activity is required for its stabilization. Taken together, our findings highlight a potentially valuable feature of CDK4/6 inhibitors as epigenetic modulators to facilitate activation of senescence programs in tumor cells. Cancer Res; 76(11); 3252-64. ©2016 AACR.
Collapse
Affiliation(s)
- Mariana Acevedo
- Department of Biochemistry and Molecular Medicine, Université de Montréal, Montréal, Québec, Canada
| | - Mathieu Vernier
- Department of Biochemistry and Molecular Medicine, Université de Montréal, Montréal, Québec, Canada
| | - Lian Mignacca
- Department of Biochemistry and Molecular Medicine, Université de Montréal, Montréal, Québec, Canada
| | - Frédéric Lessard
- Department of Biochemistry and Molecular Medicine, Université de Montréal, Montréal, Québec, Canada
| | - Geneviève Huot
- Department of Biochemistry and Molecular Medicine, Université de Montréal, Montréal, Québec, Canada
| | - Olga Moiseeva
- Department of Biochemistry and Molecular Medicine, Université de Montréal, Montréal, Québec, Canada
| | - Véronique Bourdeau
- Department of Biochemistry and Molecular Medicine, Université de Montréal, Montréal, Québec, Canada.
| | - Gerardo Ferbeyre
- Department of Biochemistry and Molecular Medicine, Université de Montréal, Montréal, Québec, Canada.
| |
Collapse
|
16
|
Schwarzenbacher D, Stiegelbauer V, Deutsch A, Ress AL, Aigelsreiter A, Schauer S, Wagner K, Langsenlehner T, Resel M, Gerger A, Ling H, Ivan C, Calin GA, Hoefler G, Rinner B, Pichler M. Low spinophilin expression enhances aggressive biological behavior of breast cancer. Oncotarget 2016; 6:11191-202. [PMID: 25857299 PMCID: PMC4484449 DOI: 10.18632/oncotarget.3586] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Accepted: 02/19/2015] [Indexed: 11/25/2022] Open
Abstract
Spinophilin, a putative tumor suppressor gene, has been shown to be involved in the pathogenesis of certain types of cancer, but its role has never been systematically explored in breast cancer. In this study, we determined for the first time the expression pattern of spinophilin in human breast cancer molecular subtypes (n = 489) and correlated it with survival (n = 921). We stably reduced spinophilin expression in breast cancer cells and measured effects on cellular growth, apoptosis, anchorage-independent growth, migration, invasion and self-renewal capacity in vitro and metastases formation in vivo. Microarray profiling was used to determine the most abundantly expressed genes in spinophilin-silenced breast cancer cells. Spinophilin expression was significantly lower in basal-like breast cancer (p<0.001) and an independent poor prognostic factor in breast cancer patients (hazard ratio = 1.93, 95% confidence interval: 1.24-3.03; p = 0.004) A reduction of spinophilin levels increased cellular growth in breast cancer cells (p<0.05), without influencing activation of apoptosis. Anchorage-independent growth, migration and self-renewal capacity in vitro and metastatic potential in vivo were also significantly increased in spinophilin-silenced cells (p<0.05). Finally, we identified several differentially expressed genes in spinophilin-silenced cells. According to our data, low levels of spinophilin are associated with aggressive behavior of breast cancer.
Collapse
Affiliation(s)
| | - Verena Stiegelbauer
- Division of Oncology, Department of Internal Medicine, Medical University of Graz, Austria
| | - Alexander Deutsch
- Division of Hematology, Department of Internal Medicine, Medical University of Graz, Austria
| | - Anna Lena Ress
- Division of Oncology, Department of Internal Medicine, Medical University of Graz, Austria
| | | | - Silvia Schauer
- Institute of Pathology, Medical University of Graz, Austria
| | - Karin Wagner
- Center for Medical Research, Medical University of Graz, Austria
| | - Tanja Langsenlehner
- Department of Therapeutic Radiology and Oncology, Medical University of Graz, Austria
| | - Margit Resel
- Division of Oncology, Department of Internal Medicine, Medical University of Graz, Austria
| | - Armin Gerger
- Division of Oncology, Department of Internal Medicine, Medical University of Graz, Austria
| | - Hui Ling
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, TX, USA
| | - Cristina Ivan
- Center for RNA Interference and Non-Coding RNAs, The University of Texas MD Anderson Cancer Center, TX, USA
| | - George Adrian Calin
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, TX, USA.,Center for RNA Interference and Non-Coding RNAs, The University of Texas MD Anderson Cancer Center, TX, USA
| | - Gerald Hoefler
- Institute of Pathology, Medical University of Graz, Austria
| | - Beate Rinner
- Center for Medical Research, Medical University of Graz, Austria
| | - Martin Pichler
- Division of Oncology, Department of Internal Medicine, Medical University of Graz, Austria.,Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, TX, USA
| |
Collapse
|
17
|
Wachowska M, Muchowicz A, Golab J. Targeting Epigenetic Processes in Photodynamic Therapy-Induced Anticancer Immunity. Front Oncol 2015; 5:176. [PMID: 26284197 PMCID: PMC4519687 DOI: 10.3389/fonc.2015.00176] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Accepted: 07/16/2015] [Indexed: 01/04/2023] Open
Abstract
Photodynamic therapy (PDT) of cancer is an approved therapeutic procedure that generates oxidative stress leading to cell death of tumor and stromal cells. Cell death resulting from oxidative damage to intracellular components leads to the release of damage-associated molecular patterns (DAMPs) that trigger robust inflammatory response and creates local conditions for effective sampling of tumor-associated antigens (TAA) by antigen-presenting cells. The latter can trigger development of TAA-specific adaptive immune response. However, due to a number of mechanisms, including epigenetic regulation of TAA expression, tumor cells evade immune recognition. Therefore, numerous approaches are being developed to combine PDT with immunotherapies to allow development of systemic immunity. In this review, we describe immunoregulatory mechanisms of epigenetic treatments that were shown to restore the expression of epigenetically silenced or down-regulated major histocompatibility complex molecules as well as TAA. We also discuss the results of our recent studies showing that epigenetic treatments based on administration of methyltransferase inhibitors in combination with PDT can release effective mechanisms leading to development of antitumor immunity and potentiated antitumor effects.
Collapse
Affiliation(s)
| | - Angelika Muchowicz
- Department of Immunology, Medical University of Warsaw , Warsaw , Poland
| | - Jakub Golab
- Department of Immunology, Medical University of Warsaw , Warsaw , Poland
| |
Collapse
|
18
|
Héninger E, Krueger TEG, Lang JM. Augmenting antitumor immune responses with epigenetic modifying agents. Front Immunol 2015; 6:29. [PMID: 25699047 PMCID: PMC4316783 DOI: 10.3389/fimmu.2015.00029] [Citation(s) in RCA: 109] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Accepted: 01/14/2015] [Indexed: 12/31/2022] Open
Abstract
Epigenetic silencing of immune-related genes is a striking feature of the cancer genome that occurs in the process of tumorigenesis. This phenomena impacts antigen processing and antigen presentation by tumor cells and facilitates evasion of immunosurveillance. Further modulation of the tumor microenvironment by altered expression of immunosuppressive cytokines impairs antigen-presenting cells and cytolytic T-cell function. The potential reversal of immunosuppression by epigenetic modulation is therefore a promising and versatile therapeutic approach to reinstate endogenous immune recognition and tumor lysis. Pre-clinical studies have identified multiple elements of the immune system that can be modulated by epigenetic mechanisms and result in improved antigen presentation, effector T-cell function, and breakdown of suppressor mechanisms. Recent clinical studies are utilizing epigenetic therapies prior to, or in combination with, immune therapies to improve clinical outcomes.
Collapse
Affiliation(s)
- Erika Héninger
- University of Wisconsin Carbone Cancer Center , Madison, WI , USA
| | | | - Joshua M Lang
- University of Wisconsin Carbone Cancer Center , Madison, WI , USA ; Department of Medicine, University of Wisconsin , Madison, WI , USA
| |
Collapse
|
19
|
Xia C, Leon-Ferre R, Laux D, Deutsch J, Smith BJ, Frees M, Milhem M. Treatment of resistant metastatic melanoma using sequential epigenetic therapy (decitabine and panobinostat) combined with chemotherapy (temozolomide). Cancer Chemother Pharmacol 2014; 74:691-7. [PMID: 25062770 PMCID: PMC4175037 DOI: 10.1007/s00280-014-2501-1] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2013] [Accepted: 05/27/2014] [Indexed: 01/14/2023]
Abstract
Purpose To explore the safety and tolerability of combining two epigenetic drugs: decitabine (a DNA methyltransferase inhibitor) and panobinostat (a histone deacetylase inhibitor), with chemotherapy with temozolomide (an alkylating agent). The purpose of such combination is to evaluate the use of epigenetic priming to overcome resistance of melanoma to chemotherapy. Methods A Phase I clinical trial enrolling patients aged 18 years or older, with recurrent or unresectable stage III or IV melanoma of any site. This trial was conducted with full Institutional Review Board approval and was registered with the National Institutes of Health under the clinicaltrials.gov identifier NCT00925132. Patients were treated with subcutaneous decitabine 0.1 or 0.2 mg/kg three times weekly for 2 weeks (starting on day 1), in combination with oral panobinostat 10, 20, or 30 mg every 96 h (starting on day 8), and oral temozolomide 150 mg/m2/day on days 9 through 13. In cycle 2, temozolomide was increased to 200 mg/m2/day if neutropenia or thrombocytopenia had not occurred. Each cycle lasted 6 weeks, and patients could receive up to six cycles. Patients who did not demonstrate disease progression were eligible to enter a maintenance protocol with combination of weekly panobinostat and thrice-weekly decitabine until tumor progression, unacceptable toxicity, or withdrawal of consent. Results Twenty patients were initially enrolled, with 17 receiving treatment. The median age was 56 years. Eleven (65 %) were male, and 6 (35 %) were female. Eleven (64.7 %) had cutaneous melanoma, 4 (23.5 %) had ocular melanoma, and 2 (11.8 %) had mucosal melanoma. All patients received at least one treatment cycle and were evaluable for toxicity. Patients received a median of two 6-week treatment cycles (range 1–6). None of the patients experienced DLT. MTD was not reached. Adverse events attributed to treatment included grade 3 lymphopenia (24 %), anemia (12 %), neutropenia (12 %), and fatigue (12 %), as well as grade 2 leukopenia (30 %), neutropenia (23 %), nausea (23 %), and lymphopenia (18 %). The most common reason for study discontinuation was disease progression. Conclusions This triple agent of dual epigenetic therapy in combination with traditional chemotherapy was generally well tolerated by the cohort and appeared safe to be continued in a Phase II trial. No DLTs were observed, and MTD was not reached.
Collapse
Affiliation(s)
- Chang Xia
- Division of Hematology, Oncology and Bone and Marrow Transplantation, Department of Internal Medicine, University of Iowa Hospitals and Clinics, 200 Hawkins Drive, Iowa City, IA, 52242, USA
| | | | | | | | | | | | | |
Collapse
|
20
|
Abate-Daga D, Speiser DE, Chinnasamy N, Zheng Z, Xu H, Feldman SA, Rosenberg SA, Morgan RA. Development of a T cell receptor targeting an HLA-A*0201 restricted epitope from the cancer-testis antigen SSX2 for adoptive immunotherapy of cancer. PLoS One 2014; 9:e93321. [PMID: 24681846 PMCID: PMC3969312 DOI: 10.1371/journal.pone.0093321] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Accepted: 03/04/2014] [Indexed: 11/18/2022] Open
Abstract
The clinical success of adoptive immunotherapy of cancer relies on the selection of target antigens that are highly expressed in tumor cells but absent in essential normal tissues. A group of genes that encode the cancer/testis or cancer germline antigens have been proposed as ideal targets for immunotherapy due to their high expression in multiple cancer types and their restricted expression in immunoprivileged normal tissues. In the present work we report the isolation and characterization of human T cell receptors (TCRs) with specificity for synovial sarcoma X breakpoint 2 (SSX2), a cancer/testis antigen expressed in melanoma, prostate cancer, lymphoma, multiple myeloma and pancreatic cancer, among other tumors. We isolated seven HLA-A2 restricted T cell receptors from natural T cell clones derived from tumor-infiltrated lymph nodes of two SSX2-seropositive melanoma patients, and selected four TCRs for cloning into retroviral vectors. Peripheral blood lymphocytes (PBL) transduced with three of four SSX2 TCRs showed SSX241-49 (KASEKIFYV) peptide specific reactivity, tumor cell recognition and tetramer binding. One of these, TCR-5, exhibited tetramer binding in both CD4 and CD8 cells and was selected for further studies. Antigen-specific and HLA-A*0201-restricted interferon-γ release, cell lysis and lymphocyte proliferation was observed following culture of TCR engineered human PBL with relevant tumor cell lines. Codon optimization was found to increase TCR-5 expression in transduced T cells, and this construct has been selected for development of clinical grade viral vector producing cells. The tumor-specific pattern of expression of SSX2, along with the potent and selective activity of TCR-5, makes this TCR an attractive candidate for potential TCR gene therapy to treat multiple cancer histologies.
Collapse
Affiliation(s)
- Daniel Abate-Daga
- Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
- * E-mail:
| | - Daniel E. Speiser
- Department of Oncology, Ludwig Center, University of Lausanne, Lausanne, Switzerland
| | - Nachimuthu Chinnasamy
- Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Zhili Zheng
- Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Hui Xu
- Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Steven A. Feldman
- Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Steven A. Rosenberg
- Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Richard A. Morgan
- Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| |
Collapse
|
21
|
Greve KBV, Pøhl M, Olsen KE, Nielsen O, Ditzel HJ, Gjerstorff MF. SSX2-4 expression in early-stage non-small cell lung cancer. ACTA ACUST UNITED AC 2014; 83:344-9. [PMID: 24645645 DOI: 10.1111/tan.12340] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2013] [Revised: 02/19/2014] [Accepted: 02/20/2014] [Indexed: 11/29/2022]
Abstract
The expression of cancer/testis antigens SSX2, SSX3, and SSX4 in non-small cell lung cancers (NSCLC) was examined, since they are considered promising targets for cancer immunotherapy due to their immunogenicity and testis-restricted normal tissue expression. We characterized three SSX antibodies and performed immunohistochemical staining of 25 different normal tissues and 143 NSCLCs. The antibodies differed in binding to two distinctive splice variants of SSX2 that exhibited different subcellular staining patterns, suggesting that the two splice variants display different functions. SSX2-4 expression was only detected in 5 of 143 early-stage NSCLCs, which is rare compared to other cancer/testis antigens (e.g. MAGE-A and GAGE). However, further studies are needed to determine whether SSX can be used as a prognostic or predictive biomarker in NSCLC.
Collapse
Affiliation(s)
- K B V Greve
- Department of Cancer and Inflammation Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | | | | | | | | | | |
Collapse
|
22
|
Smith HA, Rekoske BT, McNeel DG. DNA vaccines encoding altered peptide ligands for SSX2 enhance epitope-specific CD8+ T-cell immune responses. Vaccine 2014; 32:1707-15. [PMID: 24492013 DOI: 10.1016/j.vaccine.2014.01.048] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2013] [Revised: 12/19/2013] [Accepted: 01/18/2014] [Indexed: 12/22/2022]
Abstract
Plasmid DNA serves as a simple and easily modifiable form of antigen delivery for vaccines. The USDA approval of DNA vaccines for several non-human diseases underscores the potential of this type of antigen delivery method as a cost-effective approach for the treatment or prevention of human diseases, including cancer. However, while DNA vaccines have demonstrated safety and immunological effect in early phase clinical trials, they have not consistently elicited robust anti-tumor responses. Hence many recent efforts have sought to increase the immunological efficacy of DNA vaccines, and we have specifically evaluated several target antigens encoded by DNA vaccine as treatments for human prostate cancer. In particular, we have focused on SSX2 as one potential target antigen, given its frequent expression in metastatic prostate cancer. We have previously identified two peptides, p41-49 and p103-111, as HLA-A2-restricted SSX2-specific epitopes. In the present study we sought to determine whether the efficacy of a DNA vaccine could be enhanced by an altered peptide ligand (APL) strategy wherein modifications were made to anchor residues of these epitopes to enhance or ablate their binding to HLA-A2. A DNA vaccine encoding APL modified to increase epitope binding elicited robust peptide-specific CD8+ T cells producing Th1 cytokines specific for each epitope. Ablation of one epitope in a DNA vaccine did not enhance immune responses to the other epitope. These results demonstrate that APL encoded by a DNA vaccine can be used to elicit increased numbers of antigen-specific T cells specific for multiple epitopes simultaneously, and suggest this could be a general approach to improve the immunogenicity of DNA vaccines encoding tumor antigens.
Collapse
Affiliation(s)
- Heath A Smith
- Department of Oncology, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Brian T Rekoske
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Douglas G McNeel
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA.
| |
Collapse
|
23
|
Abstract
Tumor cells frequently exhibit widespread epigenetic aberrations that significantly alter the repertoire of expressed proteins. In particular, it has been known for nearly 25 years that tumors frequently reactivate genes whose expression is typically restricted to germ cells. These gene products are classified as cancer/testis antigens (CTAs) owing to their biased expression pattern and their immunogenicity in cancer patients. While these genes have been pursued as targets for anticancer vaccines, whether these reactivated testis proteins have roles in supporting tumorigenic features is less studied. Recent evidence now indicates that these proteins can be directly employed by the tumor cell regulatory environment to support cell-autonomous behaviors. Here, we review the history of the CTA field and present recent findings indicating that CTAs can play functional roles in supporting tumorigenesis.
Collapse
|
24
|
Lymphocyte cytosolic protein 1 is a chronic lymphocytic leukemia membrane-associated antigen critical to niche homing. Blood 2013; 122:3308-16. [PMID: 24009233 DOI: 10.1182/blood-2013-05-504597] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Membrane antigens are critical to the pathogenesis of chronic lymphocytic leukemia (CLL) as they facilitate microenvironment homing, proliferation, and survival. Targeting the CLL membrane and associated signaling patterns is a current focus of therapeutic development. Many tumor membrane targets are simultaneously targeted by humoral immunity, thus forming recognizable immunoglobulin responses. We sought to use this immune response to identify novel membrane-associated targets for CLL. Using a novel strategy, we interrogated CLL membrane-specific autologous immunoglobulin G reactivity. Our analysis unveiled lymphocyte cytosolic protein 1 (LCP1), a lymphocyte-specific target that is highly expressed in CLL. LCP1 plays a critical role in B-cell biology by crosslinking F-actin filaments, thereby solidifying cytoskeletal structures and providing a scaffold for critical signaling pathways. Small interfering RNA knockdown of LCP1 blocked migration toward CXCL12 in transwell assays and to bone marrow in an in vivo xenotransplant model, confirming a role for LCP1 in leukemia migration. Furthermore, we demonstrate that the Bruton's tyrosine kinase inhibitor ibrutinib or the PI3K inhibitor idelalisib block B-cell receptor induced activation of LCP1. Our data demonstrate a novel strategy to identify cancer membrane target antigens using humoral anti-tumor immunity. In addition, we identify LCP1 as a membrane-associated target in CLL with confirmed pathogenic significance. This clinical trial was registered at clinicaltrials.gov; study ID number: OSU-0025 OSU-0156.
Collapse
|
25
|
Kulkarni P, Shiraishi T, Rajagopalan K, Kim R, Mooney SM, Getzenberg RH. Cancer/testis antigens and urological malignancies. Nat Rev Urol 2012; 9:386-96. [PMID: 22710665 DOI: 10.1038/nrurol.2012.117] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Cancer/testis antigens (CTAs) are a group of tumour-associated antigens (TAAs) that display normal expression in the adult testis--an immune-privileged organ--but aberrant expression in several types of cancers, particularly in advanced cancers with stem cell-like characteristics. There has been an explosion in CTA-based research since CTAs were first identified in 1991 and MAGE-1 was shown to elicit an autologous cytotoxic T-lymphocyte (CTL) response in a patient with melanoma. The resulting data have not only highlighted a role for CTAs in tumorigenesis, but have also underscored the translational potential of these antigens for detecting and treating many types of cancers. Studies that have investigated the use of CTAs for the clinical management of urological malignancies indicate that these TAAs have potential roles as novel biomarkers, with increased specificity and sensitivity compared to those currently used in the clinic, and therapeutic targets for cancer immunotherapy. Increasing evidence supports the utilization of these promising tools for urological indications.
Collapse
Affiliation(s)
- Prakash Kulkarni
- James Buchanan Brady Urological Institute, 600 North Wolfe Street, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.
| | | | | | | | | | | |
Collapse
|
26
|
Downstream and intermediate interactions of synovial sarcoma-associated fusion oncoproteins and their implication for targeted therapy. Sarcoma 2012; 2012:249219. [PMID: 22550415 PMCID: PMC3329658 DOI: 10.1155/2012/249219] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2011] [Accepted: 01/09/2012] [Indexed: 12/14/2022] Open
Abstract
Synovial sarcoma (SS), an aggressive type of soft tissue tumor, occurs mostly in adolescents and young adults. The origin and molecular mechanism of the development of SS remain only partially known. Over 90% of SS cases are characterized by the t(X;18)(p11.2;q11.2) translocation, which results mainly in the formation of
SS18-SSX1 or SS18-SSX2 fusion genes. In recent years, several reports describing direct and indirect interactions of SS18-SSX1/SSX2 oncoproteins have been published. These reports suggest that the fusion proteins particularly affect the cell growth, cell proliferation, TP53 pathway, and chromatin remodeling mechanisms, contributing to SS oncogenesis. Additional research efforts are required to fully explore the protein-protein interactions of SS18-SSX oncoproteins and the pathways that are regulated by these partnerships for the development of effective targeted therapy.
Collapse
|
27
|
Vaccines targeting the cancer-testis antigen SSX-2 elicit HLA-A2 epitope-specific cytolytic T cells. J Immunother 2012; 34:569-80. [PMID: 21904219 DOI: 10.1097/cji.0b013e31822b5b1d] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The cancer-testis antigen synovial sarcoma X breakpoint-2 (SSX-2) is a potentially attractive target for tumor immunotherapy based upon its tissue-restricted expression to germline cells and its frequent expression in malignancies. The goal of this study was to evaluate genetic vaccine encoding SSX-2 to prioritize human leukocyte antigen (HLA)-A2-specific epitopes and determine if a DNA vaccine can elicit SSX-2-specific cytotoxic T lymphocytes (CTLs) capable of lysing prostate cancer cells. HLA-A2-restricted epitopes were identified based on their in vitro binding affinity for HLA-A2 and by the ability of a genetic vaccine to elicit peptide-specific CTL in A2/DR1 (HLA-A2.1+/HLA-DR1+/H-2 class I-/class II-knockout) transgenic mice. We found that SSX-2 peptides p41-49 (KASEKIFYV) and p103-111 (RLQGISPKI) had high affinity for HLA-A2 and were immunogenic in vivo; however, peptide p103-111 was immunodominant with robust peptide-specific immune responses elicited in mice vaccinated with a plasmid DNA vaccine encoding SSX-2. Furthermore, p103-111-specific CTLs were able to lyse an HLA-A2+ prostate cancer cell line. The immunodominance of this epitope was found not to be due to a putative HLA-DR1 epitope (p98-112) flanking p103-111. Finally, we demonstrated that SSX-2 epitope-specific CTLs could be detected and cultured from the peripheral blood of HLA-A2+ prostate cancer patients, notably patients with advanced prostate cancer. Overall, we conclude that SSX-2 peptide p103-111 is an immunodominant HLA-A2-restricted epitope, and epitope-specific CD8 T cells can be detected in patients with prostate cancer, suggesting that tolerance to SSX-2 can be circumvented in vivo. Together, these findings suggest that SSX-2 may be a relevant target antigen for prostate cancer vaccine approaches.
Collapse
|
28
|
Smith HA, Cronk RJ, Lang JM, McNeel DG. Expression and immunotherapeutic targeting of the SSX family of cancer-testis antigens in prostate cancer. Cancer Res 2011; 71:6785-95. [PMID: 21880588 DOI: 10.1158/0008-5472.can-11-2127] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Recent U.S. Food and Drug Administration approval of the first immunotherapy for prostate cancer encourages efforts to improve immune targeting of this disease. The synovial sarcoma X chromosome breakpoint (SSX) proteins comprise a set of cancer-testis antigens that are upregulated in MHC class I-deficient germline cells and in various types of advanced cancers with a poor prognosis. Humoral and cell-mediated immune responses to the SSX family member SSX2 can arise spontaneously in prostate cancer patients. Thus, SSX2 and other proteins of the SSX family may offer useful targets for tumor immunotherapy. In this study, we evaluated the expression of SSX family members in prostate cancer cell lines and tumor biopsies to identify which members might be most appropriate for immune targeting. We found that SSX2 was expressed most frequently in prostate cell lines, but that SSX1 and SSX5 were also expressed after treatment with the DNA demethylating agent 5-aza-2'-deoxycytidine. Immunohistochemical analysis of microarrayed tissue biopsies confirmed a differential level of SSX protein expression in human prostate cancers. Notably, SSX expression in patient tumor samples was restricted to metastatic lesions (5/22; 23%) and no expression was detected in primary prostate tumors examined (0/73; P < 0.001). We determined that cross-reactive immune responses to a dominant HLA-A2-specific SSX epitope (p103-111) could be elicited by immunization of A2/DR1 transgenic mice with SSX vaccines. Our findings suggest that multiple SSX family members are expressed in metastatic prostate cancers which are amenable to simultaneous targeting.
Collapse
Affiliation(s)
- Heath A Smith
- Department of Medicine and Oncology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | | | | | | |
Collapse
|
29
|
Xie C, Kim HJ, Haw JG, Kalbasi A, Gardner BK, Li G, Rao J, Chia D, Liong M, Punzalan RR, Marks LS, Pantuck AJ, de la Taille A, Wang G, Mukouyama H, Zeng G. A novel multiplex assay combining autoantibodies plus PSA has potential implications for classification of prostate cancer from non-malignant cases. J Transl Med 2011; 9:43. [PMID: 21504557 PMCID: PMC3102624 DOI: 10.1186/1479-5876-9-43] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2011] [Accepted: 04/19/2011] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND The lack of sufficient specificity and sensitivity among conventional cancer biomarkers, such as prostate specific antigen (PSA) for prostate cancer has been widely recognized after several decades of clinical implications. Autoantibodies (autoAb) among others are being extensively investigated as potential substitute markers, but remain elusive. One major obstacle is the lack of a sensitive and multiplex approach for quantifying autoAb against a large panel of clinically relevant tumor-associated antigens (TAA). METHODS To circumvent preparation of phage lysates and purification of recombinant proteins, we identified B cell epitopes from a number of previously defined prostate cancer-associated antigens (PCAA). Peptide epitopes from cancer/testis antigen NY-ESO-1, XAGE-1b, SSX-2,4, as well as prostate cancer overexpressed antigen AMACR, p90 autoantigen, and LEDGF were then conjugated with seroMAP microspheres to allow multiplex measurement of autoAb present in serum samples. Moreover, simultaneous quantification of autoAb plus total PSA was achieved in one reaction, and termed the "A+PSA" assay. RESULTS Peptide epitopes from the above 6 PCAA were identified and confirmed that autoAb against these peptide epitopes reacted specifically with the full-length protein. A pilot study was conducted with the A+PSA assay using pre-surgery sera from 131 biopsy-confirmed prostate cancer patients and 121 benign prostatic hyperplasia and/or prostatitis patients. A logistic regression-based A+PSA index was found to enhance sensitivities and specificities over PSA alone in distinguishing prostate cancer from nonmalignant cases. The A+PSA index also reduced false positive rate and improved the area under a receiver operating characteristic curve. CONCLUSIONS The A+PSA assay represents a novel platform that integrates autoAb signatures with a conventional cancer biomarker, which may aid in the diagnosis and prognosis of prostate cancer and others.
Collapse
Affiliation(s)
- Chong Xie
- Department of Urology, Zhongshan Hospital of Fudan University, No.180 Fenglin Road, Shanghai 200032, China
| | - Hyun J Kim
- Department of Radiology, David Geffen School of Medicine at UCLA, 10833 Le Conte Ave, Los Angeles, CA 90095-1721, USA
| | - Jonathan G Haw
- Department of Urology, David Geffen School of Medicine at UCLA, 10833 Le Conte Ave, Los Angeles, CA 90095-1738, USA
| | - Anusha Kalbasi
- Department of Urology, David Geffen School of Medicine at UCLA, 10833 Le Conte Ave, Los Angeles, CA 90095-1738, USA
| | - Brian K Gardner
- Department of Medicine, David Geffen School of Medicine at UCLA, 10833 Le Conte Ave, Los Angeles, CA 90095-1732, USA
| | - Gang Li
- Department of Biostatistics, UCLA School of Public Health, 10833 Le Conte Ave, Los Angeles, CA 90095-1772, USA
| | - Jianyu Rao
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, 10833 Le Conte Ave, Los Angeles, CA 90095-1738, USA
| | - David Chia
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, 10833 Le Conte Ave, Los Angeles, CA 90095-1738, USA
| | - Monty Liong
- Department of Chemistry and Biochemistry, 607 Charles E. Young Drive East, Los Angeles, CA 90095-1569, USA
| | - Rubio R Punzalan
- Advanced Medical Analysis, LLC, 1941 Walker Ave, Monrovia, CA 91016, USA
| | - Leonard S Marks
- Department of Urology, David Geffen School of Medicine at UCLA, 10833 Le Conte Ave, Los Angeles, CA 90095-1738, USA
| | - Allan J Pantuck
- Department of Urology, David Geffen School of Medicine at UCLA, 10833 Le Conte Ave, Los Angeles, CA 90095-1738, USA
| | | | - Guomin Wang
- Department of Urology, Zhongshan Hospital of Fudan University, No.180 Fenglin Road, Shanghai 200032, China
| | - Hideki Mukouyama
- Department of Urology, Okinawa Nambu Tokushukai Hospital, 80 Hokama, Yaese-cho, Shimajiri-gun, Okinawa 901-0417, Japan
| | - Gang Zeng
- Department of Urology, David Geffen School of Medicine at UCLA, 10833 Le Conte Ave, Los Angeles, CA 90095-1738, USA
| |
Collapse
|
30
|
Park JH, Song MH, Lee CH, Lee MK, Park YM, Old L, Lee SY. Expression of the human cancer/testis antigen NY-SAR-35 is activated by CpG island hypomethylation. Biotechnol Lett 2011; 33:1085-91. [PMID: 21318630 DOI: 10.1007/s10529-011-0559-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2010] [Accepted: 01/31/2011] [Indexed: 11/28/2022]
Abstract
The novel cancer/testis antigen gene, NY-SAR-35, is expressed exclusively in normal testis and in various histological types of tumor. However, the NY-SAR-35 gene expression is observed to be aberrant in several cancer cell lines and tissues. The analysis of methylation status of the NY-SAR-35 gene promoter in various cancer cell lines showed that its expression was related to methylation of the promoter region. Treatment of human cancer cell lines with the demethylating agent 5-aza-2'-deoxycytidine activated the expression of the NY-SAR-35 gene. In addition, transfection experiments on various fragments of the CpG-rich gene promoter indicate that in vitro methylation of the NY-SAR-35 gene promoter results in the loss of promoter activity. The expression of NY-SAR-35 is therefore activated by hypomethylation of the CpG island in the gene promoter.
Collapse
Affiliation(s)
- Jin-Hyung Park
- Department of Biochemistry, School of Medicine, Pusan National University, Beomeori, Mulgeumeup, Yangsan, 626-770, Republic of Korea
| | | | | | | | | | | | | |
Collapse
|
31
|
Maricque BB, Eickhoff JC, McNeel DG. Antibody responses to prostate-associated antigens in patients with prostatitis and prostate cancer. Prostate 2011; 71:134-46. [PMID: 20632317 PMCID: PMC2978272 DOI: 10.1002/pros.21229] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
BACKGROUND An important focus of tumor immunotherapy has been the identification of appropriate antigenic targets. Serum-based screening approaches have led to the discovery of hundreds of tumor-associated antigens recognized by IgG. Our efforts to identify immunologically recognized proteins in prostate cancer have yielded a multitude of antigens; however, prioritizing these antigens as targets for evaluation in immunotherapies has been challenging. In this report, we set out to determine whether the evaluation of multiple antigenic targets would allow the identification of a subset of antigens that are common immunologic targets in patients with prostate cancer. METHODS Using a phage immunoblot approach, we evaluated IgG responses in patients with prostate cancer (n = 126), patients with chronic prostatitis (n = 45), and men without prostate disease (n = 53). RESULTS We found that patients with prostate cancer or prostatitis have IgG specific for multiple common antigens. A subset of 23 proteins was identified to which IgG were detected in 38% of patients with prostate cancer and 33% patients with prostatitis versus 6% of controls (P < 0.001 and P = 0.003, respectively). Responses to multiple members were not higher in patients with advanced disease, suggesting antibody immune responses occur early in the natural history of cancer progression. CONCLUSIONS These findings suggest an association between inflammatory conditions of the prostate and prostate cancer, and suggest that IgG responses to a panel of commonly recognized prostate antigens could be potentially used in the identification of patients at risk for prostate cancer or as a tool to identify immune responses elicited to prostate tissue.
Collapse
Affiliation(s)
- Brett B. Maricque
- University of Wisconsin Carbone Comprehensive Cancer Center, 1111 Highland Avenue, Madison, WI 53705
| | - Jens C. Eickhoff
- Colorado State University, Department of Statistics, Fort Collins, CO 80523
| | - Douglas G. McNeel
- University of Wisconsin Carbone Comprehensive Cancer Center, 1111 Highland Avenue, Madison, WI 53705
| |
Collapse
|
32
|
Moreno-Bost A, Szmania S, Stone K, Garg T, Hoerring A, Szymonifka J, Shaughnessy J, Barlogie B, Prentice HG, van Rhee F. Epigenetic modulation of MAGE-A3 antigen expression in multiple myeloma following treatment with the demethylation agent 5-azacitidine and the histone deacetlyase inhibitor MGCD0103. Cytotherapy 2010; 13:618-28. [PMID: 21171821 DOI: 10.3109/14653249.2010.529893] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
BACKGROUND AIMS Immunotherapy targeting MAGE-A3 in multiple myeloma (MM) could eradicate highly aggressive and proliferative clonal cell populations responsible for relapse. However, expression of many cancer-testis antigens, including MAGE-A3, can be heterogeneous, leading to the potential for tumor escape despite MAGE-A3-induced immunity. We hypothesized that a combination of the hypomethylating agent 5-azacitidine (5AC) and the histone deacetylase inhibitor (HDACi) MGCD0103 (MGC) could induce MAGE-A3 expression in MAGE-A3-negative MM, resulting in recognition and killing of MM cells by MAGE-A3-specific cytotoxic T lymphocytes (CTL). METHODS Gene expression analyses of MAGE-A3 expression in primary MM patient samples at diagnosis and relapse were completed to identify populations that would benefit from MAGE-A3 immunotherapy. MM cell lines were treated with 5AC and MGC. Real-time polymerase chain reaction (PCR) and Western blotting were performed to assess MAGE-A3 RNA and protein levels, respectively. Chromium-release assays and interferon (IFN) secretion assays were employed to ascertain MAGE-A3 CTL specificity against treated targets. RESULTS Gene expression analysis revealed that MAGE-A3 is expressed in MM patients at diagnosis (25%) and at relapse (49%). We observed de novo expression of MAGE-A3 RNA and protein in MAGE-A3-negative cell lines treated with 5AC. MGC treatment alone did not induce expression but sequential 5AC/MGC treatment led to enhanced expression and augmented recognition by MAGE-A3-specific CTL, as assessed by (51)Cr-release assays (P = 0.047) and enzyme-linked immunosorbent assay (ELISA) for IFN-γ secretion (P = 0.004). CONCLUSIONS MAGE-A3 is an attractive target for immunotherapy of MM and epigenetic modulation by 5AC, and MGC can induce MAGE-A3 expression and facilitate killing by MAGE-A3-specific CTL.
Collapse
Affiliation(s)
- Amberly Moreno-Bost
- Myeloma Institute for Research and Therapy, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
IgG responses to tissue-associated antigens as biomarkers of immunological treatment efficacy. J Biomed Biotechnol 2010; 2011:454861. [PMID: 21197272 PMCID: PMC3010827 DOI: 10.1155/2011/454861] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2010] [Accepted: 11/12/2010] [Indexed: 01/05/2023] Open
Abstract
We previously demonstrated that IgG responses to a panel of 126 prostate tissue-associated antigens are common in patients with prostate cancer. In the current report we questioned whether changes in IgG responses to this panel might be used as a measure of immune response, and potentially antigen spread, following prostate cancer-directed immune-active therapies. Sera were obtained from prostate cancer patients prior to and three months following treatment with androgen deprivation therapy (n = 34), a poxviral vaccine (n = 31), and a DNA vaccine (n = 21). Changes in IgG responses to individual antigens were identified by phage immunoblot. Patterns of IgG recognition following three months of treatment were evaluated using a machine-learned Bayesian Belief Network (ML-BBN). We found that different antigens were recognized following androgen deprivation compared with vaccine therapies. While the number of clinical responders was low in the vaccine-treated populations, we demonstrate that ML-BBN can be used to develop potentially predictive models.
Collapse
|
34
|
de Vrij J, Willemsen RA, Lindholm L, Hoeben RC, Bangma CH, Barber C, Behr JP, Briggs S, Carlisle R, Cheng WS, Dautzenberg IJC, de Ridder C, Dzojic H, Erbacher P, Essand M, Fisher K, Frazier A, Georgopoulos LJ, Jennings I, Kochanek S, Koppers-Lalic D, Kraaij R, Kreppel F, Magnusson M, Maitland N, Neuberg P, Nugent R, Ogris M, Remy JS, Scaife M, Schenk-Braat E, Schooten E, Seymour L, Slade M, Szyjanowicz P, Totterman T, Uil TG, Ulbrich K, van der Weel L, van Weerden W, Wagner E, Zuber G. Adenovirus-derived vectors for prostate cancer gene therapy. Hum Gene Ther 2010; 21:795-805. [PMID: 19947826 DOI: 10.1089/hum.2009.203] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Prostate cancer is a leading cause of death among men in Western countries. Whereas the survival rate approaches 100% for patients with localized cancer, the results of treatment in patients with metastasized prostate cancer at diagnosis are much less successful. The patients are usually presented with a variety of treatment options, but therapeutic interventions in prostate cancer are associated with frequent adverse side effects. Gene therapy and oncolytic virus therapy may constitute new strategies. Already a wide variety of preclinical studies has demonstrated the therapeutic potential of such approaches, with oncolytic prostate-specific adenoviruses as the most prominent vector. The state of the art and future prospects of gene therapy in prostate cancer are reviewed, with a focus on adenoviral vectors. We summarize advances in adenovirus technology for prostate cancer treatment and highlight areas where further developments are necessary.
Collapse
Affiliation(s)
- Jeroen de Vrij
- Department of Molecular Cell Biology, Leiden University Medical Center , 2300 RC Leiden, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
The SSX family of cancer-testis antigens as target proteins for tumor therapy. Clin Dev Immunol 2010; 2010:150591. [PMID: 20981248 PMCID: PMC2963798 DOI: 10.1155/2010/150591] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2010] [Accepted: 09/01/2010] [Indexed: 01/01/2023]
Abstract
Cancer-testis antigens (CTAs) represent an expanding class of tumor-associated proteins defined on the basis of their tissue-restricted expression to testis or ovary germline cells and frequent ectopic expression in tumor tissue. The expression of CTA in MHC class I-deficient germline cells makes these proteins particularly attractive as immunotherapeutic targets because they serve as essentially tumor-specific antigens for MHC class I-restricted CD8+ T cells. Moreover, because CTAs are expressed in many types of cancer, any therapeutic developed to target these antigens might have efficacy for multiple cancer types. Of particular interest among CTAs is the synovial sarcoma X chromosome breakpoint (SSX) family of proteins, which includes ten highly homologous family members. Expression of SSX proteins in tumor tissues has been associated with advanced stages of disease and worse patient prognosis. Additionally, both humoral and cell-mediated immune responses to SSX proteins have been demonstrated in patients with tumors of varying histological origin, which indicates that natural immune responses can be spontaneously generated to these antigens in cancer patients. The current review will describe the history and identification of this family of proteins, as well as what is known of their function, expression in normal and malignant tissues, and immunogenicity.
Collapse
|
36
|
Dubovsky JA, Wang D, Powers JJ, Berchmans E, Smith MA, Wright KL, Sotomayor EM, Pinilla-Ibarz JA. Restoring the functional immunogenicity of chronic lymphocytic leukemia using epigenetic modifiers. Leuk Res 2010; 35:394-404. [PMID: 20863567 DOI: 10.1016/j.leukres.2010.08.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2010] [Revised: 06/23/2010] [Accepted: 08/03/2010] [Indexed: 10/19/2022]
Abstract
Chronic lymphocytic leukemia (CLL) is a malignancy arising from immune cells (B-lymphocytes) endowed with intrinsic antigen-presenting capabilities. Such a function however is lost during malignant transformation and CLL cells are well known for their inability to process and present antigens to the T-cell arm of the immune system. Instead, malignant CLL cells elicit a vast array of immune regulatory mechanisms conducive to T-cell dysfunction and immunosuppression. Previously, we have shown that treatment of CLL cells with the demethylating agent 5-aza-2'-deoxycytidine unleashed target antigen expression. Here we show for the first time that combining two epigenetic modifiers, 5-aza-2'-deoxycytidine and the histone deacetylase inhibitor LAQ824 effectively restores the immunogenicity of CLL cell lines as well as primary cells obtained from CLL patients. Indeed, such a combination induces the expression of novel and highly antigenic cancer-testis antigens (CTAs) and costimulatory molecules. These changes facilitate the formation of robust supramolecular activation complexes (SMAC) between CLL cells and responder T-cells leading to intracellular signaling, lytic granule mobilization, and polarization of functional and relevant T-cell responses. This cascade of T-cell activating events triggered by CLL cells with restored APC function, points to combined epigenetic modifier treatment as a potential immunotherapeutic strategy for CLL patients.
Collapse
Affiliation(s)
- Jason A Dubovsky
- Department of Malignant Hematology, Immunology, and Experimental Therapeutics, H. Lee Moffitt Cancer Center, Tampa, FL, USA
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Expression of the Y-Encoded TSPY is Associated with Progression of Prostate Cancer. Genes (Basel) 2010; 1:283-93. [PMID: 24710046 PMCID: PMC3954091 DOI: 10.3390/genes1020283] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2010] [Revised: 09/01/2010] [Accepted: 09/02/2010] [Indexed: 01/20/2023] Open
Abstract
TSPY is a Y-encoded gene that is expressed in normal testicular germ cells and various cancer types including germ cell tumor, melanoma, hepatocellular carcinoma, and prostate cancer. Currently, the correlation between TSPY expression and oncogenic development has not been established, particularly in somatic cancers. To establish such correlation, we analyzed the expression of TSPY, in reference to its interactive oncoprotein, EEF1A, tumor biomarker, AMACR, and normal basal cell biomarker, p63, in 41 cases of clinical prostate cancers (CPCa), 17 cases of latent prostate cancers (LPCa), and 19 cases of non-cancerous prostate (control) by immunohistochemistry. Our results show that TSPY was detected more frequently (78%) in the clinical prostate cancer specimens than those of latent prostate cancer (47%) and control (50%). In the latent cancer group, the levels of TSPY expression could be correlated with increasing Gleason grades. TSPY expression was detected in seven out of nine high-grade latent cancer samples (Gleason 7 and more). The expression of the TSPY binding partner EEF1A was detectable in all prostate specimens, but the levels were higher in cancer cells in clinical and latent prostate cancer specimens than normal prostatic cells. These observations suggest that expressions of TSPY and its binding partner EEF1A are associated with the development and progression of prostate cancer.
Collapse
|
38
|
Cheever MA, Allison JP, Ferris AS, Finn OJ, Hastings BM, Hecht TT, Mellman I, Prindiville SA, Viner JL, Weiner LM, Matrisian LM. The prioritization of cancer antigens: a national cancer institute pilot project for the acceleration of translational research. Clin Cancer Res 2009; 15:5323-37. [PMID: 19723653 DOI: 10.1158/1078-0432.ccr-09-0737] [Citation(s) in RCA: 1047] [Impact Index Per Article: 65.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The purpose of the National Cancer Institute pilot project to prioritize cancer antigens was to develop a well-vetted, priority-ranked list of cancer vaccine target antigens based on predefined and preweighted objective criteria. An additional aim was for the National Cancer Institute to test a new approach for prioritizing translational research opportunities based on an analytic hierarchy process for dealing with complex decisions. Antigen prioritization involved developing a list of "ideal" cancer antigen criteria/characteristics, assigning relative weights to those criteria using pairwise comparisons, selecting 75 representative antigens for comparison and ranking, assembling information on the predefined criteria for the selected antigens, and ranking the antigens based on the predefined, preweighted criteria. Using the pairwise approach, the result of criteria weighting, in descending order, was as follows: (a) therapeutic function, (b) immunogenicity, (c) role of the antigen in oncogenicity, (d) specificity, (e) expression level and percent of antigen-positive cells, (f) stem cell expression, (g) number of patients with antigen-positive cancers, (h) number of antigenic epitopes, and (i) cellular location of antigen expression. None of the 75 antigens had all of the characteristics of the ideal cancer antigen. However, 46 were immunogenic in clinical trials and 20 of them had suggestive clinical efficacy in the "therapeutic function" category. These findings reflect the current status of the cancer vaccine field, highlight the possibility that additional organized efforts and funding would accelerate the development of therapeutically effective cancer vaccines, and accentuate the need for prioritization.
Collapse
Affiliation(s)
- Martin A Cheever
- Fred Hutchinson Cancer Research Center, Seattle, Washington, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Dubovsky JA, McNeel DG, Powers JJ, Gordon J, Sotomayor EM, Pinilla-Ibarz JA. Treatment of chronic lymphocytic leukemia with a hypomethylating agent induces expression of NXF2, an immunogenic cancer testis antigen. Clin Cancer Res 2009; 15:3406-15. [PMID: 19401350 DOI: 10.1158/1078-0432.ccr-08-2099] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Critical to the success of active immunotherapy against cancer is the identification of immunologically recognized cancer-specific proteins with low tolerogenic potential. Cancer testis antigens (CTA), in particular, fulfill this requirement as a result of their aberrant expression restricted to cancer cells and lack of expression in normal tissues bypassing tolerogenic mechanisms against self. Although CTAs have been extensively studied in solid malignancies, little is known regarding their expression in chronic lymphocytic leukemia (CLL). EXPERIMENTAL DESIGN Using a two-pronged approach we evaluated the immunogenicity of 29 CTAs in 22 patients with CLL and correlated these results to reverse transcriptase PCR data from CLL cell lines and patient cells. RESULTS We identified IgG-specific antibodies for one antigen, NXF2, and confirmed this response by ELISA and Western blot. We found that treatment of CLL with 5-aza-2'-deoxycytidine can induce expression of NXF2 that lasted for several weeks after treatment. Treatment also increased levels of MHC and costimulatory molecules (CD80, CD86, and CD40) necessary for antigen presentation. In addition, we identified other promising antigens that may have potential immunotherapeutic application. CONCLUSIONS Our findings suggest that NXF2 could be further pursued as an immunotherapeutic target in CLL, and that treatment with demethylating agents could be exploited to specifically modulate CTA expression and effective antigen presentation in malignant B cells.
Collapse
|