1
|
Shaikh U, Shah P, Jones V, Ramos-Rodriguez AJ, Sriharan A, Loo E, Khan WA, Simmons B, Cloutier JM. Subungual melanoma with cartilaginous differentiation: Molecular insights. J Cutan Pathol 2024; 51:576-582. [PMID: 38666479 DOI: 10.1111/cup.14630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 02/02/2024] [Accepted: 04/15/2024] [Indexed: 07/09/2024]
Abstract
Melanoma's rare capacity to undergo heterologous differentiation can create significant diagnostic challenges. The molecular mechanisms underlying this phenomenon are not well understood. We present an unusual case of subungual melanoma exhibiting extensive cartilaginous differentiation and provide insights into its molecular and cytogenomic features. Histopathologically, the tumor was predominantly composed of nodules of malignant cartilage in association with a smaller population of nested epithelioid to rhabdoid cells. Immunohistochemically, the tumor cells in both components were positive for S100, SOX10, and PRAME, and were negative for Melan-A and HMB-45. Molecular analysis by whole exome DNA sequence did not detect any pathogenic variants in genes commonly implicated in melanoma. Additional analysis by SNP chromosomal microarray revealed a complex genome characterized by numerous chromosomal losses and gains, including a homozygous deletion of the CDKN2A locus and a heterozygous deletion of the locus containing EXT2, a tumor suppressor implicated in hereditary multiple osteochondromas and secondary chondrosarcomas. This case underscores the importance of recognizing cartilaginous differentiation as a rare manifestation of melanoma, particularly at subungual sites, and suggests that at least some of these melanomas may be driven by non-canonical molecular pathways.
Collapse
Affiliation(s)
- Umayr Shaikh
- School of Medicine, Georgetown University, Washington, DC, USA
| | - Payal Shah
- Department of Dermatology, Dartmouth Health, Lebanon, New Hampshire, USA
| | - Victoria Jones
- Department of Pathology and Laboratory Medicine, One Medical Center Drive, Dartmouth Health, Dartmouth Hitchcock Medical Center, Lebanon, New Hampshire, USA
| | - Alvaro J Ramos-Rodriguez
- Department of Pathology and Laboratory Medicine, One Medical Center Drive, Dartmouth Health, Dartmouth Hitchcock Medical Center, Lebanon, New Hampshire, USA
| | - Aravindhan Sriharan
- Department of Pathology and Laboratory Medicine, One Medical Center Drive, Dartmouth Health, Dartmouth Hitchcock Medical Center, Lebanon, New Hampshire, USA
| | - Eric Loo
- Department of Pathology and Laboratory Medicine, One Medical Center Drive, Dartmouth Health, Dartmouth Hitchcock Medical Center, Lebanon, New Hampshire, USA
| | - Wahab A Khan
- Department of Pathology and Laboratory Medicine, One Medical Center Drive, Dartmouth Health, Dartmouth Hitchcock Medical Center, Lebanon, New Hampshire, USA
| | - Brian Simmons
- Department of Dermatology, Dartmouth Health, Lebanon, New Hampshire, USA
| | - Jeffrey M Cloutier
- Department of Pathology and Laboratory Medicine, One Medical Center Drive, Dartmouth Health, Dartmouth Hitchcock Medical Center, Lebanon, New Hampshire, USA
| |
Collapse
|
2
|
Gelmi MC, Gezgin G, van der Velden PA, Luyten GPM, Luk SJ, Heemskerk MHM, Jager MJ. PRAME Expression: A Target for Cancer Immunotherapy and a Prognostic Factor in Uveal Melanoma. Invest Ophthalmol Vis Sci 2023; 64:36. [PMID: 38149971 PMCID: PMC10755595 DOI: 10.1167/iovs.64.15.36] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 11/15/2023] [Indexed: 12/28/2023] Open
Abstract
Purpose Uveal melanoma (UM) is a rare disease with a high mortality, and new therapeutic options are being investigated. Preferentially Expressed Antigen in Melanoma (PRAME) is a cancer testis antigen, expressed in the testis, but also in cancers, including uveal melanoma. PRAME is considered a target for immune therapy in several cancers, and PRAME-specific T cell clones have been shown to kill UM cells. Methods We studied the literature on PRAME expression in hematological and solid malignancies, including UM, and its role as a target for immunotherapy. The distribution of tumor features was compared between PRAME-high and PRAME-low UM in a 64-patient cohort from the Leiden University Medical Center (LUMC) and in the Cancer Genome Atlas (TCGA) cohort of 80 cases and differential gene expression analysis was performed in the LUMC cohort. Results PRAME is expressed in many malignancies, it is frequently associated with a negative prognosis, and can be the target of T cell receptor (TCR)-transduced T cells, a promising treatment option with high avidity and safety. In UM, PRAME is expressed in 26% to 45% of cases and is correlated with a worse prognosis. In the LUMC and the TCGA cohorts, high PRAME expression was associated with larger diameter, higher Tumor-Node-Metastasis (TNM) stage, more frequent gain of chromosome 8q, and an inflammatory phenotype. Conclusions We confirm that PRAME is associated with poor prognosis in UM and has a strong connection with extra copies of 8q. We show that PRAME-specific immunotherapy in an adjuvant setting is promising in treatment of malignancies, including UM.
Collapse
Affiliation(s)
- Maria Chiara Gelmi
- Department of Ophthalmology, Leiden University Medical Center, Leiden, The Netherlands
| | - Gulçin Gezgin
- Department of Ophthalmology, Leiden University Medical Center, Leiden, The Netherlands
| | | | | | - Sietse J. Luk
- Department of Hematology, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Martine J. Jager
- Department of Ophthalmology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
3
|
van Amerongen RA, Tuit S, Wouters AK, van de Meent M, Siekman SL, Meeuwsen MH, Wachsmann TLA, Remst DFG, Hagedoorn RS, van der Steen DM, de Ru AH, Verdegaal EME, van Veelen PA, Falkenburg JHF, Heemskerk MHM. PRAME and CTCFL-reactive TCRs for the treatment of ovarian cancer. Front Immunol 2023; 14:1121973. [PMID: 37026005 PMCID: PMC10070997 DOI: 10.3389/fimmu.2023.1121973] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 02/06/2023] [Indexed: 04/08/2023] Open
Abstract
Recurrent disease emerges in the majority of patients with ovarian cancer (OVCA). Adoptive T-cell therapies with T-cell receptors (TCRs) targeting tumor-associated antigens (TAAs) are considered promising solutions for less-immunogenic 'cold' ovarian tumors. In order to treat a broader patient population, more TCRs targeting peptides derived from different TAAs binding in various HLA class I molecules are essential. By performing a differential gene expression analysis using mRNA-seq datasets, PRAME, CTCFL and CLDN6 were selected as strictly tumor-specific TAAs, with high expression in ovarian cancer and at least 20-fold lower expression in all healthy tissues of risk. In primary OVCA patient samples and cell lines we confirmed expression and identified naturally expressed TAA-derived peptides in the HLA class I ligandome. Subsequently, high-avidity T-cell clones recognizing these peptides were isolated from the allo-HLA T-cell repertoire of healthy individuals. Three PRAME TCRs and one CTCFL TCR of the most promising T-cell clones were sequenced, and transferred to CD8+ T cells. The PRAME TCR-T cells demonstrated potent and specific antitumor reactivity in vitro and in vivo. The CTCFL TCR-T cells efficiently recognized primary patient-derived OVCA cells, and OVCA cell lines treated with demethylating agent 5-aza-2'-deoxycytidine (DAC). The identified PRAME and CTCFL TCRs are promising candidates for the treatment of patients with ovarian cancer, and are an essential addition to the currently used HLA-A*02:01 restricted PRAME TCRs. Our selection of differentially expressed genes, naturally expressed TAA peptides and potent TCRs can improve and broaden the use of T-cell therapies for patients with ovarian cancer or other PRAME or CTCFL expressing cancers.
Collapse
Affiliation(s)
| | - Sander Tuit
- Department of Hematology, Leiden University Medical Center, Leiden, Netherlands
| | - Anne K. Wouters
- Department of Hematology, Leiden University Medical Center, Leiden, Netherlands
| | - Marian van de Meent
- Department of Hematology, Leiden University Medical Center, Leiden, Netherlands
| | - Sterre L. Siekman
- Department of Hematology, Leiden University Medical Center, Leiden, Netherlands
| | - Miranda H. Meeuwsen
- Department of Hematology, Leiden University Medical Center, Leiden, Netherlands
| | | | - Dennis F. G. Remst
- Department of Hematology, Leiden University Medical Center, Leiden, Netherlands
| | - Renate S. Hagedoorn
- Department of Hematology, Leiden University Medical Center, Leiden, Netherlands
| | | | - Arnoud H. de Ru
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, Netherlands
| | - Els M. E. Verdegaal
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden, Netherlands
| | - Peter A. van Veelen
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, Netherlands
| | | | - Mirjam H. M. Heemskerk
- Department of Hematology, Leiden University Medical Center, Leiden, Netherlands
- *Correspondence: Mirjam H. M. Heemskerk,
| |
Collapse
|
4
|
The generation and application of antigen-specific T cell therapies for cancer and viral-associated disease. Mol Ther 2022; 30:2130-2152. [PMID: 35149193 PMCID: PMC9171249 DOI: 10.1016/j.ymthe.2022.02.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 11/27/2021] [Accepted: 02/03/2022] [Indexed: 11/23/2022] Open
Abstract
Immunotherapy with antigen-specific T cells is a promising, targeted therapeutic option for patients with cancer as well as for immunocompromised patients with virus infections. In this review, we characterize and compare current manufacturing protocols for the generation of T cells specific to viral and non-viral tumor-associated antigens. Specifically, we discuss: (1) the different methodologies to expand virus-specific T cell and non-viral tumor-associated antigen-specific T cell products, (2) an overview of the immunological principles involved when developing such manufacturing protocols, and (3) proposed standardized methodologies for the generation of polyclonal, polyfunctional antigen-specific T cells irrespective of donor source. Ex vivo expanded cells have been safely administered to treat numerous patients with virus-associated malignancies, hematologic malignancies, and solid tumors. Hence, we have performed a comprehensive review of the clinical trial results evaluating the safety, feasibility, and efficacy of these products in the clinic. In summary, this review seeks to provide new insights regarding antigen-specific T cell technology to benefit a rapidly expanding T cell therapy field.
Collapse
|
5
|
Methylation-mediated silencing of protein kinase C zeta induces apoptosis avoidance through ATM/CHK2 inactivation in dedifferentiated chondrosarcoma. Br J Cancer 2022; 126:1289-1300. [PMID: 35017658 PMCID: PMC9042856 DOI: 10.1038/s41416-021-01695-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 12/10/2021] [Accepted: 12/23/2021] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Dedifferentiated chondrosarcoma (DDCS) is an aggressive bone tumour with a poor prognosis and no effective treatment. Because changes in DNA methylation play critical roles in DDCS, we explored the roles that DNA methylation plays in oncogenesis to potentially identify an effective epigenetic treatment. METHODS We identified genes downregulated in DDCS vs. conventional chondrosarcoma (CCS) due to DNA methylation using in silico analysis. The results were validated in DDCS clinical samples, and the molecular functions of the genes of interest were investigated in multiple chondrosarcoma cell lines (NDCS-1, SW1353, and OUMS-27). The therapeutic effect of decitabine, a DNA methyltransferase inhibitor, was evaluated in vitro and in vivo. RESULTS PRKCZ was specifically downregulated by DNA methylation in DDCS. Overexpression of PRKCZ decreased the proliferation of NDCS-1 and SW1353 cells. PRKCZ directly bound to and activated ATM, which was followed by phosphorylation of CHK2 and subsequent apoptosis. Decitabine increased PRKCZ expression through de-methylating the promoter region of PRKCZ, which activated the ATM/CHK2 pathway and inhibited cell proliferation by inducing apoptosis. CONCLUSIONS Increased DNA methylation and reduced expression of PRKCZ prevents apoptosis via inactivation of the ATM/CHK2 pathway in DDCS. Decitabine-induced expression of PRKCZ represents a promising therapy for DDCS.
Collapse
|
6
|
Micaily I, Roche M, Ibrahim MY, Martinez-Outschoorn U, Mallick AB. Metabolic Pathways and Targets in Chondrosarcoma. Front Oncol 2021; 11:772263. [PMID: 34938658 PMCID: PMC8685273 DOI: 10.3389/fonc.2021.772263] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 11/18/2021] [Indexed: 11/13/2022] Open
Abstract
Chondrosarcomas are the second most common primary bone malignancy. Chondrosarcomas are characterized by the production of cartilaginous matrix and are generally resistant to radiation and chemotherapy and the outcomes are overall poor. Hence, there is strong interest in determining mechanisms of cancer aggressiveness and therapeutic resistance in chondrosarcomas. There are metabolic alterations in chondrosarcoma that are linked to the epigenetic state and tumor microenvironment that drive treatment resistance. This review focuses on metabolic changes in chondrosarcoma, and the relationship between signaling via isocitrate dehydrogenase 1 and 2 (IDH1 and IDH2), hedgehog, PI3K-mTOR-AKT, and SRC, as well as histone acetylation and angiogenesis. Also, potential treatment strategies targeting metabolism will be discussed including potential synergy with immunotherapies.
Collapse
Affiliation(s)
- Ida Micaily
- Department of Medical Oncology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Megan Roche
- Department of Medical Oncology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Mohammad Y. Ibrahim
- Saint Francis Medical Center, Seton Hall University, Trenton, NJ, United States
| | | | - Atrayee Basu Mallick
- Department of Medical Oncology, Thomas Jefferson University, Philadelphia, PA, United States
| |
Collapse
|
7
|
Sheikh TN, Chen X, Xu X, McGuire JT, Ingham M, Lu C, Schwartz GK. Growth Inhibition and Induction of Innate Immune Signaling of Chondrosarcomas with Epigenetic Inhibitors. Mol Cancer Ther 2021; 20:2362-2371. [PMID: 34552007 PMCID: PMC8643315 DOI: 10.1158/1535-7163.mct-21-0066] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 06/16/2021] [Accepted: 09/10/2021] [Indexed: 11/16/2022]
Abstract
Chondrosarcomas are inherently resistant to chemotherapy and radiotherapy, pointing to an unmet need for new treatment options. Immune checkpoint inhibitors, which have shown remarkable promise in multiple solid cancer types, have limited efficacy in chondrosarcomas. Mutations in IDH1/2 genes, which result in progressive increases in DNA and histone methylation, are observed in 50% of conventional chondrosarcomas, suggesting that epigenetic dysregulation represents a potential barrier for tumor progression and target for therapeutic intervention. Here, we demonstrated that combined treatment of FDA-approved inhibitors of DNA methyltransferases (DNMTs) 5-aza-2'-deoxycytidine (5-aza), and histone deacetylases (HDACs) suberanilohydroxamic acid (SAHA) impaired the proliferation of chondrosarcoma cell lines in vitro and in xenograft studies. Transcriptomic analysis reveals that chondrosarcoma cells treated with 5-aza and SAHA markedly elevated the expression of IFN-stimulated genes including PD-L1, indicating that these epigenetic drugs induced a potent innate immune response. We demonstrated that 5-aza and SAHA resulted in both genomic and epigenomic instability, as shown by elevated DNA damage response and derepression of retrotransposons, respectively, which in turn activated pattern recognition receptors (PRRs) and the downstream IFN signaling pathways. Importantly, the cytotoxic effects of 5-aza and SAHA can be rescued by depletion of PRRs such as cGAS and MAVS, and potentiated by depletion of the RNA-editing enzyme ADAR1. Together, our results demonstrate preclinical activity of combined DNMT and HDAC inhibition against chondrosarcomas and suggest that targeted epigenetic therapies could represent a new therapeutic approach in the treatment of chondrosarcomas, and this is being tested in an ongoing clinical trial (NCT04340843).
Collapse
Affiliation(s)
- Tahir N Sheikh
- Department of Genetics and Development, Columbia University, New York, New York
- Division of Hematology/Oncology, Department of Medicine, Columbia University, New York, New York
| | - Xiao Chen
- Department of Genetics and Development, Columbia University, New York, New York
| | - Xinjing Xu
- Department of Genetics and Development, Columbia University, New York, New York
| | - John T McGuire
- Department of Genetics and Development, Columbia University, New York, New York
| | - Matthew Ingham
- Division of Hematology/Oncology, Department of Medicine, Columbia University, New York, New York
- Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York
| | - Chao Lu
- Department of Genetics and Development, Columbia University, New York, New York.
- Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York
| | - Gary K Schwartz
- Division of Hematology/Oncology, Department of Medicine, Columbia University, New York, New York.
- Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York
| |
Collapse
|
8
|
Clemente O, Ottaiano A, Di Lorenzo G, Bracigliano A, Lamia S, Cannella L, Pizzolorusso A, Di Marzo M, Santorsola M, De Chiara A, Fazioli F, Tafuto S. Is immunotherapy in the future of therapeutic management of sarcomas? J Transl Med 2021; 19:173. [PMID: 33902630 PMCID: PMC8077947 DOI: 10.1186/s12967-021-02829-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 04/12/2021] [Indexed: 12/19/2022] Open
Abstract
Sarcomas are rare, ubiquitous and heterogeneous tumors usually treated with surgery, chemotherapy, target therapy, and radiotherapy. However, 25-50% of patients experience local relapses and/or distant metastases after chemotherapy with an overall survival about 12-18 months. Recently, immuno-therapy has revolutionized the cancer treatments with initial indications for non-small cell lung cancer (NSCLC) and melanoma (immune-checkpoint inhibitors).Here, we provide a narrative review on the topic as well as a critical description of the currently available trials on immunotherapy treatments in patients with sarcoma. Given the promising results obtained with anti-PD-1 monoclonal antibodies (pembrolizumab and nivolumab) and CAR-T cells, we strongly believe that these new immunotherapeutic approaches, along with an innovative characterization of tumor genetics, will provide an exciting opportunity to ameliorate the therapeutic management of sarcomas.
Collapse
Affiliation(s)
- Ottavia Clemente
- Sarcomas and Rare Tumors Unit, Istituto Nazionale Tumori, IRCCS - Fondazione "G. Pascale", 80131, Naples, Italy
| | - Alessandro Ottaiano
- Division of Innovative Therapies, Istituto Nazionale Tumori, IRCCS - Fondazione "G. Pascale", 80131, Naples, Italy
| | - Giuseppe Di Lorenzo
- Sarcomas and Rare Tumors Unit, Istituto Nazionale Tumori, IRCCS - Fondazione "G. Pascale", 80131, Naples, Italy
| | - Alessandra Bracigliano
- Nuclear Medicine Unit, Istituto Nazionale Tumori, IRCCS - Fondazione "G. Pascale, 80131, Naples, Italy
| | - Sabrina Lamia
- Sarcomas and Rare Tumors Unit, Istituto Nazionale Tumori, IRCCS - Fondazione "G. Pascale", 80131, Naples, Italy
| | - Lucia Cannella
- Sarcomas and Rare Tumors Unit, Istituto Nazionale Tumori, IRCCS - Fondazione "G. Pascale", 80131, Naples, Italy
| | - Antonio Pizzolorusso
- Sarcomas and Rare Tumors Unit, Istituto Nazionale Tumori, IRCCS - Fondazione "G. Pascale", 80131, Naples, Italy
| | - Massimiliano Di Marzo
- Department of Abdominal Oncology, Istituto Nazionale Tumori, IRCCS - Fondazione "G. Pascale", 80131, Naples, Italy
| | - Mariachiara Santorsola
- Division of Innovative Therapies, Istituto Nazionale Tumori, IRCCS - Fondazione "G. Pascale", 80131, Naples, Italy
| | - Annarosaria De Chiara
- Histopathology of Lymphomas and Sarcomas SSD, Istituto Nazionale Tumori, IRCCS - Fondazione "G. Pascale", 80131, Naples, Italy
| | - Flavio Fazioli
- Orthopedic Oncology Unit, Istituto Nazionale Tumori, IRCCS - Fondazione "G. Pascale", 80131, Naples, Italy
| | - Salvatore Tafuto
- Sarcomas and Rare Tumors Unit, Istituto Nazionale Tumori, IRCCS - Fondazione "G. Pascale", 80131, Naples, Italy.
| |
Collapse
|
9
|
Lezcano C, Müller AM, Frosina D, Hernandez E, Geronimo JA, Busam KJ, Jungbluth AA. Immunohistochemical Detection of Cancer-Testis Antigen PRAME. Int J Surg Pathol 2021; 29:826-835. [PMID: 33890816 DOI: 10.1177/10668969211012085] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Cancer-testis (CT) antigens were identified by their ability to elicit T- or B-cell immune responses in the autologous host. They are typically expressed in a wide variety of neoplasms and in normal adult tissues are restricted to testicular germ cells. PReferentially expressed Antigen of Melanoma (PRAME) is a member of the family of nonclassical CT antigens being expressed in a few other normal tissues besides testis. Interestingly, knowledge about the protein expression of many CT antigens is still incomplete due to the limited availability of reagents for their immunohistochemical detection. Here, we tested several commercially available serological reagents and identified a monoclonal antibody suitable for the immunohistochemical detection of PRAME in formalin-fixed paraffin-embedded specimens. We also tested a wide array of normal and neoplastic tissues. PRAME protein expression in normal tissues is congruent with original molecular data being present in the testis, and at low levels in the endometrium, adrenal cortex, and adult as well as fetal ovary. In tumors, there is diffuse PRAME immunoreactivity in most metastatic melanomas, myxoid liposarcomas, and synovial sarcomas. Other neoplasms such as seminomas and carcinomas of various origins including endometrial, serous ovarian, mammary ductal, lung, and renal showed an intermediate proportion of cases and variable extent of tumor cells positive for PRAME protein expression. As seen with other CT antigens, hepatocellular and colorectal carcinoma, Leydig cell tumors, mesothelioma, and leiomyosarcoma are poor expressers of PRAME.
Collapse
Affiliation(s)
| | | | - Denise Frosina
- 5803Memorial Sloan-Kettering Cancer Center, New York, USA
| | | | | | - Klaus J Busam
- 5803Memorial Sloan-Kettering Cancer Center, New York, USA
| | | |
Collapse
|
10
|
Zając A, Król SK, Rutkowski P, Czarnecka AM. Biological Heterogeneity of Chondrosarcoma: From (Epi) Genetics through Stemness and Deregulated Signaling to Immunophenotype. Cancers (Basel) 2021; 13:1317. [PMID: 33804155 PMCID: PMC8001927 DOI: 10.3390/cancers13061317] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 03/05/2021] [Indexed: 12/11/2022] Open
Abstract
Chondrosarcoma (ChS) is a primary malignant bone tumor. Due to its heterogeneity in clinical outcomes and resistance to chemo- and radiotherapies, there is a need to develop new potential therapies and molecular targets of drugs. Many genes and pathways are involved in in ChS progression. The most frequently mutated genes are isocitrate dehydrogenase ½ (IDH1/2), collagen type II alpha 1 chain (COL2A1), and TP53. Besides the point mutations in ChS, chromosomal aberrations, such as 12q13 (MDM2) amplification, the loss of 9p21 (CDKN21/p16/INK4A and INK4A-p14ARF), and several gene fusions, commonly occurring in sarcomas, have been found. ChS involves the hypermethylation of histone H3 and the decreased methylation of some transcription factors. In ChS progression, changes in the phosphatidylinositol 3-kinase/protein kinase B/mammalian target of rapamycin (PI3K-AKT-mTOR) and hedgehog pathways are known to play a role in tumor growth and chondrocyte proliferation. Due to recent discoveries regarding the potential of immunotherapy in many cancers, in this review we summarize the current state of knowledge concerning cellular markers of ChS and tumor-associated immune cells. This review compares the latest discoveries in ChS biology from gene alterations to specific cellular markers, including advanced molecular pathways and tumor microenvironment, which can help in discovering new potential checkpoints in inhibitory therapy.
Collapse
Affiliation(s)
- Agnieszka Zając
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland; (A.Z.); (P.R.)
| | - Sylwia K. Król
- Department of Molecular and Translational Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland;
| | - Piotr Rutkowski
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland; (A.Z.); (P.R.)
| | - Anna M. Czarnecka
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland; (A.Z.); (P.R.)
- Department of Experimental Pharmacology, Mossakowski Medical Research Centre, Polish Academy of Sciences, 02-176 Warsaw, Poland
| |
Collapse
|
11
|
Nowicki TS, Farrell C, Morselli M, Rubbi L, Campbell KM, Macabali MH, Berent-Maoz B, Comin-Anduix B, Pellegrini M, Ribas A. Epigenetic Suppression of Transgenic T-cell Receptor Expression via Gamma-Retroviral Vector Methylation in Adoptive Cell Transfer Therapy. Cancer Discov 2020; 10:1645-1653. [PMID: 32699033 DOI: 10.1158/2159-8290.cd-20-0300] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 05/21/2020] [Accepted: 07/07/2020] [Indexed: 11/16/2022]
Abstract
Transgenic T-cell receptor (TCR) adoptive cell therapies recognizing tumor antigens are associated with robust initial response rates, but frequent disease relapse. This usually occurs in the setting of poor long-term persistence of cells expressing the transgenic TCR, generated using murine stem cell virus (MSCV) γ-retroviral vectors. Analysis of clinical transgenic adoptive cell therapy products in vivo revealed that despite strong persistence of the transgenic TCR DNA sequence over time, its expression was profoundly decreased over time at the RNA and protein levels. Patients with the greatest degrees of expression suppression displayed significant increases in DNA methylation over time within the MSCV promoter region, as well as progressive increases in DNA methylation within the entire MSCV vector over time. These increases in vector methylation occurred independently of its integration site within the host genomes. These results have significant implications for the design of future viral vector gene-engineered adoptive cell transfer therapies. SIGNIFICANCE: Cellular immunotherapies' reliance on retroviral vectors encoding foreign genetic material can be vulnerable to progressive acquisition of DNA methylation and subsequent epigenetic suppression of the transgenic product in TCR adoptive cell therapy. This must be considered in the design of future generations of cellular immunotherapies for cancer.This article is highlighted in the In This Issue feature, p. 1611.
Collapse
Affiliation(s)
- Theodore S Nowicki
- Division of Pediatric Hematology-Oncology, Department of Pediatrics, University of California, Los Angeles, Los Angeles, California. .,Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, California.,Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, California
| | - Colin Farrell
- Department of Human Genetics, University of California, Los Angeles, Los Angeles, California
| | - Marco Morselli
- Institute for Quantitative and Computational Biosciences - The Collaboratory, University of California, Los Angeles, Los Angeles, California.,Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, Los Angeles, California
| | - Liudmilla Rubbi
- Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, Los Angeles, California
| | - Katie M Campbell
- Division of Hematology-Oncology, Department of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Mignonette H Macabali
- Division of Hematology-Oncology, Department of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Beata Berent-Maoz
- Division of Hematology-Oncology, Department of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Begoña Comin-Anduix
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, California.,Division of Surgical Oncology, Department of Surgery, University of California, Los Angeles, Los Angeles, California
| | - Matteo Pellegrini
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, California.,Institute for Quantitative and Computational Biosciences - The Collaboratory, University of California, Los Angeles, Los Angeles, California.,Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, Los Angeles, California
| | - Antoni Ribas
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, California.,Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, California.,Division of Hematology-Oncology, Department of Medicine, University of California, Los Angeles, Los Angeles, California.,Division of Surgical Oncology, Department of Surgery, University of California, Los Angeles, Los Angeles, California.,Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, California
| |
Collapse
|
12
|
Maryamchik E, Gallagher KME, Preffer FI, Kadauke S, Maus MV. New directions in chimeric antigen receptor T cell [CAR-T] therapy and related flow cytometry. CYTOMETRY PART B-CLINICAL CYTOMETRY 2020; 98:299-327. [PMID: 32352629 DOI: 10.1002/cyto.b.21880] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 04/01/2020] [Accepted: 04/07/2020] [Indexed: 12/12/2022]
Abstract
Chimeric antigen receptor (CAR) T cells provide a promising approach to the treatment of hematologic malignancies and solid tumors. Flow cytometry is a powerful analytical modality, which plays an expanding role in all stages of CAR T therapy, from lymphocyte collection, to CAR T cell manufacturing, to in vivo monitoring of the infused cells and evaluation of their function in the tumor environment. Therefore, a thorough understanding of the new directions is important for designing and implementing CAR T-related flow cytometry assays in the clinical and investigational settings. However, the speed of new discoveries and the multitude of clinical and preclinical trials make it challenging to keep up to date in this complex field. In this review, we summarize the current state of CAR T therapy, highlight the areas of emergent research, discuss applications of flow cytometry in modern cell therapy, and touch upon several considerations particular to CAR detection and assessing the effectiveness of CAR T therapy.
Collapse
Affiliation(s)
- Elena Maryamchik
- Department of Pathology and Laboratory Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | | | - Frederic I Preffer
- Clinical Cytometry, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Stephan Kadauke
- Department of Pathology and Laboratory Medicine, Cell and Gene Therapy Laboratory, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Marcela V Maus
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Cellular Immunotherapy Program, Department of Medicine, Boston, Massachusetts, USA
| |
Collapse
|
13
|
Heymann MF, Schiavone K, Heymann D. Bone sarcomas in the immunotherapy era. Br J Pharmacol 2020; 178:1955-1972. [PMID: 31975481 DOI: 10.1111/bph.14999] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 12/23/2019] [Accepted: 01/07/2020] [Indexed: 11/30/2022] Open
Abstract
Bone sarcomas are primary bone tumours found mainly in children and adolescents, as osteosarcoma and Ewing's sarcoma, and in adults in their 40s as chondrosarcoma. The last four decades the development of therapeutic approaches was based on drug combinations have shown no real improvement in overall survival. Recently oncoimmunology has allowed a better understand of the crucial role played by the immune system in the oncologic process. This led to clinical trials with the aim of reprogramming the immune system to facilitate cancer cell recognition. Immune infiltrates of bone sarcomas have been characterized and their molecular profiling identified as immune therapeutic targets. Unfortunately, the clinical responses in trials remain anecdotal but highlight the necessity to improve the characterization of tumour micro-environment to unlock the immunotherapeutic response, especially in their paediatric forms. Bone sarcomas have entered the immunotherapy era and here we overview the recent developments in immunotherapies in these sarcomas. LINKED ARTICLES: This article is part of a themed issue on The molecular pharmacology of bone and cancer-related bone diseases. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v178.9/issuetoc.
Collapse
Affiliation(s)
- Marie-Françoise Heymann
- Université de Nantes, INSERM, CRCINA, Institut de Cancérologie de l'Ouest, Saint-Herblain, France.,"Tumor Heterogeneity and Precision Medicine", Institut de Cancérologie de l'Ouest, Saint Herblain, France.,INSERM, European Associated Laboratory "Sarcoma Research Unit", Department of Oncology and Metabolism, Medical School, University of Sheffield, Sheffield, UK
| | - Kristina Schiavone
- Université de Nantes, INSERM, CRCINA, Institut de Cancérologie de l'Ouest, Saint-Herblain, France.,"Tumor Heterogeneity and Precision Medicine", Institut de Cancérologie de l'Ouest, Saint Herblain, France
| | - Dominique Heymann
- Université de Nantes, INSERM, CRCINA, Institut de Cancérologie de l'Ouest, Saint-Herblain, France.,"Tumor Heterogeneity and Precision Medicine", Institut de Cancérologie de l'Ouest, Saint Herblain, France.,INSERM, European Associated Laboratory "Sarcoma Research Unit", Department of Oncology and Metabolism, Medical School, University of Sheffield, Sheffield, UK
| |
Collapse
|
14
|
Xu Y, Zou R, Wang J, Wang ZW, Zhu X. The role of the cancer testis antigen PRAME in tumorigenesis and immunotherapy in human cancer. Cell Prolif 2020; 53:e12770. [PMID: 32022332 PMCID: PMC7106952 DOI: 10.1111/cpr.12770] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 01/01/2020] [Accepted: 01/15/2020] [Indexed: 12/24/2022] Open
Abstract
Preferentially expressed antigen in melanoma (PRAME), which belongs to the cancer/testis antigen (CTA) gene family, plays a pivotal role in multiple cellular processes and immunotherapy response in human cancers. PRAME is highly expressed in different types of cancers and is involved in cell proliferation, apoptosis, differentiation and metastasis as well as the outcomes of patients with cancer. In this review article, we discuss the potential roles and physiological functions of PRAME in various types of cancers. Moreover, this review highlights immunotherapeutic strategies that target PRAME in human malignancies. Therefore, the modulation of PRAME might be useful for the treatment of patients with cancer.
Collapse
Affiliation(s)
- Yichi Xu
- Departmant of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Ruanmin Zou
- Departmant of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jing Wang
- Departmant of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zhi-Wei Wang
- Departmant of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Xueqiong Zhu
- Departmant of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
15
|
Abstract
AbstractChondrosarcomas are rare cancers of bone that arise from the malignant transformation of cells of chondrocytic lineage. They are known to be resistant to systemic cytotoxic chemotherapy and radiotherapy. The mainstay of management of localised disease is en bloc surgical resection with curative intent. Metastatic chondrosarcoma has a dismal prognosis, and to date, there are no proven effective systemic therapies in the advanced setting. Genomic studies have demonstrated that 50 to 80% of chondrosarcomas harbour a mutation in either the IDH1 or IDH2 gene. IDH inhibitors are currently under investigation in clinical trials, after showing promising results in phase 1 studies in IDH mutated cancers. In chondrosarcoma, IDH mutations represent an attractive target, however, early results with IDH inhibitors in IDH mutated chondrosarcoma are modest and the final results of ongoing trials are eagerly awaited.
Collapse
|
16
|
Abstract
Components of the tumor microenvironment (TME) are known to play an essential role during malignant progression, but often in a context-dependent manner. In bone and soft tissue sarcomas, disease-regulatory activities in the TME remain largely uncharacterized. This chapter introduces the cellular, structural, and chemical composition of the sarcoma TME from a pathobiological and therapeutic perspective.Sarcomas are malignant tumors with diverse features when it comes to primary tumor appearance, metastatic potential, and response to treatment. Many of the classic subtypes are mainly composed of malignant cells and are therefore assumed to be committed to autocrine signaling. Some of the tumors are infiltrated by immune cells and contain necrotic areas or excessive amounts of extracellular matrix (ECM) that regulates tissue stiffness and interstitial fluid pressure. Vascular invasion and blood vessel characteristics can in some instances be considered in the prognostic setting.Further insights into the disease-regulatory activities of the sarcoma TME will provide essential knowledge on how to develop successful combination treatments targeting not only malignant cells, but also their routes of nutrition and ability to shield themselves toward existing therapy.
Collapse
|
17
|
Wei R, Dean DC, Thanindratarn P, Hornicek FJ, Guo W, Duan Z. Cancer testis antigens in sarcoma: Expression, function and immunotherapeutic application. Cancer Lett 2019; 479:54-60. [PMID: 31634526 DOI: 10.1016/j.canlet.2019.10.024] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 09/23/2019] [Accepted: 10/15/2019] [Indexed: 02/06/2023]
Abstract
Sarcomas are a group of heterogeneous malignancies of mesenchymal origin. Patient outcomes remain especially grim for those with recurrent or metastatic disease, and current therapeutic strategies have not significantly improved outcomes over the past few decades. This has led to a number of studies assessing novel therapies. Cancer testis antigens (CTAs) are tumor-associated antigens with physiologic expression in the testis and various malignancies, including sarcomas. Genes encoding CTAs include MAGE, NY-ESO-1, PRAME, TRAG-3/CSAGE, and SSX. The importance and function of CTAs in tumorigenesis have gained recognition in recent years. They are also proving as robust diagnostic and prognostic biomarkers. Therapeutically, antigens derived from CTAs are highly recognizable by T lymphocytes and therefore capable of generating a potent antitumor immune response. CTAs are, therefore, promising targets for novel immunotherapies. Here we review the emerging works on expression, function, and immunotherapeutic application of CTAs in sarcoma therapy.
Collapse
Affiliation(s)
- Ran Wei
- Department of Orthopedic Surgery, Sarcoma Biology Laboratory, David Geffen School of Medicine at UCLA, 615 Charles E. Young Dr. South, Los Angeles, CA, 90095, USA; Musculoskeletal Tumor Center, Peking University People's Hospital, No. 11 Xizhimen South Street, Xicheng District, Beijing, 100044, China.
| | - Dylan C Dean
- Department of Orthopedic Surgery, Sarcoma Biology Laboratory, David Geffen School of Medicine at UCLA, 615 Charles E. Young Dr. South, Los Angeles, CA, 90095, USA.
| | - Pichaya Thanindratarn
- Department of Orthopedic Surgery, Sarcoma Biology Laboratory, David Geffen School of Medicine at UCLA, 615 Charles E. Young Dr. South, Los Angeles, CA, 90095, USA.
| | - Francis J Hornicek
- Department of Orthopedic Surgery, Sarcoma Biology Laboratory, David Geffen School of Medicine at UCLA, 615 Charles E. Young Dr. South, Los Angeles, CA, 90095, USA.
| | - Wei Guo
- Musculoskeletal Tumor Center, Peking University People's Hospital, No. 11 Xizhimen South Street, Xicheng District, Beijing, 100044, China.
| | - Zhenfeng Duan
- Department of Orthopedic Surgery, Sarcoma Biology Laboratory, David Geffen School of Medicine at UCLA, 615 Charles E. Young Dr. South, Los Angeles, CA, 90095, USA.
| |
Collapse
|
18
|
Chew GL, Campbell AE, De Neef E, Sutliff NA, Shadle SC, Tapscott SJ, Bradley RK. DUX4 Suppresses MHC Class I to Promote Cancer Immune Evasion and Resistance to Checkpoint Blockade. Dev Cell 2019; 50:658-671.e7. [PMID: 31327741 PMCID: PMC6736738 DOI: 10.1016/j.devcel.2019.06.011] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 04/22/2019] [Accepted: 06/14/2019] [Indexed: 12/22/2022]
Abstract
Advances in cancer immunotherapies make it critical to identify genes that modulate antigen presentation and tumor-immune interactions. We report that DUX4, an early embryonic transcription factor that is normally silenced in somatic tissues, is re-expressed in diverse solid cancers. Both cis-acting inherited genetic variation and somatically acquired mutations in trans-acting repressors contribute to DUX4 re-expression in cancer. Although many DUX4 target genes encode self-antigens, DUX4-expressing cancers were paradoxically characterized by reduced markers of anti-tumor cytolytic activity and lower major histocompatibility complex (MHC) class I gene expression. We demonstrate that DUX4 expression blocks interferon-γ-mediated induction of MHC class I, implicating suppressed antigen presentation in DUX4-mediated immune evasion. Clinical data in metastatic melanoma confirmed that DUX4 expression was associated with significantly reduced progression-free and overall survival in response to anti-CTLA-4. Our results demonstrate that cancers can escape immune surveillance by reactivating a normal developmental pathway and identify a therapeutically relevant mechanism of cell-intrinsic immune evasion.
Collapse
Affiliation(s)
- Guo-Liang Chew
- Computational Biology Program, Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; Basic Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Amy E Campbell
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Emma De Neef
- Computational Biology Program, Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; Basic Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA
| | - Nicholas A Sutliff
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Sean C Shadle
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; Molecular and Cellular Biology Program, University of Washington, Seattle, WA 98195, USA
| | - Stephen J Tapscott
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; Department of Neurology, University of Washington, Seattle, WA 98195, USA.
| | - Robert K Bradley
- Computational Biology Program, Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; Basic Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA.
| |
Collapse
|
19
|
Kreuzinger C, von der Decken I, Wolf A, Gamperl M, Koller J, Karacs J, Pfaffinger S, Bartl T, Reinthaller A, Grimm C, Singer CF, Braicu EI, Cunnea P, Gourley C, Smeets D, Boeckx B, Lambrechts D, Perco P, Horvat R, Berns EMJJ, Cacsire Castillo-Tong D. Patient-derived cell line models revealed therapeutic targets and molecular mechanisms underlying disease progression of high grade serous ovarian cancer. Cancer Lett 2019; 459:1-12. [PMID: 31150822 DOI: 10.1016/j.canlet.2019.05.032] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 05/24/2019] [Accepted: 05/24/2019] [Indexed: 01/07/2023]
Abstract
High grade serous ovarian cancer (HGSOC) is the most frequent type of ovarian cancer. Most patients have primary response to platinum-based chemotherapy but frequently relapse, which leads to patient death. A lack of well documented and characterized patient-derived HGSOC cell lines is so far a major barrier to define tumor specific therapeutic targets and to study the molecular mechanisms underlying disease progression. We established 34 patient-derived HGSOC cell lines and characterized them at cellular and molecular level. Particularly, we demonstrated that a cancer-testis antigen PRAME and Estrogen Receptor could serve as therapeutic targets. Notably, data from the cell lines did not demonstrate acquired resistance due to tumor recurrence that matched with clinical observations. Finally, we presented that all HGSOC had no or very low CDKN1A (p21) expression due to loss of wild-type TP53, suggesting that loss of cell cycle control is the determinant for tumorigenesis and progression. In conclusion, patient-derived cell lines reveal that PRAME is a potential tumor specific therapeutic target in HGSOC and counteracting the down-regulation of p21 caused by loss of wild-type TP53 might be the key to impede disease progression.
Collapse
Affiliation(s)
- Caroline Kreuzinger
- Translational Gynecology Group, Department of Obstetrics and Gynecology, Comprehensive Cancer Center, Medical University of Vienna, 1090, Vienna, Austria
| | - Isabel von der Decken
- Translational Gynecology Group, Department of Obstetrics and Gynecology, Comprehensive Cancer Center, Medical University of Vienna, 1090, Vienna, Austria
| | - Andrea Wolf
- Translational Gynecology Group, Department of Obstetrics and Gynecology, Comprehensive Cancer Center, Medical University of Vienna, 1090, Vienna, Austria
| | - Magdalena Gamperl
- Translational Gynecology Group, Department of Obstetrics and Gynecology, Comprehensive Cancer Center, Medical University of Vienna, 1090, Vienna, Austria
| | - Julia Koller
- Translational Gynecology Group, Department of Obstetrics and Gynecology, Comprehensive Cancer Center, Medical University of Vienna, 1090, Vienna, Austria
| | - Jasmine Karacs
- Translational Gynecology Group, Department of Obstetrics and Gynecology, Comprehensive Cancer Center, Medical University of Vienna, 1090, Vienna, Austria
| | - Stephanie Pfaffinger
- Translational Gynecology Group, Department of Obstetrics and Gynecology, Comprehensive Cancer Center, Medical University of Vienna, 1090, Vienna, Austria
| | - Thomas Bartl
- Translational Gynecology Group, Department of Obstetrics and Gynecology, Comprehensive Cancer Center, Medical University of Vienna, 1090, Vienna, Austria
| | - Alexander Reinthaller
- Translational Gynecology Group, Department of Obstetrics and Gynecology, Comprehensive Cancer Center, Medical University of Vienna, 1090, Vienna, Austria
| | - Christoph Grimm
- Translational Gynecology Group, Department of Obstetrics and Gynecology, Comprehensive Cancer Center, Medical University of Vienna, 1090, Vienna, Austria
| | - Christian F Singer
- Department of Gynecology and Gynecologic Oncology, Gynecologic Cancer Unit, Comprehensive Cancer Center, Medical University of Vienna, 1090, Vienna, Austria
| | - Elena Ioana Braicu
- Tumor Bank Ovarian Cancer Network, Department of Gynecology, Charité Universitätsmedizin Berlin, 13353, Berlin, Germany; Department of Gynecology, Charité Universitätsmedizin Berlin, 13353, Berlin, Germany
| | - Paula Cunnea
- Ovarian Cancer Action Research Centre, Department of Surgery and Cancer, Imperial College London, London, W12 0HS, United Kingdom
| | - Charlie Gourley
- Nicola Murray Centre for Ovarian Cancer Research, Cancer Research UK Edinburgh Centre, MRC IGMM, University of Edinburgh, Western General Hospital, Edinburgh, EH4 2XR, United Kingdom
| | - Dominiek Smeets
- KU Leuven, Department of Human Genetics, Laboratory for Translational Genetics, 3000, Leuven, Belgium; VIB, VIB Center for Cancer Biology, Laboratory for Translational Genetics, 3000, Leuven, Belgium
| | - Bram Boeckx
- KU Leuven, Department of Human Genetics, Laboratory for Translational Genetics, 3000, Leuven, Belgium; VIB, VIB Center for Cancer Biology, Laboratory for Translational Genetics, 3000, Leuven, Belgium
| | - Diether Lambrechts
- KU Leuven, Department of Human Genetics, Laboratory for Translational Genetics, 3000, Leuven, Belgium; VIB, VIB Center for Cancer Biology, Laboratory for Translational Genetics, 3000, Leuven, Belgium
| | - Paul Perco
- Emergentec Biodevelopment GmbH, 1180, Vienna, Austria
| | - Reinhard Horvat
- Department of Clinical Pathology, Medical University of Vienna, 1090, Vienna, Austria
| | - Els M J J Berns
- Department of Medical Oncology, Erasmus MC Cancer Institute, 3000 CA, Rotterdam, the Netherlands
| | - Dan Cacsire Castillo-Tong
- Translational Gynecology Group, Department of Obstetrics and Gynecology, Comprehensive Cancer Center, Medical University of Vienna, 1090, Vienna, Austria.
| |
Collapse
|
20
|
Pender A, Jones RL, Pollack S. Optimising Cancer Vaccine Design in Sarcoma. Cancers (Basel) 2018; 11:E1. [PMID: 30577459 PMCID: PMC6356514 DOI: 10.3390/cancers11010001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 12/15/2018] [Accepted: 12/18/2018] [Indexed: 12/14/2022] Open
Abstract
Immunotherapeutics are increasingly recognized as a key tool in the armamentarium against malignancy. The success of immune checkpoint-targeting drugs and adoptive cell therapy has refocused attention on the potential anti-cancer effect of eliciting a tumour-specific immunological response. Sarcomas are a rare and diverse group of tumours with a limited prognosis in advanced disease despite systemic therapeutics. Various vaccine strategies including peptide vaccines against cancer testis antigens, dendritic cell vaccines, and viral vectors have been trialled in sarcoma with growing evidence of efficacy. Here, we review the principles of successful vaccine development and how these have been applied thus far to the treatment of sarcoma.
Collapse
Affiliation(s)
- Alexandra Pender
- Sarcoma Unit, Department of Medicine, The Royal Marsden NHS Foundation Trust, London SW3 6JJ, UK.
| | - Robin L Jones
- Sarcoma Unit, Department of Medicine, The Royal Marsden NHS Foundation Trust, London SW3 6JJ, UK.
- Division of Clinical Studies, The Institute of Cancer Research, London SW3 6JB, UK.
| | - Seth Pollack
- Fred Hutchinson Cancer Research Center, University of Washington, Seattle, WA 98109, USA.
- Division of Oncology, University of Washington, Seattle, WA 98195, USA.
| |
Collapse
|
21
|
Wagner MJ, Ricciotti RW, Mantilla J, Loggers ET, Pollack SM, Cranmer LD. Response to PD1 inhibition in conventional chondrosarcoma. J Immunother Cancer 2018; 6:94. [PMID: 30253794 PMCID: PMC6156853 DOI: 10.1186/s40425-018-0413-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Accepted: 09/18/2018] [Indexed: 12/31/2022] Open
Abstract
Background Chondrosarcoma is one of the most common malignant bone tumors in adults. Conventional chondrosarcoma represents around 85% of all chondrosarcomas and is notoriously difficult to treat with chemotherapy. Case presentation We describe a 67-year-old man with metastatic conventional chondrosarcoma who was treated with nivolumab. Treatment was discontinued after restaging showed increased tumor burden, which later proved to be pseudoprogression. The patient restarted nivolumab and continues to have a near complete response. Conclusion Conventional chondrosarcoma may be sensitive to checkpoint inhibitors. Further, this case demonstrates clearly the phenomenon of pseudo-progression in this disease, a factor that must be considered in the design of clinical trials and clinical care. This case supports additional study of immunomodulatory agents in this deadly disease.
Collapse
Affiliation(s)
- Michael J Wagner
- Division of Medical Oncology, University of Washington School of Medicine, 825 Eastlake Avenue E, Seattle, WA, 98109, USA. .,Clinical Research Division, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave N, Seattle, WA, 98109, USA.
| | - Robert W Ricciotti
- Department of Pathology, University of Washington School of Medicine, 1959 NE Pacific St, Seattle, WA, 98195, USA
| | - Jose Mantilla
- Department of Pathology, University of Washington School of Medicine, 1959 NE Pacific St, Seattle, WA, 98195, USA
| | - Elizabeth T Loggers
- Division of Medical Oncology, University of Washington School of Medicine, 825 Eastlake Avenue E, Seattle, WA, 98109, USA.,Clinical Research Division, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave N, Seattle, WA, 98109, USA
| | - Seth M Pollack
- Division of Medical Oncology, University of Washington School of Medicine, 825 Eastlake Avenue E, Seattle, WA, 98109, USA.,Clinical Research Division, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave N, Seattle, WA, 98109, USA
| | - Lee D Cranmer
- Division of Medical Oncology, University of Washington School of Medicine, 825 Eastlake Avenue E, Seattle, WA, 98109, USA. .,Clinical Research Division, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave N, Seattle, WA, 98109, USA.
| |
Collapse
|
22
|
Mesiano G, Grignani G, Fiorino E, Leuci V, Rotolo R, D'Ambrosio L, Salfi C, Gammaitoni L, Giraudo L, Pisacane A, Butera S, Pignochino Y, Basiricó M, Capozzi F, Sapino A, Aglietta M, Sangiolo D. Cytokine Induced Killer cells are effective against sarcoma cancer stem cells spared by chemotherapy and target therapy. Oncoimmunology 2018; 7:e1465161. [PMID: 30393581 PMCID: PMC6208452 DOI: 10.1080/2162402x.2018.1465161] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Revised: 03/24/2018] [Accepted: 04/10/2018] [Indexed: 12/11/2022] Open
Abstract
Metastatic bone and soft tissue sarcomas often relapse after chemotherapy (CHT) and molecular targeted therapy (mTT), maintaining a severe prognosis. A subset of sarcoma cancer stem cells (sCSC) is hypothesized to resist conventional drugs and sustain disease relapses. We investigated the immunotherapy activity of cytokine induced killer cells (CIK) against autologous sCSC that survived CHT and mTT. The experimental platform included two aggressive bone and soft tissue sarcoma models: osteosarcoma (OS) and undifferentiated-pleomorphic sarcoma (UPS). To visualize putative sCSC we engineered patient-derived sarcoma cultures (2 OS and 3 UPS) with a lentiviral sCSC-detector wherein the promoter of stem-gene Oct4 controls the expression of eGFP. We visualized a fraction of sCSC (mean 24.2 ± 5.2%) and confirmed their tumorigenicity in vivo. sCSC resulted relatively resistant to both CHT and mTT in vitro. Therapeutic doses of doxorubicin significantly enriched viable eGFP+sCSC in both OS (2.6 fold, n = 16) and UPS (2.3 fold, n = 29) compared to untreated controls. Treatment with sorafenib (for OS) and pazopanib (for UPS) also determined enrichment (1.3 fold) of viable eGFP+sCSC, even if less intense than what observed after CHT. Sarcoma cells surviving CHT and mTT were efficiently killed in vitro by autologous CIK even at minimal effector/target ratios (40:1 = 82%, 1:4 = 29%, n = 13). CIK immunotherapy did not spare sCSC that were killed as efficiently as whole sarcoma cell population. The relative chemo-resistance of sCSC and sensitivity to CIK immunotherapy was confirmed in vivo. Our findings support CIK as an innovative, clinically explorable, approach to eradicate chemo-resistant sCSC implicated in tumor relapse.
Collapse
Affiliation(s)
- Giulia Mesiano
- Medical Oncology Division, Candiolo Cancer Institute, FPO-IRCCS. Str. Prov. 142, km 3.95, I-10060, Candiolo (To), Italy
| | - Giovanni Grignani
- Medical Oncology Division, Candiolo Cancer Institute, FPO-IRCCS. Str. Prov. 142, km 3.95, I-10060, Candiolo (To), Italy
| | - Erika Fiorino
- Department of Oncology, University of Torino, Candiolo (Torino) Italy
| | - Valeria Leuci
- Medical Oncology Division, Candiolo Cancer Institute, FPO-IRCCS. Str. Prov. 142, km 3.95, I-10060, Candiolo (To), Italy.,Department of Oncology, University of Torino, Candiolo (Torino) Italy
| | - Ramona Rotolo
- Department of Oncology, University of Torino, Candiolo (Torino) Italy
| | - Lorenzo D'Ambrosio
- Medical Oncology Division, Candiolo Cancer Institute, FPO-IRCCS. Str. Prov. 142, km 3.95, I-10060, Candiolo (To), Italy.,Department of Oncology, University of Torino, Candiolo (Torino) Italy
| | - Chiara Salfi
- Department of Oncology, University of Torino, Candiolo (Torino) Italy
| | - Loretta Gammaitoni
- Medical Oncology Division, Candiolo Cancer Institute, FPO-IRCCS. Str. Prov. 142, km 3.95, I-10060, Candiolo (To), Italy
| | - Lidia Giraudo
- Medical Oncology Division, Candiolo Cancer Institute, FPO-IRCCS. Str. Prov. 142, km 3.95, I-10060, Candiolo (To), Italy
| | - Alberto Pisacane
- Pathology Division, Candiolo Cancer Institute, FPO-IRCCS, Str. Prov. 142, km 3.95, I-10060, Candiolo (To), Italy
| | - Sara Butera
- Department of Molecular Biotechnologies and Healthy Sciences, Haematology Division 1, University of Torino, Italy
| | - Ymera Pignochino
- Medical Oncology Division, Candiolo Cancer Institute, FPO-IRCCS. Str. Prov. 142, km 3.95, I-10060, Candiolo (To), Italy.,Department of Oncology, University of Torino, Candiolo (Torino) Italy
| | - Marco Basiricó
- Medical Oncology Division, Candiolo Cancer Institute, FPO-IRCCS. Str. Prov. 142, km 3.95, I-10060, Candiolo (To), Italy
| | - Federica Capozzi
- Medical Oncology Division, Candiolo Cancer Institute, FPO-IRCCS. Str. Prov. 142, km 3.95, I-10060, Candiolo (To), Italy
| | - Anna Sapino
- Pathology Division, Candiolo Cancer Institute, FPO-IRCCS, Str. Prov. 142, km 3.95, I-10060, Candiolo (To), Italy.,Department of Medical Sciences, University of Torino, Italy
| | - Massimo Aglietta
- Medical Oncology Division, Candiolo Cancer Institute, FPO-IRCCS. Str. Prov. 142, km 3.95, I-10060, Candiolo (To), Italy.,Department of Oncology, University of Torino, Candiolo (Torino) Italy
| | - Dario Sangiolo
- Medical Oncology Division, Candiolo Cancer Institute, FPO-IRCCS. Str. Prov. 142, km 3.95, I-10060, Candiolo (To), Italy.,Department of Oncology, University of Torino, Candiolo (Torino) Italy
| |
Collapse
|
23
|
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: 261] [Impact Index Per Article: 37.3] [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
|
24
|
Zhang W, Barger CJ, Eng KH, Klinkebiel D, Link PA, Omilian A, Bshara W, Odunsi K, Karpf AR. PRAME expression and promoter hypomethylation in epithelial ovarian cancer. Oncotarget 2018; 7:45352-45369. [PMID: 27322684 PMCID: PMC5216727 DOI: 10.18632/oncotarget.9977] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Accepted: 05/29/2016] [Indexed: 12/19/2022] Open
Abstract
PRAME is a cancer-testis antigen (CTA) and potential immuno-therapeutic target, but has not been well-studied in epithelial ovarian cancer (EOC) or its high grade serous (HGSC) subtype. Compared to normal ovary, PRAME expression was significantly increased most EOC, regardless of stage and grade. Interestingly, PRAME mRNA expression was associated with improved survival in the HGSC subtype. The PRAME locus was a frequent target for copy number alterations (CNA) in HGSC but most changes were heterozygous losses, indicating that elevated PRAME expression is not typically due to CNA. In contrast, PRAME promoter DNA hypomethylation was very common in EOC and HGSC and correlated with increased PRAME expression. PRAME expression and promoter hypomethylation both correlated with LINE-1 hypomethylation, a biomarker of global DNA hypomethylation. Pharmacologic or genetic disruption of DNA methyltransferase (DNMT) enzymes activated PRAME expression in EOC cells. Immunohistochemistry (IHC) of PRAME in EOC revealed frequent, but low level, protein expression, and expression was confined to epithelial cells and localized to the cytoplasm. Cytoplasmic PRAME expression was positively associated with PRAME mRNA expression and negatively associated with promoter methylation, but the latter correlation was not statistically significant. PRAME protein expression did not correlate with EOC clinicopathology or survival. In summary, PRAME is frequently expressed in EOC at the mRNA and protein levels, and DNA methylation is a key mechanism regulating its expression. These data support PRAME as an immunotherapy target in EOC, and suggest treatment with DNMT inhibitors as a means to augment PRAME immunotherapy.
Collapse
Affiliation(s)
- Wa Zhang
- Eppley Institute for Cancer Research, University of Nebraska Medical Center, Omaha, NE, USA.,Current address: Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Carter J Barger
- Eppley Institute for Cancer Research, University of Nebraska Medical Center, Omaha, NE, USA
| | - Kevin H Eng
- Department of Biostatistics and Bioinformatics, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - David Klinkebiel
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Petra A Link
- Department of Pharmacology, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Angela Omilian
- Department of Pathology, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Wiam Bshara
- Department of Pathology, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Kunle Odunsi
- Department of Gynecologic Oncology, Roswell Park Cancer Institute, Buffalo, NY, USA.,Department of Immunology, Roswell Park Cancer Institute, Buffalo, NY, USA.,Center for Immunotherapy, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Adam R Karpf
- Eppley Institute for Cancer Research, University of Nebraska Medical Center, Omaha, NE, USA.,Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
25
|
Pollack SM. The potential of the CMB305 vaccine regimen to target NY-ESO-1 and improve outcomes for synovial sarcoma and myxoid/round cell liposarcoma patients. Expert Rev Vaccines 2018; 17:107-114. [PMID: 29280411 PMCID: PMC6521962 DOI: 10.1080/14760584.2018.1419068] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
INTRODUCTION Synovial Sarcoma (SS) and Myxoid Round Cell Liposarcoma (MRCL) are devastating sarcoma subtypes with few treatment options and poor outcomes in the advanced setting. However, both these diseases may be ideal for novel immunotherapies targeting the cancer-testis antigen, NY-ESO-1. AREAS COVERED In this review, we discuss the novel NY-ESO-1 targeted vaccine regimen, CMB305. This regimen uses a unique integration-deficient, dendritic-cell targeting lentiviral vector from the ZVex® platform, LV305, in order to prime NY-ESO-1 specific T cells. LV305 has single agent activity, and, in one case, caused a durable partial response in a refractory SS patient. CMB305 also includes a boost from a NY-ESO-1 protein vaccine given along with a potent toll-like-4 receptor agonist, glycopyranosyl lipid A. CMB305 induces NY-ESO-1 specific T cell responses in both SS and MRC patients and these patients had excellent overall survival (OS) outcomes in the initial phase I study. EXPERT COMMENTARY CMB305 is a therapeutic vaccine regimen targeting NY-ESO-1 based on the lentiviral vaccine vector, LV305. Phase I studies have proven this vaccine is active immunologically. Data suggesting this vaccine may improve OS for SS and MRCL patients is exciting but early, and on-going work is testing the impact of CMB305 on patient outcomes.
Collapse
Affiliation(s)
- Seth M Pollack
- a Clinical Research Division , Fred Hutchinson Cancer Research Center , Seattle , WA , USA
- b Department of Medicine , University of Washington , Seattle , WA , USA
| |
Collapse
|
26
|
Abstract
PURPOSE OF REVIEW The prognosis of patients with unresectable or metastatic chondrosarcoma of the bone is poor. Chondrosarcomas are in general resistant to chemotherapy and radiotherapy. This review discusses recent developments in the characterization of molecular pathways involved in the oncogenesis of chondrosarcoma that should be explored to improve prognosis of patients with advanced chondrosarcoma. RECENT FINDINGS The different oncogenic pathways for chondrosarcoma have become better defined. These include alterations in pathways such as isocitrate dehydrogenase mutation, hedgehog signalling, the retinoblastoma protein and p53 pathways, apoptosis and survival mechanisms, and several tyrosine kinases. These specific alterations can be employed for use in clinical interventions in advanced chondrosarcoma. SUMMARY As many different genetic alterations in chondrosarcoma have been identified, it is of the utmost importance to classify druggable targets that may improve the prognosis of chondrosarcoma patients. In recent years an increased number of trials evaluating targeted therapies are being conducted. As chondrosarcoma is an orphan disease consequently all studies are performed with small numbers of patients. The results of clinical studies so far have been largely disappointing. Therapeutic intervention studies of these new targets emerging from preclinical studies are of highest importance to improve prognosis of chondrosarcoma patients with advanced disease.
Collapse
|
27
|
Torabi A, Amaya CN, Wians FH, Bryan BA. PD-1 and PD-L1 expression in bone and soft tissue sarcomas. Pathology 2017; 49:506-513. [PMID: 28688724 DOI: 10.1016/j.pathol.2017.05.003] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Revised: 05/01/2017] [Accepted: 05/04/2017] [Indexed: 01/29/2023]
Abstract
PD-1 and its ligands have been shown to play a significant role in evasion of malignant tumour cells from the immune system. Last year, the Unites States Food and Drug Administration (FDA) approved anti-PD-1 inhibitors for treatment of non-small cell lung carcinoma and recently expanded the use of immunotherapy for metastatic urothelial cell carcinoma and Hodgkin lymphoma. However, studies on expression of PD-1 and its ligand in malignant bone and soft tissue sarcoma are sparse. In this study, we evaluated PD-1 and PD-L1 expression on variants of liposarcomas and rhabdomyosarcomas, osteosarcomas and chondrosarcomas. Tissue microarrays (TMAs) for liposarcomas (well differentiated, myxoid/round cell, and pleomorphic), rhabdomyosarcomas (alveolar, embryonal, pleomorphic, and spindle cell), conventional osteosarcomas and chondrosarcomas were stained for PD-1 and PD-L1 antibodies. Adipose tissue, skeletal muscle, bone, osteochondroma and lipoma were used as control and benign counterparts. Western blot was performed to evaluate expression of PD-1 and PD-L1 in four sarcoma cell lines. Osteosarcomas, chondrosarcomas, and all variants of liposarcomas and rhabdomyosarcomas over-expressed PD-1 relative to normal tissue. Expression of PD-1 in rhabdomyosarcomas was associated with higher tumour stage. Only one case of pleomorphic liposarcoma, one case of pleomorphic rhabdomyosarcoma and two cases of alveolar rhabdomyosarcomas were positive for PD-L1. Normal adipose tissue, skeletal muscle, and bone were negative for both PD-1 and PD-L1 and lipomas and osteochondroma weakly expressed PD-1 but not PD-L1. Western blot confirmed the presence of PD-1 protein in all four sarcoma cell lines. Overall, our results showed cytoplasmic expression of PD-1 in the bone and soft tissue sarcomas, while PD-L1 was negative. Whether these data are an indication for effectiveness of immunotherapy in the management of malignant bone and soft tissue sarcomas remains to be elucidated.
Collapse
Affiliation(s)
- Alireza Torabi
- Department of Pathology, Texas Tech University Health Sciences Center, Paul L. Foster School of Medicine, El Paso, Texas, United States.
| | - Clarissa N Amaya
- Department of Biomedical Sciences, Texas Tech University Health Sciences Center, Paul L. Foster School of Medicine, El Paso, Texas, United States
| | - Frank H Wians
- Department of Pathology, Texas Tech University Health Sciences Center, Paul L. Foster School of Medicine, El Paso, Texas, United States
| | - Brad A Bryan
- Department of Biomedical Sciences, Texas Tech University Health Sciences Center, Paul L. Foster School of Medicine, El Paso, Texas, United States
| |
Collapse
|
28
|
Abstract
Elevated expression of preferentially expressed antigen in melanoma (PRAME) has been implicated in disease progression in a variety of cancers. However, the mechanisms underlying the transcriptional regulation of PRAME remain largely unexplored. Initially, we observed that PRAME was elevated in proportion to the malignant potential of melanoma cells. From the in silico prediction of PRAME gene structure, we identified the putative myeloid zinc finger 1 (MZF1) binding sites, which overlap with a CpG-rich region located in the first intron. The transcription factor MZF1 increased PRAME expression via its direct binding to the intron DNA. Upon treatment with a DNA methylation inhibitor, 5-aza-2'-deoxycitidine (5-azaC), together with ectopic expression of MZF1, PRAME expression was significantly enhanced at both the protein and mRNA levels. More pronounced MZF1 binding to the PRAME DNA was observed in the presence of 5-azaC. DNA methylation was inversely correlated with PRAME expression in melanoma cells. Finally, we observed that MZF1, like PRAME, promotes the colony-forming ability in melanoma cells. Overall, our findings suggest that MZF1, via stimulation of PRAME expression, may be a potential prognostic and therapeutic target in melanoma.
Collapse
|
29
|
Roszik J, Wang WL, Livingston JA, Roland CL, Ravi V, Yee C, Hwu P, Futreal A, Lazar AJ, Patel SR, Conley AP. Overexpressed PRAME is a potential immunotherapy target in sarcoma subtypes. Clin Sarcoma Res 2017. [PMID: 28630682 PMCID: PMC5471883 DOI: 10.1186/s13569-017-0077-3] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Background PRAME (preferentially expressed antigen in melanoma), a member of the cancer-testis antigen family, has been shown to have increased expression in solid tumors, including sarcoma, and PRAME-specific therapies are currently in development for other cancers such as melanoma. Methods To map the landscape of PRAME expression in sarcoma, we used publicly available data from The Cancer Genome Atlas (TCGA) and the Cancer Cell Line Encyclopedia (CCLE) projects and determined which sarcoma subtypes and subsets are associated with increased PRAME expression. We also analyzed how PRAME expression correlates with survival and expression of markers related to antigen presentation and T cell function. Furthermore, tumor and normal tissue expression comparisons were performed using data from the genotype-tissue expression (GTEx) project. Results We found that uterine carcinosarcoma highly overexpresses the PRAME antigen, and synovial sarcomas and multifocal leiomyosarcomas also show high expressions suggesting that PRAME may be an effective target of immunotherapies of these tumors. However, we also discovered that PRAME expression negatively correlates with genes involved in antigen presentation, and in synovial sarcoma MHC class I antigen presentation deficiencies are also present, potentially limiting the efficacy of immunotherapies of this malignancy. Conclusions We determined that uterine carcinosarcoma, synovial sarcoma, and leiomyosarcoma patients would potentially benefit from PRAME-specific immunotherapies. Tumor escape through loss of antigen presentation needs to be further studied.
Collapse
Affiliation(s)
- Jason Roszik
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030 USA.,Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030 USA
| | - Wei-Lien Wang
- Department of Pathology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030 USA
| | - John A Livingston
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030 USA
| | - Christina L Roland
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030 USA
| | - Vinod Ravi
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030 USA
| | - Cassian Yee
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030 USA
| | - Patrick Hwu
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030 USA.,Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030 USA
| | - Andrew Futreal
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030 USA
| | - Alexander J Lazar
- Department of Pathology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030 USA
| | - Shreyaskumar R Patel
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030 USA
| | - Anthony P Conley
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030 USA
| |
Collapse
|
30
|
Pollack SM, He Q, Yearley JH, Emerson R, Vignali M, Zhang Y, Redman MW, Baker KK, Cooper S, Donahue B, Loggers ET, Cranmer LD, Spraker MB, Seo YD, Pillarisetty VG, Ricciotti RW, Hoch BL, McClanahan TK, Murphy E, Blumenschein WM, Townson SM, Benzeno S, Riddell SR, Jones RL. T-cell infiltration and clonality correlate with programmed cell death protein 1 and programmed death-ligand 1 expression in patients with soft tissue sarcomas. Cancer 2017; 123:3291-3304. [PMID: 28463396 PMCID: PMC5568958 DOI: 10.1002/cncr.30726] [Citation(s) in RCA: 194] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Revised: 03/01/2017] [Accepted: 03/16/2017] [Indexed: 12/15/2022]
Abstract
BACKGROUND Patients with metastatic sarcomas have poor outcomes and although the disease may be amenable to immunotherapies, information regarding the immunologic profiles of soft tissue sarcoma (STS) subtypes is limited. METHODS The authors identified patients with the common STS subtypes: leiomyosarcoma, undifferentiated pleomorphic sarcoma (UPS), synovial sarcoma (SS), well‐differentiated/dedifferentiated liposarcoma, and myxoid/round cell liposarcoma. Gene expression, immunohistochemistry for programmed cell death protein (PD‐1) and programmed death‐ligand 1 (PD‐L1), and T‐cell receptor Vβ gene sequencing were performed on formalin‐fixed, paraffin‐embedded tumors from 81 patients. Differences in liposarcoma subsets also were evaluated. RESULTS UPS and leiomyosarcoma had high expression levels of genes related to antigen presentation and T‐cell infiltration. UPS were found to have higher levels of PD‐L1 (P≤.001) and PD‐1 (P≤.05) on immunohistochemistry and had the highest T‐cell infiltration based on T‐cell receptor sequencing, significantly more than SS, which had the lowest (P≤.05). T‐cell infiltrates in UPS also were more oligoclonal compared with SS and liposarcoma (P≤.05). A model adjusted for STS histologic subtype found that for all sarcomas, T‐cell infiltration and clonality were highly correlated with PD‐1 and PD‐L1 expression levels (P≤.01). CONCLUSIONS In the current study, the authors provide the most detailed overview of the immune microenvironment in sarcoma subtypes to date. UPS, which is a more highly mutated STS subtype, provokes a substantial immune response, suggesting that it may be well suited to treatment with immune checkpoint inhibitors. The SS and liposarcoma subsets are less mutated but do express immunogenic self‐antigens, and therefore strategies to improve antigen presentation and T‐cell infiltration may allow for successful immunotherapy in patients with these diagnoses. Cancer 2017;123:3291‐304. © 2017 The Authors. Cancer published by Wiley Periodicals, Inc. on behalf of American Cancer Society. This is an open access article under the terms of the Creative Commons Attribution NonCommercial License, which permits use, distribution and reproduction in any medium, provided the original work is properly cited and is not used for commercial purposes. To the author's knowledge, the current study provides the most comprehensive characterization of the sarcoma tumor immune microenvironment to date through the use of gene expression analysis, immunohistochemistry, and T‐cell receptor sequencing. The results demonstrate that some sarcoma subtypes, such as synovial sarcoma, are immunologically quiet, whereas others, such as undifferentiated pleomorphic sarcoma, are highly inflammatory and could be susceptible to immune checkpoint inhibition.
Collapse
Affiliation(s)
- Seth M Pollack
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington.,Department of Medicine, University of Washington, Seattle, Washington
| | - Qianchuan He
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington.,Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | | | - Ryan Emerson
- Adaptive Biotechnologies Corporation, Seattle, Washington
| | | | - Yuzheng Zhang
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington.,Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Mary W Redman
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Kelsey K Baker
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Sara Cooper
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Bailey Donahue
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Elizabeth T Loggers
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington.,Department of Medicine, University of Washington, Seattle, Washington
| | - Lee D Cranmer
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington.,Department of Medicine, University of Washington, Seattle, Washington
| | - Matthew B Spraker
- Department of Radiation Oncology, University of Washington, Seattle, Washington
| | - Y David Seo
- Department of Surgery, University of Washington, Seattle, Washington
| | | | | | - Benjamin L Hoch
- Department of Pathology, University of Washington, Seattle, Washington
| | | | | | | | | | - Sharon Benzeno
- Adaptive Biotechnologies Corporation, Seattle, Washington
| | - Stanley R Riddell
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington.,Department of Medicine, University of Washington, Seattle, Washington.,Institute for Advanced Study, Technical University of Munich, Munich, Germany
| | - Robin L Jones
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington.,Department of Medicine, University of Washington, Seattle, Washington.,Royal Marsden Hospital and Institute of Cancer Research, London
| |
Collapse
|
31
|
Lee A, Huang P, DeMatteo RP, Pollack SM. Immunotherapy for Soft Tissue Sarcoma: Tomorrow Is Only a Day Away. Am Soc Clin Oncol Educ Book 2017; 35:281-90. [PMID: 27249707 DOI: 10.1200/edbk_157439] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Despite the advances taking place for patients with many types of cancer, to date there has been little success in meeting the great need for novel treatments of advanced soft tissue sarcoma with effective immunologic therapies. Here, we review recent clinical and preclinical data that indicate immune responses against sarcomas occur spontaneously and can also be successfully provoked. Efforts to manipulate the sarcoma immune microenvironment have the potential to eradicate disease and may also sensitize tumors to other tumor-targeted immunotherapeutic approaches. Other approaches, including vaccines and genetic engineering of T cells, offer a promising opportunity to actively direct cytotoxic lymphocytes toward antigen-bearing sarcomas. Drawing parallels with recent advances made in other cancer types, we identify ways in which sarcomas can be included in the ongoing immunotherapy revolution.
Collapse
Affiliation(s)
- Alex Lee
- From the Department of Medical Oncology, Royal Marsden Hospital London, United Kingdom; Institute of Cancer Research, London, United Kingdom; Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY; Fred Hutchinson Cancer Research Center, Division of Oncology, University of Washington, Seattle, WA
| | - Paul Huang
- From the Department of Medical Oncology, Royal Marsden Hospital London, United Kingdom; Institute of Cancer Research, London, United Kingdom; Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY; Fred Hutchinson Cancer Research Center, Division of Oncology, University of Washington, Seattle, WA
| | - Ronald P DeMatteo
- From the Department of Medical Oncology, Royal Marsden Hospital London, United Kingdom; Institute of Cancer Research, London, United Kingdom; Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY; Fred Hutchinson Cancer Research Center, Division of Oncology, University of Washington, Seattle, WA
| | - Seth M Pollack
- From the Department of Medical Oncology, Royal Marsden Hospital London, United Kingdom; Institute of Cancer Research, London, United Kingdom; Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY; Fred Hutchinson Cancer Research Center, Division of Oncology, University of Washington, Seattle, WA
| |
Collapse
|
32
|
Jones RL. Understanding sarcomas and other rare tumors: an interview with Robin L Jones. Future Oncol 2016; 13:391-394. [PMID: 27924638 DOI: 10.2217/fon-2016-0469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Robin L Jones speaks to Jade Parker, Commissioning Editor: Robin Jones is a medical oncologist specializing in the treatment of bone and soft tissue sarcomas and Head of the Sarcoma Unit at The Royal Marsden. He completed his medical training at Guy's and St Thomas' Hospital, and his oncology training at The Royal Marsden. His postgraduate research degree, with Professor Dowsett at the Institute of Cancer Research (ICR), evaluated potential predictive and prognostic factors in breast cancer patients treated with neoadjuvant chemotherapy. In January 2010 he was appointed Associate Professor and Director of the Sarcoma Program at the University of Washington and Fred Hutchinson Cancer Research Center in Seattle. He led a successful, grant funded program and continued his translational and clinical trial-based research. The laboratory work with Dr Seth Pollack evaluated novel immunotherapeutic targets in bone and soft tissue sarcomas, and has led to a number of early-phase immunotherapeutic clinical trials in sarcoma. He returned to The Royal Marsden and Institute of Cancer Research in December 2014, as Sarcoma Clinical Trials Team Leader and Consultant Medical Oncologist. He has experience in conducting Phase I, II and III trials, as well as translational studies in sarcoma. He is continuing a number of trials of investigational agents as well as laboratory-based immunotherapy studies.
Collapse
Affiliation(s)
- Robin L Jones
- Royal Marsden Hospital/Institute of Cancer Research, Fulham Road, London, SW3 6JJ, UK
| |
Collapse
|
33
|
Current Immunotherapies for Sarcoma: Clinical Trials and Rationale. Sarcoma 2016; 2016:9757219. [PMID: 27703409 PMCID: PMC5039267 DOI: 10.1155/2016/9757219] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Accepted: 08/24/2016] [Indexed: 12/21/2022] Open
Abstract
Sarcoma tumors are rare and heterogeneous, yet they possess many characteristics that may facilitate immunotherapeutic responses. Both active strategies including vaccines and passive strategies involving cellular adoptive immunotherapy have been applied clinically. Results of these clinical trials indicate a distinct benefit for select patients. The recent breakthrough of immunologic checkpoint inhibition is being rapidly introduced to a variety of tumor types including sarcoma. It is anticipated that these emerging immunotherapies will exhibit clinical efficacy for a variety of sarcomas. The increasing ability to tailor immunologic therapies to sarcoma patients will undoubtedly generate further enthusiasm and clinical research for this treatment modality.
Collapse
|
34
|
Analysis of PD-L1, T-cell infiltrate and HLA expression in chondrosarcoma indicates potential for response to immunotherapy specifically in the dedifferentiated subtype. Mod Pathol 2016; 29:1028-37. [PMID: 27312065 DOI: 10.1038/modpathol.2016.108] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2016] [Revised: 04/27/2016] [Accepted: 05/01/2016] [Indexed: 02/07/2023]
Abstract
Therapies targeting the programmed cell death 1 (PD-1) or its ligand (PD-L1) promote antitumor T-cell activity, leading to unprecedented long-lasting tumor responses in some advanced cancers. Because of radiotherapy and chemotherapy resistance, no effective treatments have been defined for advanced chondrosarcomas. We here report an immunohistochemical analysis of PD-L1 expression in a large series of conventional, mesenchymal, clear cell and dedifferentiated chondrosarcomas using tissue microarrays. In the PD-L1-positive tumors, we analyzed the immune microenvironment (T-cell and macrophage infiltration as well as HLA class I expression) using whole sections. PD-L1 expression was absent in conventional (n=119), mesenchymal (n=19) and clear cell (n=20) chondrosarcomas. Forty-one percent (9 of the 22) of dedifferentiated chondrosarcomas displayed PD-L1 positivity. These results were confirmed in an independent cohort using whole tissue sections of dedifferentiated chondrosarcomas in which PD-L1 expression was detected in 52% (11 of the 21) of cases. PD-L1 expression was exclusively found in the dedifferentiated component and expression positively correlated with other immune parameters such as high number of tumor-infiltrating lymphocytes (P=0.014) and positive HLA class I expression (P=0.024) but not with patient overall survival (P=0.22). The presence of PD-L1 expression in association with immune-infiltrating cells and HLA class I expression in nearly 50% of the dedifferentiated chondrosarcomas provides rationale for including these patients in clinical trials with PD-1/PD-L1-targeted therapies.
Collapse
|
35
|
TCR-engineered T cells to treat tumors: Seeing but not touching? Semin Immunol 2016; 28:10-21. [PMID: 26997556 DOI: 10.1016/j.smim.2016.03.002] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Revised: 03/02/2016] [Accepted: 03/04/2016] [Indexed: 12/17/2022]
Abstract
Adoptive transfer of T cells gene-engineered with T cell receptors (TCRs) has proven its feasibility and therapeutic potential in the treatment of malignant tumors. To ensure further clinical development of TCR gene therapy, it is necessary to accurately select TCRs that demonstrate antigen-selective responses that are restricted to tumor cells and, at the same time, include strategies that restore or enhance the entry, migration and local accumulation of T cells in tumor tissues. Here, we present the current standing of TCR-engineered T cell therapy, discuss and propose procedures to select TCRs as well as strategies to sensitize the tumor to T cell trafficking, and provide a rationale for combination therapies with TCR-engineered T cells.
Collapse
|
36
|
Esfandiary A, Ghafouri-Fard S. New York esophageal squamous cell carcinoma-1 and cancer immunotherapy. Immunotherapy 2016; 7:411-39. [PMID: 25917631 DOI: 10.2217/imt.15.3] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
New York esophageal squamous cell carcinoma 1 (NY-ESO-1) is a known cancer testis gene with exceptional immunogenicity and prevalent expression in many cancer types. These characteristics have made it an appropriate vaccine candidate with the potential application against various malignancies. This article reviews recent knowledge about the NY-ESO-1 biology, function, immunogenicity and expression in cancers as well as and the results of clinical trials with this antigen.
Collapse
Affiliation(s)
- Ali Esfandiary
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran 19857-17443, Iran
| | | |
Collapse
|
37
|
Vγ9Vδ2 T cells and zoledronate mediate antitumor activity in an orthotopic mouse model of human chondrosarcoma. Tumour Biol 2015; 37:7333-44. [DOI: 10.1007/s13277-015-4615-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Accepted: 12/08/2015] [Indexed: 12/11/2022] Open
|
38
|
Gérard C, Baudson N, Ory T, Segal L, Louahed J. A Comprehensive Preclinical Model Evaluating the Recombinant PRAME Antigen Combined With the AS15 Immunostimulant to Fight Against PRAME-expressing Tumors. J Immunother 2015; 38:311-20. [PMID: 26325375 PMCID: PMC4661047 DOI: 10.1097/cji.0000000000000095] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Accepted: 07/22/2015] [Indexed: 12/30/2022]
Abstract
The PRAME tumor antigen is a potential target for immunotherapy. We assessed the immunogenicity, the antitumor activity, and the safety and the tolerability of a recombinant PRAME protein (recPRAME) combined with the AS15 immunostimulant (recPRAME+ AS15) in preclinical studies in mice and Cynomolgus monkeys. Four groups of 12 CB6F1 mice received 4 injections of phosphate-buffered saline (PBS), recPRAME, AS15, or recPRAME+AS15. Immunized mice were injected with tumor cells expressing PRAME (CT26-PRAME) 2 weeks or 2 months after the last injection. The mean tumor surface was measured twice a week. Two groups of 10 monkeys received 7 injections of saline or recPRAME+ AS15. T-cell responses were measured by flow cytometry using intracellular cytokine staining (ICS). In CB6F1 mice, repeated injections of recPRAME+ AS15 induced high PRAME-specific antibody titers and mostly CD4+ T cells producing cytokines. This immune response was long-lasting in these animals and was associated with protection against a challenge with PRAME-expressing tumor cells (CT26-PRAME) applied either 2 weeks or 2 months after the last injection; these data indicate the induction of an immune memory. In HLA-A02.01/HLA-DR1 transgenic mice, recPRAME+ AS15 induced both CD4+ and CD8+ T-cell responses, indicating that this antigen can be processed by the human leukocyte antigen and is potentially immunogenic in humans. In addition, a repeated-dose toxicity study in monkeys showed that 7 biweekly injections of recPRAME+ AS15 were well tolerated, and induced PRAME-specific antibodies and T cells. In conclusion, these preclinical data indicate that repeated injections of the PRAME cancer immunotherapeutic are immunogenic and have an acceptable safety profile.
Collapse
|
39
|
NY-ESO-1 (CTAG1B) expression in mesenchymal tumors. Mod Pathol 2015; 28:587-95. [PMID: 25412843 DOI: 10.1038/modpathol.2014.155] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Revised: 10/09/2014] [Accepted: 10/15/2014] [Indexed: 12/29/2022]
Abstract
New York esophageal squamous cell carcinoma 1 (NY-ESO-1, CTAG1B) is a cancer-testis antigen and currently a focus of several targeted immunotherapeutic strategies. We performed a large-scale immunohistochemical expression study of NY-ESO-1 using tissue microarrays of mesenchymal tumors from three institutions in an international collaboration. A total of 1132 intermediate and malignant and 175 benign mesenchymal lesions were enrolled in this study. Immunohistochemical staining was performed on tissue microarrays using a monoclonal antibody for NY-ESO-1. Among mesenchymal tumors, myxoid liposarcomas showed the highest positivity for NY-ESO-1 (88%), followed by synovial sarcomas (49%), myxofibrosarcomas (35%), and conventional chondrosarcomas (28%). Positivity of NY-ESO-1 in the remaining mesenchymal tumors was consistently low, and no immunoreactivity was observed in benign mesenchymal lesions. On the basis of these findings, nearly 90% of myxoid liposarcomas, as well as a significant proportion of synovial sarcomas, myxofibrosarcomas, and conventional chondrosarcomas are good candidates for immunotherapy targeting NY-ESO-1.
Collapse
|
40
|
Li X, Mei Q, Nie J, Fu X, Han W. Decitabine: a promising epi-immunotherapeutic agent in solid tumors. Expert Rev Clin Immunol 2015; 11:363-75. [DOI: 10.1586/1744666x.2015.1002397] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
41
|
Abstract
Cancer testis antigens, such as NY-ESO-1, are expressed in a variety of prevalent tumors and represent potential targets for T-cell receptor (TCR) gene therapy. DNA encoding a murine anti-NY-ESO-1 TCR gene (mTCR) was isolated from immunized HLA-A*0201 transgenic mice and inserted into a γ-retroviral vector. Two mTCR vectors were produced and used to transduce human PBL. Transduced cells were cocultured with tumor target cell lines and T2 cells pulsed with the NY-ESO-1 peptide, and assayed for cytokine release and cell lysis activity. The most active TCR construct was selected for production of a master cell bank for clinical use. mTCR-transduced PBL maintained TCR expression in short-term and long-term culture, ranging from 50% to 90% efficiency 7-11 days after stimulation and 46%-82% 10-20 days after restimulation. High levels of interferon-γ secretion were observed (1000-12000 pg/mL), in tumor coculture assays and recognition of peptide-pulsed cells was observed at 0.1 ng/mL, suggesting that the new mTCR had high avidity for antigen recognition. mTCR-transduced T cells also specifically lysed human tumor targets. In all assays, the mTCR was equivalent or better than the comparable human TCR. As the functional activity of TCR-transduced cells may be affected by the formation of mixed dimers, mTCRs, which are less likely to form mixed dimers with endogenous hTCRs, may be more effective in vivo. This new mTCR targeted to NY-ESO-1 represents a novel potential therapeutic option for adoptive cell-transfer therapy for a variety of malignancies.
Collapse
|
42
|
Pollack SM, Jones RL, Farrar EA, Lai IP, Lee SM, Cao J, Pillarisetty VG, Hoch BL, Gullett A, Bleakley M, Conrad EU, Eary JF, Shibuya KC, Warren EH, Carstens JN, Heimfeld S, Riddell SR, Yee C. Tetramer guided, cell sorter assisted production of clinical grade autologous NY-ESO-1 specific CD8(+) T cells. J Immunother Cancer 2014; 2:36. [PMID: 25317334 PMCID: PMC4196009 DOI: 10.1186/s40425-014-0036-y] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Accepted: 09/09/2014] [Indexed: 12/31/2022] Open
Abstract
Background Adoptive T cell therapy represents an attractive modality for the treatment of patients with cancer. Peripheral blood mononuclear cells have been used as a source of antigen specific T cells but the very low frequency of T cells recognizing commonly expressed antigens such as NY-ESO-1 limit the applicability of this approach to other solid tumors. To overcome this, we tested a strategy combining IL-21 modulation during in vitro stimulation with first-in-class use of tetramer-guided cell sorting to generate NY-ESO-1 specific cytotoxic T lymphocytes (CTL). Methods CTL generation was evaluated in 6 patients with NY-ESO-1 positive sarcomas, under clinical manufacturing conditions and characterized for phenotypic and functional properties. Results Following in vitro stimulation, T cells stained with NY-ESO-1 tetramer were enriched from frequencies as low as 0.4% to >90% after single pass through a clinical grade sorter. NY-ESO-1 specific T cells were generated from all 6 patients. The final products expanded on average 1200-fold to a total of 36 billion cells, were oligoclonal and contained 67-97% CD8+, tetramer+ T cells with a memory phenotype that recognized endogenous NY-ESO-1. Conclusion This study represents the first series using tetramer-guided cell sorting to generate T cells for adoptive therapy. This approach, when used to target more broadly expressed tumor antigens such as WT-1 and additional Cancer-Testis antigens will enhance the scope and feasibility of adoptive T cell therapy. Electronic supplementary material The online version of this article (doi:10.1186/s40425-014-0036-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Seth M Pollack
- Clinical Research Division, D3-100 Fred Hutchinson Cancer Research Center, 1100 Fairview Ave, Seattle, WA 98109 USA ; Department of Medicine, University of Washington, Seattle, WA USA
| | - Robin L Jones
- Clinical Research Division, D3-100 Fred Hutchinson Cancer Research Center, 1100 Fairview Ave, Seattle, WA 98109 USA ; Department of Medicine, University of Washington, Seattle, WA USA
| | - Erik A Farrar
- Clinical Research Division, D3-100 Fred Hutchinson Cancer Research Center, 1100 Fairview Ave, Seattle, WA 98109 USA
| | - Ivy P Lai
- Clinical Research Division, D3-100 Fred Hutchinson Cancer Research Center, 1100 Fairview Ave, Seattle, WA 98109 USA ; Institute for Advanced Study, Technical University of Munich, Munich, Germany
| | - Sylvia M Lee
- Clinical Research Division, D3-100 Fred Hutchinson Cancer Research Center, 1100 Fairview Ave, Seattle, WA 98109 USA ; Department of Medicine, University of Washington, Seattle, WA USA
| | - Jianhong Cao
- Clinical Research Division, D3-100 Fred Hutchinson Cancer Research Center, 1100 Fairview Ave, Seattle, WA 98109 USA
| | - Venu G Pillarisetty
- Clinical Research Division, D3-100 Fred Hutchinson Cancer Research Center, 1100 Fairview Ave, Seattle, WA 98109 USA ; Department of Surgery, University of Washington, Seattle, WA USA
| | - Benjamin L Hoch
- Department of Pathology, University of Washington, Seattle, WA USA
| | - Ashley Gullett
- Department of Pathology, University of Washington, Seattle, WA USA
| | - Marie Bleakley
- Clinical Research Division, D3-100 Fred Hutchinson Cancer Research Center, 1100 Fairview Ave, Seattle, WA 98109 USA ; Department of Pediatrics, University of Washington, Seattle, WA USA
| | - Ernest U Conrad
- Department of Orthopedics, University of Washington, Seattle, WA USA
| | - Janet F Eary
- Department of Radiology, University of Alabama, Birmingham, AL USA
| | - Kendall C Shibuya
- Clinical Research Division, D3-100 Fred Hutchinson Cancer Research Center, 1100 Fairview Ave, Seattle, WA 98109 USA
| | - Edus H Warren
- Clinical Research Division, D3-100 Fred Hutchinson Cancer Research Center, 1100 Fairview Ave, Seattle, WA 98109 USA ; Department of Medicine, University of Washington, Seattle, WA USA
| | - Jason N Carstens
- Clinical Research Division, D3-100 Fred Hutchinson Cancer Research Center, 1100 Fairview Ave, Seattle, WA 98109 USA
| | - Shelly Heimfeld
- Clinical Research Division, D3-100 Fred Hutchinson Cancer Research Center, 1100 Fairview Ave, Seattle, WA 98109 USA
| | - Stanley R Riddell
- Clinical Research Division, D3-100 Fred Hutchinson Cancer Research Center, 1100 Fairview Ave, Seattle, WA 98109 USA ; Department of Medicine, University of Washington, Seattle, WA USA ; Institute for Advanced Study, Technical University of Munich, Munich, Germany
| | - Cassian Yee
- Clinical Research Division, D3-100 Fred Hutchinson Cancer Research Center, 1100 Fairview Ave, Seattle, WA 98109 USA ; Department of Medicine, University of Washington, Seattle, WA USA ; Department of Melanoma Medical Oncology, UT MD Anderson Cancer Center, 7455 Fannin St, Unit 904, Houston, TX 77054 USA
| |
Collapse
|
43
|
|
44
|
Sigalotti L, Fratta E, Coral S, Maio M. Epigenetic drugs as immunomodulators for combination therapies in solid tumors. Pharmacol Ther 2013; 142:339-50. [PMID: 24384533 DOI: 10.1016/j.pharmthera.2013.12.015] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2013] [Accepted: 12/10/2013] [Indexed: 12/14/2022]
Abstract
Continuously improving knowledge of the fine mechanisms regulating cross-talk between immune cells, and of their multi-faceted interactions with cancer cells, has prompted the development of several novel immunotherapeutic strategies for cancer treatment. Among these, modulation of the host's immune system by targeting immunological synapses has shown notable clinical efficacy in different tumor types. Despite this, objective clinical responses and, more importantly, long-term survival are achieved only by a fraction of patients; therefore, identification of the mechanism(s) responsible for the differential effectiveness of immune checkpoint blockade in specific patient populations is an area of intense investigation. Neoplastic cells can activate multiple mechanisms to escape from immune control; among these, epigenetic reprogramming is emerging as a key player. Selected tumor-associated antigens, Human Leukocyte Antigens, and accessory/co-stimulatory molecules required for efficient recognition of neoplastic cells by the immune system have been shown to be epigenetically silenced or down-regulated in cancer. Consistent with the inherent reversibility of epigenetic silencing, "epigenetic" drugs, such as inhibitors of DNA methyltransferases and of histone deacetylases, can restore the functional expression of these down-regulated molecules, thus improving the recognition of cancer cells by both the innate and adaptive immune responses. This review focuses on the immunomodulatory activity of epigenetic drugs and on their proposed clinical use in novel combined chemo-immunotherapeutic regimens for the treatment of solid tumors.
Collapse
Affiliation(s)
- Luca Sigalotti
- Cancer Bioimmunotherapy Unit, Centro di Riferimento Oncologico Aviano, National Cancer Institute, Aviano, Italy
| | - Elisabetta Fratta
- Cancer Bioimmunotherapy Unit, Centro di Riferimento Oncologico Aviano, National Cancer Institute, Aviano, Italy
| | - Sandra Coral
- Medical Oncology and Immunotherapy, Department of Oncology, University Hospital of Siena, Istituto Toscano Tumori, Siena, Italy
| | - Michele Maio
- Medical Oncology and Immunotherapy, Department of Oncology, University Hospital of Siena, Istituto Toscano Tumori, Siena, Italy.
| |
Collapse
|
45
|
Kunert A, Straetemans T, Govers C, Lamers C, Mathijssen R, Sleijfer S, Debets R. TCR-Engineered T Cells Meet New Challenges to Treat Solid Tumors: Choice of Antigen, T Cell Fitness, and Sensitization of Tumor Milieu. Front Immunol 2013; 4:363. [PMID: 24265631 PMCID: PMC3821161 DOI: 10.3389/fimmu.2013.00363] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2013] [Accepted: 10/24/2013] [Indexed: 01/18/2023] Open
Abstract
Adoptive transfer of T cells gene-engineered with antigen-specific T cell receptors (TCRs) has proven its feasibility and therapeutic potential in the treatment of malignant tumors. To ensure further clinical development of TCR gene therapy, it is necessary to target immunogenic epitopes that are related to oncogenesis and selectively expressed by tumor tissue, and implement strategies that result in optimal T cell fitness. In addition, in particular for the treatment of solid tumors, it is equally necessary to include strategies that counteract the immune-suppressive nature of the tumor micro-environment. Here, we will provide an overview of the current status of TCR gene therapy, and redefine the following three challenges of improvement: “choice of target antigen”; “fitness of T cells”; and “sensitization of tumor milieu.” We will categorize and discuss potential strategies to address each of these challenges, and argue that advancement of clinical TCR gene therapy critically depends on developments toward each of the three challenges.
Collapse
Affiliation(s)
- Andre Kunert
- Laboratory of Experimental Tumor Immunology, Erasmus MC Cancer Institute , Rotterdam , Netherlands ; Department of Medical Oncology, Erasmus MC Cancer Institute , Rotterdam , Netherlands
| | | | | | | | | | | | | |
Collapse
|
46
|
Yao Y, Zhou J, Wang L, Gao X, Ning Q, Jiang M, Wang J, Wang L, Yu L. Increased PRAME-specific CTL killing of acute myeloid leukemia cells by either a novel histone deacetylase inhibitor chidamide alone or combined treatment with decitabine. PLoS One 2013; 8:e70522. [PMID: 23940586 PMCID: PMC3734248 DOI: 10.1371/journal.pone.0070522] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2012] [Accepted: 06/25/2013] [Indexed: 12/15/2022] Open
Abstract
As one of the best known cancer testis antigens, PRAME is overexpressed exclusively in germ line tissues such as the testis as well as in a variety of solid and hematological malignant cells including acute myeloid leukemia. Therefore, PRAME has been recognized as a promising target for both active and adoptive anti-leukemia immunotherapy. However, in most patients with PRAME-expressing acute myeloid leukemia, PRAME antigen-specific CD8(+) CTL response are either undetectable or too weak to exert immune surveillance presumably due to the inadequate PRAME antigen expression and PRAME-specific antigen presentation by leukemia cells. In this study, we observed remarkably increased PRAME mRNA expression in human acute myeloid leukemia cell lines and primary acute myeloid leukemia cells after treatment with a novel subtype-selective histone deacetylase inhibitor chidamide in vitro. PRAME expression was further enhanced in acute myeloid leukemia cell lines after combined treatment with chidamide and DNA demethylating agent decitabine. Pre-treatment of an HLA-A0201(+) acute myeloid leukemia cell line THP-1 with chidamide and/or decitabine increased sensitivity to purified CTLs that recognize PRAME(100-108) or PRAME(300-309) peptide presented by HLA-A0201. Chidamide-induced epigenetic upregulation of CD86 also contributed to increased cytotoxicity of PRAME antigen-specific CTLs. Our data thus provide a new line of evidence that epigenetic upregulation of cancer testis antigens by a subtype-selective HDAC inhibitor or in combination with hypomethylating agent increases CTL cytotoxicity and may represent a new opportunity in future design of treatment strategy targeting specifically PRAME-expressing acute myeloid leukemia.
Collapse
Affiliation(s)
- Yushi Yao
- Department of Hematology and BMT Center, Chinese PLA General Hospital, Beijing, China
| | - Jihao Zhou
- Department of Hematology and BMT Center, Chinese PLA General Hospital, Beijing, China
| | - Lixin Wang
- Department of Hematology, Navy General Hospital, Beijing, China
| | - Xiaoning Gao
- Department of Hematology and BMT Center, Chinese PLA General Hospital, Beijing, China
| | - Qiaoyang Ning
- Department of Hematology and BMT Center, Chinese PLA General Hospital, Beijing, China
| | - Mengmeng Jiang
- Department of Hematology and BMT Center, Chinese PLA General Hospital, Beijing, China
| | - Jia Wang
- Department of Hematology and BMT Center, Chinese PLA General Hospital, Beijing, China
| | - Lili Wang
- Department of Hematology and BMT Center, Chinese PLA General Hospital, Beijing, China
| | - Li Yu
- Department of Hematology and BMT Center, Chinese PLA General Hospital, Beijing, China
- * E-mail:
| |
Collapse
|
47
|
Stiles JM, Rowntree RK, Amaya C, Diaz D, Kokta V, Mitchell DC, Bryan BA. Gene expression analysis reveals marked differences in the transcriptome of infantile hemangioma endothelial cells compared to normal dermal microvascular endothelial cells. Vasc Cell 2013; 5:6. [PMID: 23531100 PMCID: PMC3655845 DOI: 10.1186/2045-824x-5-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2013] [Accepted: 03/13/2013] [Indexed: 01/11/2023] Open
Abstract
Background Infantile hemangiomas are benign vascular tumors primarily found on the skin in 10% of the pediatric population. The etiology of this disease is largely unknown and while large scale genomic studies have examined the transcriptomes of infantile hemangioma tumors as a whole, no study to date has compared the global gene expression profiles of pure infantile hemangioma endothelial cells (HEMECs) to that of normal human dermal microvascular endothelial cells (HDMVECs). Methods To shed light on the molecular differences between these normal and aberrant dermal endothelial cell types, we performed whole genome microarray analysis on purified cultures of HEMECs and HDMVECs. We then utilized qPCR and immunohistochemistry to confirm our microarray results. Results Our array analysis identified 125 genes whose expression was upregulated and 104 genes whose expression was downregulated by greater than two fold in HEMECs compared to HDMVECs. Bioinformatics analysis revealed three major classifications of gene functions that were altered in HEMECs including cell adhesion, cell cycle, and arachidonic acid production. Several of these genes have been reported to be critical regulators and/or mutated in cancer, vascular tumors, and vascular malformations. We confirmed the expression of a subset of these differentially expressed genes (ANGPT2, ANTXR1, SMARCE1, RGS5, CTAG2, LTBP2, CLDN11, and KISS1) using qPCR and utilized immunohistochemistry on a panel of paraffin embedded infantile hemangioma tumor tissues to demonstrate that the cancer/testis antigen CTAG2 is highly abundant in vessel-dense proliferating infantile hemangiomas and with significantly reduced levels during tumor involution as vascular density decreases. Conclusion Our data reveal that the transcriptome of HEMECs is reflective of a pro-proliferative cell type with altered adhesive characteristics. Moveover, HEMECs show altered expression of many genes that are important in the progression and prognosis of metastatic cancers.
Collapse
Affiliation(s)
- Jessica M Stiles
- Department of Biomedical Sciences, Paul L, Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, TX, USA.
| | | | | | | | | | | | | |
Collapse
|