1
|
Genty G, Sandoval-Castillo J, Beheregaray LB, Möller LM. Into the Blue: Exploring genetic mechanisms behind the evolution of baleen whales. Gene 2024; 929:148822. [PMID: 39103058 DOI: 10.1016/j.gene.2024.148822] [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: 03/12/2024] [Revised: 07/29/2024] [Accepted: 08/02/2024] [Indexed: 08/07/2024]
Abstract
Marine ecosystems are ideal for studying evolutionary adaptations involved in lineage diversification due to few physical barriers and reduced opportunities for strict allopatry compared to terrestrial ecosystems. Cetaceans (whales, dolphins, and porpoises) are a diverse group of mammals that successfully adapted to various habitats within the aquatic environment around 50 million years ago. While the overall adaptive transition from terrestrial to fully aquatic species is relatively well understood, the radiation of modern whales is still unclear. Here high-quality genomes derived from previously published data were used to identify genomic regions that potentially underpinned the diversification of baleen whales (Balaenopteridae). A robust molecular phylogeny was reconstructed based on 10,159 single copy and complete genes for eight mysticetes, seven odontocetes and two cetacean outgroups. Analysis of positive selection across 3,150 genes revealed that balaenopterids have undergone numerous idiosyncratic and convergent genomic variations that may explain their diversification. Genes associated with aging, survival and homeostasis were enriched in all species. Additionally, positive selection on genes involved in the immune system were disclosed for the two largest species, blue and fin whales. Such genes can potentially be ascribed to their morphological evolution, allowing them to attain greater length and increased cell number. Further evidence is presented about gene regions that might have contributed to the extensive anatomical changes shown by cetaceans, including adaptation to distinct environments and diets. This study contributes to our understanding of the genomic basis of diversification in baleen whales and the molecular changes linked to their adaptive radiation, thereby enhancing our understanding of cetacean evolution.
Collapse
Affiliation(s)
- Gabrielle Genty
- Cetacean Ecology, Behaviour and Evolution Lab, College of Science and Engineering, Flinders University, GPO Box 2100, Adelaide, SA 5001, Australia; Molecular Ecology Lab, College of Science and Engineering, Flinders University, GPO Box 2100, Adelaide, SA 5001, Australia.
| | - Jonathan Sandoval-Castillo
- Molecular Ecology Lab, College of Science and Engineering, Flinders University, GPO Box 2100, Adelaide, SA 5001, Australia
| | - Luciano B Beheregaray
- Molecular Ecology Lab, College of Science and Engineering, Flinders University, GPO Box 2100, Adelaide, SA 5001, Australia
| | - Luciana M Möller
- Cetacean Ecology, Behaviour and Evolution Lab, College of Science and Engineering, Flinders University, GPO Box 2100, Adelaide, SA 5001, Australia; Molecular Ecology Lab, College of Science and Engineering, Flinders University, GPO Box 2100, Adelaide, SA 5001, Australia
| |
Collapse
|
2
|
Fu Y, Zhu X, Ren L, Wan J, Wang H. Syringeable Near-Infrared Light-Activated In Situ Immunogenic Hydrogel Boosts the Cancer-Immunity Cycle to Enhance Anticancer Immunity. ACS NANO 2024; 18:14877-14892. [PMID: 38809421 DOI: 10.1021/acsnano.3c08425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/30/2024]
Abstract
Effective anticancer immunity depends on properly activating multiple stepwise events in the cancer-immunity cycle. An immunologically "cold" tumor microenvironment (TME) engenders immune evasion and refractoriness to conventional checkpoint blockade immunotherapy. Here, we combine nanoparticle formulations and an in situ formed hydrogel scaffold to treat accessible tumors locally and to stimulate systemic immunity against metastatic tumor lesions. The nanoparticles encapsulate poly(ε-caprolactone)-derived cytotoxic chemotherapy and adjuvant of Toll-like receptor 7/8 through a reactive oxygen species (ROS)-cleavable linker that can be self-activated by the coassembled neighboring photosensitizer following near-infrared (NIR) laser irradiation. Further development results in syringeable, NIR light-responsive, and immunogenic hydrogel (iGEL) that can be implanted peritumorally and deposited into the tumor surgical bed. Upon NIR laser irradiation, the generated ROS induces iGEL degradation and bond cleavage in the polymer-drug conjugates, triggering the immunogenic cell death cascade in cancer cells and spontaneously releasing encapsulated agents to rewire the cancer-immunity cycle. Notably, upon application in multiple preclinical models of melanoma and triple-negative breast cancer, which are aggressive and refractory to conventional immunotherapy, iGEL induces durable remission of established tumors, extends postsurgical tumor-free survival, and inhibits metastatic burden. The result of this study is a locally administrable immunogenic hydrogel for triggering host systemic immunity to improve immunotherapeutic efficacy with minimal off-target side effects.
Collapse
Affiliation(s)
- Yang Fu
- The First Affiliated Hospital; NHC Key Laboratory of Combined Multi-Organ Transplantation, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 310003, P. R. China
| | - Xiaoxiao Zhu
- Department of Medical Oncology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 310016, P. R. China
| | - Lulu Ren
- The First Affiliated Hospital; Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 310003, P. R. China
| | - Jianqin Wan
- The First Affiliated Hospital; Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 310003, P. R. China
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, Shandong Province 250117, P. R. China
| | - Hangxiang Wang
- The First Affiliated Hospital; Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 310003, P. R. China
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, Shandong Province 250117, P. R. China
- Department of Hepatobiliary Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang Province 325000, P. R. China
| |
Collapse
|
3
|
Venzel R, Campos MCP, de Oliveira LP, Dan Lins RV, Siena ÁDD, Mesquita KT, Moreira Dos Santos TP, Nohata N, Arruda LCM, Sales-Campos H, Neto MPC. Clinical and molecular overview of immunotherapeutic approaches for malignant skin melanoma: Past, present and future. Crit Rev Oncol Hematol 2023; 186:103988. [PMID: 37086955 DOI: 10.1016/j.critrevonc.2023.103988] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 03/25/2023] [Accepted: 04/11/2023] [Indexed: 04/24/2023] Open
Abstract
Traditional therapeutic approaches for malignant melanoma, have proved to be limited and/or ineffective, especially with respect to their role in improving patient survival and tumor recurrence. In this regard, immunotherapy has been demonstrated to be a promising therapeutic alternative, boosting antitumor responses through the modulation of cell signaling pathways involved in the effector mechanisms of the immune system, particularly, the so-called "immunological checkpoints". Clinical studies on the efficacy and safety of immunotherapeutic regimens, alone or in combination with other antitumor approaches, have increased dramatically in recent decades, with very encouraging results. Hence, this review will discuss the current immunotherapeutic regimens used to treat malignant melanoma, as well as the molecular and cellular mechanisms involved. In addition, current clinical studies that have investigated the use, efficacy, and adverse events of immunotherapy in melanoma will also be discussed.
Collapse
Affiliation(s)
- Raphaelly Venzel
- Institute of Health and Biotechnology, Federal University of Amazonas, Coari, Brazil
| | | | | | | | | | | | - Tálita Pollyana Moreira Dos Santos
- Department of Oral Biology, School of Dental Medicine, University at Buffalo, Buffalo, NY, USA; Head & Neck Surgery, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Nijiro Nohata
- Oncology Science Unit, MSD K.K, Chiyoda-ku, Tokyo, Japan
| | | | - Helioswilton Sales-Campos
- Institute of Tropical Pathology and Public Health, Federal University of Goiás, Goiânia, Goiás, GO, Brazil
| | | |
Collapse
|
4
|
Therapeutic Vaccines Targeting Neoantigens to Induce T-Cell Immunity against Cancers. Pharmaceutics 2022; 14:pharmaceutics14040867. [PMID: 35456701 PMCID: PMC9029780 DOI: 10.3390/pharmaceutics14040867] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 04/11/2022] [Accepted: 04/13/2022] [Indexed: 12/12/2022] Open
Abstract
Cancer immunotherapy has achieved multiple clinical benefits and has become an indispensable component of cancer treatment. Targeting tumor-specific antigens, also known as neoantigens, plays a crucial role in cancer immunotherapy. T cells of adaptive immunity that recognize neoantigens, but do not induce unwanted off-target effects, have demonstrated high efficacy and low side effects in cancer immunotherapy. Tumor neoantigens derived from accumulated genetic instability can be characterized using emerging technologies, such as high-throughput sequencing, bioinformatics, predictive algorithms, mass-spectrometry analyses, and immunogenicity validation. Neoepitopes with a higher affinity for major histocompatibility complexes can be identified and further applied to the field of cancer vaccines. Therapeutic vaccines composed of tumor lysates or cells and DNA, mRNA, or peptides of neoantigens have revoked adaptive immunity to kill cancer cells in clinical trials. Broad clinical applicability of these therapeutic cancer vaccines has emerged. In this review, we discuss recent progress in neoantigen identification and applications for cancer vaccines and the results of ongoing trials.
Collapse
|
5
|
A decade of checkpoint blockade immunotherapy in melanoma: understanding the molecular basis for immune sensitivity and resistance. Nat Immunol 2022; 23:660-670. [PMID: 35241833 DOI: 10.1038/s41590-022-01141-1] [Citation(s) in RCA: 306] [Impact Index Per Article: 102.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Accepted: 01/18/2022] [Indexed: 12/30/2022]
Abstract
Ten years since the immune checkpoint inhibitor ipilimumab was approved for advanced melanoma, it is time to reflect on the lessons learned regarding modulation of the immune system to treat cancer and on novel approaches to further extend the efficacy of current and emerging immunotherapies. Here, we review the studies that led to our current understanding of the melanoma immune microenvironment in humans and the mechanistic work supporting these observations. We discuss how this information is guiding more precise analyses of the mechanisms of action of immune checkpoint blockade and novel immunotherapeutic approaches. Lastly, we review emerging evidence supporting the negative impact of melanoma metabolic adaptation on anti-tumor immunity and discuss how to counteract such mechanisms for more successful use of immunotherapy.
Collapse
|
6
|
Development of Pharmaceutical Nanomedicines: From the Bench to the Market. Pharmaceutics 2022; 14:pharmaceutics14010106. [PMID: 35057002 PMCID: PMC8777701 DOI: 10.3390/pharmaceutics14010106] [Citation(s) in RCA: 129] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Revised: 12/15/2021] [Accepted: 12/30/2021] [Indexed: 12/13/2022] Open
Abstract
Nanotechnology plays a significant role in the field of medicine and in drug delivery, mainly due to the major limitations affecting the conventional pharmaceutical agents, and older formulations and delivery systems. The effect of nanotechnology on healthcare is already being felt, as various nanotechnology applications have been developed, and several nanotechnology-based medicines are now on the market. Across many parts of the world, nanotechnology draws increasing investment from public authorities and the private sector. Most conventional drug-delivery systems (CDDSs) have an immediate, high drug release after administration, leading to increased administration frequency. Thus, many studies have been carried out worldwide focusing on the development of pharmaceutical nanomedicines for translation into products manufactured by local pharmaceutical companies. Pharmaceutical nanomedicine products are projected to play a major role in the global pharmaceutical market and healthcare system. Our objectives were to examine the nanomedicines approved by the Food and Drug Administration (FDA) and the European Medicines Agency (EMA) in the global market, to briefly cover the challenges faced during their development, and to look at future perspectives. Additionally, the importance of nanotechnology in developing pharmaceutical products, the ideal properties of nanocarriers, the reasons behind the failure of some nanomedicines, and the important considerations in the development of nanomedicines will be discussed in brief.
Collapse
|
7
|
Zhu J, Yuan Y, Wan X, Yin D, Li R, Chen W, Suo C, Song H. Immunotherapy (excluding checkpoint inhibitors) for stage I to III non-small cell lung cancer treated with surgery or radiotherapy with curative intent. Cochrane Database Syst Rev 2021; 12:CD011300. [PMID: 34870327 PMCID: PMC8647093 DOI: 10.1002/14651858.cd011300.pub3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND Non-small cell lung cancer (NSCLC) is the most common lung cancer, accounting for approximately 80% to 85% of all cases. For people with localised NSCLC (stages I to III), it has been speculated that immunotherapy may be helpful for reducing postoperative recurrence rates, or improving the clinical outcomes of current treatment for unresectable tumours. This is an update of a Cochrane Review first published in 2017 and it includes two new randomised controlled trials (RCTs). OBJECTIVES To assess the effectiveness and safety of immunotherapy (excluding checkpoint inhibitors) among people with localised NSCLC of stages I to III who received curative intent of radiotherapy or surgery. SEARCH METHODS We searched the following databases (from inception to 19 May 2021): CENTRAL, MEDLINE, Embase, CINAHL, and five trial registers. We also searched conference proceedings and reference lists of included trials. SELECTION CRITERIA We included RCTs conducted in adults (≥ 18 years) diagnosed with NSCLC stage I to III after surgical resection, and those with unresectable locally advanced stage III NSCLC receiving radiotherapy with curative intent. We included participants who underwent primary surgical treatment, postoperative radiotherapy or chemoradiotherapy if the same strategy was provided for both intervention and control groups. DATA COLLECTION AND ANALYSIS Two review authors independently selected eligible trials, assessed risk of bias, and extracted data. We used survival analysis to pool time-to-event data, using hazard ratios (HRs). We used risk ratios (RRs) for dichotomous data, and mean differences (MDs) for continuous data, with 95% confidence intervals (CIs). Due to clinical heterogeneity (immunotherapeutic agents with different underlying mechanisms), we combined data by applying random-effects models. MAIN RESULTS We included 11 RCTs involving 5128 participants (this included 2 new trials with 188 participants since the last search dated 20 January 2017). Participants who underwent surgical resection or received curative radiotherapy were randomised to either an immunotherapy group or a control group. The immunological interventions were active immunotherapy Bacillus Calmette-Guérin (BCG) adoptive cell transfer (i.e. transfer factor (TF), tumour-infiltrating lymphocytes (TIL), dendritic cell/cytokine-induced killer (DC/CIK), antigen-specific cancer vaccines (melanoma-associated antigen 3 (MAGE-A3) and L-BLP25), and targeted natural killer (NK) cells. Seven trials were at high risk of bias for at least one of the risk of bias domains. Three trials were at low risk of bias across all domains and one small trial was at unclear risk of bias as it provided insufficient information. We included data from nine of the 11 trials in the meta-analyses involving 4863 participants. There was no evidence of a difference between the immunotherapy agents and the controls on any of the following outcomes: overall survival (HR 0.94, 95% CI 0.84 to 1.05; P = 0.27; 4 trials, 3848 participants; high-quality evidence), progression-free survival (HR 0.94, 95% CI 0.86 to 1.03; P = 0.19; moderate-quality evidence), adverse events (RR 1.12, 95% CI 0.97 to 1.28; P = 0.11; 4 trials, 4126 evaluated participants; low-quality evidence), and severe adverse events (RR 1.14, 95% CI 0.92 to 1.40; 6 trials, 4546 evaluated participants; low-quality evidence). Survival rates at different time points showed no evidence of a difference between immunotherapy agents and the controls. Survival rate at 1-year follow-up (RR 1.02, 95% CI 0.96 to 1.08; I2 = 57%; 7 trials, 4420 participants; low-quality evidence), 2-year follow-up (RR 1.02, 95% CI 0.93 to 1.12; 7 trials, 4420 participants; moderate-quality evidence), 3-year follow-up (RR 0.99, 95% CI 0.90 to 1.09; 7 trials, 4420 participants; I2 = 22%; moderate-quality evidence) and at 5-year follow-up (RR 0.98, 95% CI 0.86 to 1.12; I2 = 0%; 7 trials, 4389 participants; moderate-quality evidence). Only one trial reported overall response rates. Two trials provided health-related quality of life results with contradicting results. AUTHORS' CONCLUSIONS: Based on this updated review, the current literature does not provide evidence that suggests a survival benefit from adding immunotherapy (excluding checkpoint inhibitors) to conventional curative surgery or radiotherapy, for people with localised NSCLC (stages I to III). Several ongoing trials with immune checkpoints inhibitors (PD-1/PD-L1) might bring new insights into the role of immunotherapy for people with stages I to III NSCLC.
Collapse
Affiliation(s)
- Jianwei Zhu
- Department of Orthopaedics, West China Hospital, Sichuan University, Chengdu, China
| | - Yun Yuan
- West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaoyu Wan
- West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, China
| | - Dan Yin
- West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, China
| | - Rui Li
- Thoracic Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Wenwen Chen
- West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, China
| | - Chen Suo
- Key Laboratory of Public Health Safety, Fudan University, Ministry of Education, Shanghai, China
| | - Huan Song
- West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, China
- Center of Public Health Sciences, Faculty of Medicine, University of Iceland, Reykjavík, Iceland
| |
Collapse
|
8
|
Dlamini Z, Hull R, Mbatha SZ, Alaouna M, Qiao YL, Yu H, Chatziioannou A. Prognostic Alternative Splicing Signatures in Esophageal Carcinoma. Cancer Manag Res 2021; 13:4509-4527. [PMID: 34113176 PMCID: PMC8186946 DOI: 10.2147/cmar.s305464] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 04/06/2021] [Indexed: 01/10/2023] Open
Abstract
Alternative splicing (AS) is a method of increasing the number of proteins that the genome is capable of coding for, by altering the pre-mRNA during its maturation. This process provides the ability of a broad range of proteins to arise from a single gene. AS events are known to occur in up to 94% of human genes. Cumulative data have shown that aberrant AS functionality is a major factor in human diseases. This review focuses on the contribution made by aberrant AS functionality in the development and progression of esophageal cancer. The changes in the pattern of expression of alternately spliced isoforms in esophageal cancer can be used as diagnostic or prognostic biomarkers. Additionally, these can be used as targets for the development of new treatments for esophageal cancer.
Collapse
Affiliation(s)
- Zodwa Dlamini
- SAMRC Precision Prevention & Novel Drug Targets for HIV-Associated Cancers Extramural Unit, Pan African Cancer Research Institute, University of Pretoria, Pretoria, South Africa
| | - Rodney Hull
- SAMRC Precision Prevention & Novel Drug Targets for HIV-Associated Cancers Extramural Unit, Pan African Cancer Research Institute, University of Pretoria, Pretoria, South Africa
| | - Sikhumbuzo Z Mbatha
- Department of Surgery, Steve Biko Academic Hospital, University of Pretoria, Pretoria, South Africa
| | - Mohammed Alaouna
- SAMRC Precision Prevention & Novel Drug Targets for HIV-Associated Cancers Extramural Unit, Pan African Cancer Research Institute, University of Pretoria, Pretoria, South Africa.,Department of Internal Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - You-Lin Qiao
- SAMRC Precision Prevention & Novel Drug Targets for HIV-Associated Cancers Extramural Unit, Pan African Cancer Research Institute, University of Pretoria, Pretoria, South Africa.,Cancer Institute/Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, People's Republic of China
| | - Herbert Yu
- SAMRC Precision Prevention & Novel Drug Targets for HIV-Associated Cancers Extramural Unit, Pan African Cancer Research Institute, University of Pretoria, Pretoria, South Africa.,University of Hawaii Cancer Center, Honolulu, HI, USA
| | - Aristotelis Chatziioannou
- SAMRC Precision Prevention & Novel Drug Targets for HIV-Associated Cancers Extramural Unit, Pan African Cancer Research Institute, University of Pretoria, Pretoria, South Africa.,Center of Systems Biology, Biomedical Research Foundation Academy of Athens, Athens, Greece.,e-NIOS Applications PC, Kallithea, Athens, 17676, Greece
| |
Collapse
|
9
|
Anti-PD-1/PD-L1 Based Combination Immunotherapy to Boost Antigen-Specific CD8 + T Cell Response in Hepatocellular Carcinoma. Cancers (Basel) 2021; 13:cancers13081922. [PMID: 33923463 PMCID: PMC8073815 DOI: 10.3390/cancers13081922] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 03/22/2021] [Accepted: 04/14/2021] [Indexed: 12/13/2022] Open
Abstract
Simple Summary The cytotoxic T cell response against hepatocellular carcinoma antigens is exhausted and fails in its task of deleting tumoral cells. These cells are featured by the expression of negative immune checkpoints that can be modulated to restore T cell function. The blockade of the PD-1/PD-L1 pathway has shown promising results in rescuing hepatocellular carcinoma-specific CD8 T cells but only a reduced group of cases is sensitive to this treatment and the effect is usually temporary. Therefore, new anti-PD-1 based combinatory strategies are underway to increase the response by adding the effect of blocking neo-angiogenesis and other negative immune checkpoints, boosting positive immune checkpoints, blocking suppressive cytokines, or inducing the expression of tumoral neoantigens. The restoration of T cell responses with these anti-PD-1 based combinatory therapies will change the outcome of advanced hepatocellular carcinoma. Abstract Thirty to fifty percent of hepatocellular carcinomas (HCC) display an immune class genetic signature. In this type of tumor, HCC-specific CD8 T cells carry out a key role in HCC control. Those potential reactive HCC-specific CD8 T cells recognize either HCC immunogenic neoantigens or aberrantly expressed host’s antigens, but they become progressively exhausted or deleted. These cells express the negative immunoregulatory checkpoint programmed cell death protein 1 (PD-1) which impairs T cell receptor signaling by blocking the CD28 positive co-stimulatory signal. The pool of CD8 cells sensitive to anti-PD-1/PD-L1 treatment is the PD-1dim memory-like precursor pool that gives rise to the effector subset involved in HCC control. Due to the epigenetic imprints that are transmitted to the next generation, the effect of PD-1 blockade is transient, and repeated treatments lead to tumor resistance. During long-lasting disease, besides the TCR signaling impairment, T cells develop other failures that should be also set-up to increase T cell reactivity. Therefore, several PD-1 blockade-based combinatory therapies are currently under investigation such as adding antiangiogenics, anti-TGFβ1, blockade of other negative immune checkpoints, or increasing HCC antigen presentation. The effect of these combinations on CD8+ T cells is discussed in this review.
Collapse
|
10
|
CD8 + T Cell Responses during HCV Infection and HCC. J Clin Med 2021; 10:jcm10050991. [PMID: 33801203 PMCID: PMC7957882 DOI: 10.3390/jcm10050991] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 02/12/2021] [Accepted: 02/22/2021] [Indexed: 12/15/2022] Open
Abstract
Chronic hepatitis C virus (cHCV) infection is a major global health burden and the leading cause of hepatocellular carcinoma (HCC) in the Western world. The course and outcome of HCV infection is centrally influenced by CD8+ T cell responses. Indeed, strong virus-specific CD8+ T cell responses are associated with spontaneous viral clearance while failure of these responses, e.g., caused by viral escape and T cell exhaustion, is associated with the development of chronic infection. Recently, heterogeneity within the exhausted HCV-specific CD8+ T cells has been observed with implications for immunotherapeutic approaches also for other diseases. In HCC, the presence of tumor-infiltrating and peripheral CD8+ T cell responses correlates with a favorable prognosis. Thus, tumor-associated and tumor-specific CD8+ T cells are considered suitable targets for immunotherapeutic strategies. Here, we review the current knowledge of CD8+ T cell responses in chronic HCV infection and HCC and their respective failure with the potential consequences for T cell-associated immunotherapeutic approaches.
Collapse
|
11
|
Rijensky NM, Blondheim Shraga NR, Barnea E, Peled N, Rosenbaum E, Popovtzer A, Stemmer SM, Livoff A, Shlapobersky M, Moskovits N, Perry D, Rubin E, Haviv I, Admon A. Identification of Tumor Antigens in the HLA Peptidome of Patient-derived Xenograft Tumors in Mouse. Mol Cell Proteomics 2020; 19:1360-1374. [PMID: 32451349 PMCID: PMC8015002 DOI: 10.1074/mcp.ra119.001876] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 05/20/2020] [Indexed: 12/15/2022] Open
Abstract
Personalized cancer immunotherapy targeting patient-specific cancer/testis antigens (CTA) and neoantigens may benefit from large-scale tumor human leukocyte antigen (HLA) peptidome (immunopeptidome) analysis, which aims to accurately identify antigens presented by tumor cells. Although significant efforts have been invested in analyzing the HLA peptidomes of fresh tumors, it is often impossible to obtain sufficient volumes of tumor tissues for comprehensive HLA peptidome characterization. This work attempted to overcome some of these obstacles by using patient-derived xenograft tumors (PDX) in mice as the tissue sources for HLA peptidome analysis. PDX tumors provide a proxy for the expansion of the patient tumor by re-grafting them through several passages to immune-compromised mice. The HLA peptidomes of human biopsies were compared with those derived from PDX tumors. Larger HLA peptidomes were obtained from the significantly larger PDX tumors as compared with the patient biopsies. The HLA peptidomes of different PDX tumors derived from the same source tumor biopsy were very reproducible, even following subsequent passages to new naïve mice. Many CTA-derived HLA peptides were discovered, as well as several potential neoantigens/variant sequences. Taken together, the use of PDX tumors for HLA peptidome analysis serves as a highly expandable and stable source of reproducible and authentic peptidomes, opening up new opportunities for defining large HLA peptidomes when only small tumor biopsies are available. This approach provides a large source for tumor antigens identification, potentially useful for personalized immunotherapy.
Collapse
Affiliation(s)
| | | | - Eilon Barnea
- Department of Biology, Technion-Israel Institute of Technology Haifa, Israel
| | - Nir Peled
- Institute of Oncology, Davidoff Center, Rabin Medical Center and Sackler Faculty of Medicine, Tel-Aviv University, Petah Tikva, Israel
| | - Eli Rosenbaum
- Institute of Oncology, Davidoff Center, Rabin Medical Center and Sackler Faculty of Medicine, Tel-Aviv University, Petah Tikva, Israel
| | - Aron Popovtzer
- Institute of Oncology, Davidoff Center, Rabin Medical Center and Sackler Faculty of Medicine, Tel-Aviv University, Petah Tikva, Israel
| | - Solomon M Stemmer
- Davidoff Center, Rabin Medical Center, Beilinson Campus, Petach Tikva, and Felsentien medical research center, Petach Tikva, and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Alejandro Livoff
- Institute of Pathology, Barzilai University Medical Center, Ashkelon, Israel
| | - Mark Shlapobersky
- Institute of Pathology, Barzilai University Medical Center, Ashkelon, Israel
| | - Neta Moskovits
- Davidoff Center, Rabin Medical Center, Beilinson Campus, Petach Tikva, and Felsentien medical research center, Petach Tikva, Israel
| | - Dafna Perry
- The Azrieli Faculty of Medicine, Bar Ilan University, Safed, Israel
| | - Eitan Rubin
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beersheva, Israel; The Shraga Segal Department of Microbiology, Immunology and Genetics, Ben-Gurion University of the Negev, Beersheba, Israel
| | - Itzhak Haviv
- The Azrieli Faculty of Medicine, Bar Ilan University, Safed, Israel
| | - Arie Admon
- Department of Biology, Technion-Israel Institute of Technology Haifa, Israel.
| |
Collapse
|
12
|
Uncovering the Tumor Antigen Landscape: What to Know about the Discovery Process. Cancers (Basel) 2020; 12:cancers12061660. [PMID: 32585818 PMCID: PMC7352969 DOI: 10.3390/cancers12061660] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 06/11/2020] [Accepted: 06/20/2020] [Indexed: 12/14/2022] Open
Abstract
According to the latest available data, cancer is the second leading cause of death, highlighting the need for novel cancer therapeutic approaches. In this context, immunotherapy is emerging as a reliable first-line treatment for many cancers, particularly metastatic melanoma. Indeed, cancer immunotherapy has attracted great interest following the recent clinical approval of antibodies targeting immune checkpoint molecules, such as PD-1, PD-L1, and CTLA-4, that release the brakes of the immune system, thus reviving a field otherwise poorly explored. Cancer immunotherapy mainly relies on the generation and stimulation of cytotoxic CD8 T lymphocytes (CTLs) within the tumor microenvironment (TME), priming T cells and establishing efficient and durable anti-tumor immunity. Therefore, there is a clear need to define and identify immunogenic T cell epitopes to use in therapeutic cancer vaccines. Naturally presented antigens in the human leucocyte antigen-1 (HLA-I) complex on the tumor surface are the main protagonists in evocating a specific anti-tumor CD8+ T cell response. However, the methodologies for their identification have been a major bottleneck for their reliable characterization. Consequently, the field of antigen discovery has yet to improve. The current review is intended to define what are today known as tumor antigens, with a main focus on CTL antigenic peptides. We also review the techniques developed and employed to date for antigen discovery, exploring both the direct elution of HLA-I peptides and the in silico prediction of epitopes. Finally, the last part of the review analyses the future challenges and direction of the antigen discovery field.
Collapse
|
13
|
Zhang X, Huang Y, Li X, Wang Y, Yuan Y, Li M. Preparation of a new combination nanoemulsion-encapsulated MAGE1-MAGE3-MAGEn/HSP70 vaccine and study of its immunotherapeutic effect. Pathol Res Pract 2020; 216:152954. [PMID: 32321658 DOI: 10.1016/j.prp.2020.152954] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 03/31/2020] [Accepted: 04/07/2020] [Indexed: 12/26/2022]
Abstract
BACKGROUND MAGE family genes have been studied as targets for tumor immunotherapy for a long time. Here, we combined MAGE1-, MAGE3- and MAGEn-derived peptides as a cancer vaccine and tested whether a new combination nanoemulsion-encapsulated vaccine could be used to inhibit the growth of tumor cells in humanized SCID mice. METHODS The nanoemulsion-encapsulated complex protein vaccine (MAGE1, MAGE3, and MAGEn/HSP70 fusion protein; M1M3MnH) was prepared using a magnetic ultrasonic technique. After screening, human PBMCs were injected into SCID mice to mimic the human immune system. Then, the humanized SCID mice were challenged with M3-HHCC cells and immunized with nanoemulsion-encapsulated MAGE1-MAGE3-MAGEn/HSP70 [NE(M1M3MnH)] or M1M3MnH. The cellular immune responses were detected by IFN-γ ELISPOT and cytotoxicity assays. Therapeutic and tumor challenge experiments were also performed. RESULTS The results showed that the immune responses elicited by NE(M1M3MnH) were apparently stronger than those elicited by M1M3MnH, NE(-) or PBS, suggesting that this novel nanoemulsion carrier induces potent antitumor immunity against the encapsulated antigens. The results of the therapeutic and tumor challenge experiments also indicated that the new vaccine had a definite effect on SCID mice bearing human hepatic cancer. CONCLUSION Our study indicated that the combination of several tumor antigen-derived peptides may be a relatively good strategy for peptide-based cancer immunotherapy. These results suggest that the complex nanoemulsion vaccine could have broader applications for both therapy and prevention mediated by antitumor effects in the future.
Collapse
Affiliation(s)
- Xiumin Zhang
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital and School of Basic Medicine, Air Force Medical University, Xi'an, 710032, China
| | - Yang Huang
- Department of Emergency, Xijing Hospital, Air Force Medical University, Xi'an, 710032, China
| | - Xia Li
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital and School of Basic Medicine, Air Force Medical University, Xi'an, 710032, China
| | - Yanxia Wang
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital and School of Basic Medicine, Air Force Medical University, Xi'an, 710032, China
| | - Yuan Yuan
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital and School of Basic Medicine, Air Force Medical University, Xi'an, 710032, China
| | - Mingyang Li
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital and School of Basic Medicine, Air Force Medical University, Xi'an, 710032, China.
| |
Collapse
|
14
|
Tokunaga A, Sugiyama D, Maeda Y, Warner AB, Panageas KS, Ito S, Togashi Y, Sakai C, Wolchok JD, Nishikawa H. Selective inhibition of low-affinity memory CD8 + T cells by corticosteroids. J Exp Med 2019; 216:2701-2713. [PMID: 31537643 PMCID: PMC6888983 DOI: 10.1084/jem.20190738] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 08/12/2019] [Accepted: 09/03/2019] [Indexed: 12/17/2022] Open
Abstract
Corticosteroids inhibit antitumor immune responses of immune checkpoint blockade in a dose- and timing-dependent manner. Memory CD8+ T cells with low TCR affinity are selectively suppressed by corticosteroids, necessitating careful and thoughtful corticosteroid use. Patients treated with immune checkpoint blockade (ICB) sometimes experience immune-related adverse events (irAEs), requiring immuno-suppressive drugs such as corticosteroids despite the possibility that immunosuppression may impair the antitumor effects of ICB. Here, we address the dilemma of using corticosteroids for the treatment of irAEs induced by ICB. ICB augments neoantigen-specific CD8+ T cell responses, resulting in tumor regression. In our model, simultaneous, but not late, administration of corticosteroids impaired antitumor responses with reduction of CD8+ T cell proliferation. Secondary challenge using tumors with/without the neoantigen showed selective progression in tumors lacking the neoantigen when corticosteroids were administered. Corticosteroids decreased low- but not high-affinity memory T cells by suppressing fatty acid metabolism essential for memory T cells. In a small cohort of human melanoma patients, overall survival was shorter after treatment with CTLA-4 blockade in patients who received early corticosteroids or had low tumor mutation burden. Together, low-affinity memory T cells are dominantly suppressed by corticosteroids, necessitating careful and thoughtful corticosteroid use.
Collapse
Affiliation(s)
- Akihiro Tokunaga
- Division of Cancer Immunology, Research Institute/Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Japan.,Oncology Research and Development Unit, Kyowa Kirin Co., Ltd., Shizuoka, Japan
| | - Daisuke Sugiyama
- Department of Immunology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yuka Maeda
- Division of Cancer Immunology, Research Institute/Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Japan
| | - Allison Betof Warner
- Parker Institute for Cancer Immunotherapy, Swim Across America-Ludwig Collaborative Lab, Memorial Sloan-Kettering Cancer Center, New York, NY.,Weill Cornell Medical College, New York, NY
| | - Katherine S Panageas
- Departments of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Sachiko Ito
- Department of Immunology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yosuke Togashi
- Division of Cancer Immunology, Research Institute/Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Japan
| | - Chika Sakai
- Division of Cancer Immunology, Research Institute/Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Japan
| | - Jedd D Wolchok
- Parker Institute for Cancer Immunotherapy, Swim Across America-Ludwig Collaborative Lab, Memorial Sloan-Kettering Cancer Center, New York, NY .,Weill Cornell Medical College, New York, NY
| | - Hiroyoshi Nishikawa
- Division of Cancer Immunology, Research Institute/Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Japan .,Department of Immunology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
15
|
Zapka P, Dörner E, Dreschmann V, Sakamato N, Kristiansen G, Calaminus G, Vokuhl C, Leuschner I, Pietsch T. Type, Frequency, and Spatial Distribution of Immune Cell Infiltrates in CNS Germinomas: Evidence for Inflammatory and Immunosuppressive Mechanisms. J Neuropathol Exp Neurol 2019; 77:119-127. [PMID: 29237087 DOI: 10.1093/jnen/nlx106] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Central nervous system germinomas are characterized by a massive immune cell infiltrate. We systematically characterized these immune cells in 28 germinomas by immunophenotyping and image analysis. mRNA expression was analyzed by Nanostring technology and in situ RNA hybridization. Tumor infiltrating lymphocytes (TILs) were composed of 61.8% ± 3.1% (mean ± SE) CD3-positive T cells, including 45.2% ± 3.5% of CD4-positive T-helper cells, 23.4% ± 1.5% of CD8-positive cytotoxic T cells, 5.5% ± 0.9% of FoxP3-positive regulatory T cells, and 11.9% ±1.3% PD-1-positive TILs. B cells accounted for 35.8% ± 2.9% of TILs and plasma cells for 9.3% ± 1.6%. Tumor-associated macrophages consisted of clusters of activated PD-L1-positive macrophages and interspersed anti-inflammatory macrophages expressing CD163. Germinoma cells did not express PD-L1. Expression of genes encoding immune cell markers and cytokines was high and comparable to mRNA levels in lymph node tissue. IFNG and IL10 mRNA was detected in subfractions of TILs and in PD-L1-positive macrophages. Taken together, the strong immune reaction observed in germinomas involves inflammatory as well as various suppressive mechanisms. Expression of PD-1 and PD-L1 and infiltration of cytotoxic T cells are biomarkers predictive of response to anti-PD-1/PD-L1 therapies, constituting a rationale for possible novel treatment approaches.
Collapse
Affiliation(s)
- Pia Zapka
- Department of Neuropathology and Brain Tumor Reference Center, University of Bonn Medical Center, Bonn, Germany
| | - Evelyn Dörner
- Department of Neuropathology and Brain Tumor Reference Center, University of Bonn Medical Center, Bonn, Germany
| | - Verena Dreschmann
- Department of Neuropathology and Brain Tumor Reference Center, University of Bonn Medical Center, Bonn, Germany
| | - Noriaki Sakamato
- Department of Diagnostic Pathology/Neurosurgery, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Glen Kristiansen
- Department of Pathology, University of Bonn Medical Center, Bonn, Germany
| | - Gabriele Calaminus
- Department of Pediatric Haematology and Oncology, University of Bonn Medical Center, Bonn, Germany
| | - Christian Vokuhl
- Pediatric Tumor Registry, Pediatric Pathology, University Hospital of Schleswig-Holstein, Kiel, Germany
| | - Ivo Leuschner
- Pediatric Tumor Registry, Pediatric Pathology, University Hospital of Schleswig-Holstein, Kiel, Germany
| | - Torsten Pietsch
- Department of Neuropathology and Brain Tumor Reference Center, University of Bonn Medical Center, Bonn, Germany
| |
Collapse
|
16
|
Yan Y, Leontovich AA, Gerdes MJ, Desai K, Dong J, Sood A, Santamaria-Pang A, Mansfield AS, Chadwick C, Zhang R, Nevala WK, Flotte TJ, Ginty F, Markovic SN. Understanding heterogeneous tumor microenvironment in metastatic melanoma. PLoS One 2019; 14:e0216485. [PMID: 31166985 PMCID: PMC6550385 DOI: 10.1371/journal.pone.0216485] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 04/22/2019] [Indexed: 01/05/2023] Open
Abstract
A systemic analysis of the tumor-immune interactions within the heterogeneous tumor microenvironment is of particular importance for understanding the antitumor immune response. We used multiplexed immunofluorescence to elucidate cellular spatial interactions and T-cell infiltrations in metastatic melanoma tumor microenvironment. We developed two novel computational approaches that enable infiltration clustering and single cell analysis-cell aggregate algorithm and cell neighborhood analysis algorithm-to reveal and to compare the spatial distribution of various immune cells relative to tumor cell in sub-anatomic tumor microenvironment areas. We showed that the heterogeneous tumor human leukocyte antigen-1 expressions differently affect the magnitude of cytotoxic T-cell infiltration and the distributions of CD20+ B cells and CD4+FOXP3+ regulatory T cells within and outside of T-cell infiltrated tumor areas. In a cohort of 166 stage III melanoma samples, high tumor human leukocyte antigen-1 expression is required but not sufficient for high T-cell infiltration, with significantly improved overall survival. Our results demonstrate that tumor cells with heterogeneous properties are associated with differential but predictable distributions of immune cells within heterogeneous tumor microenvironment with various biological features and impacts on clinical outcomes. It establishes tools necessary for systematic analysis of the tumor microenvironment, allowing the elucidation of the "homogeneous patterns" within the heterogeneous tumor microenvironment.
Collapse
Affiliation(s)
- Yiyi Yan
- Division of Medical Oncology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Alexey A. Leontovich
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Michael J. Gerdes
- Diagnostics, Imaging and Biomedical Technologies, GE Global Research Center, Niskayuna, New York, United States of America
| | - Keyur Desai
- Diagnostics, Imaging and Biomedical Technologies, GE Global Research Center, Niskayuna, New York, United States of America
| | - Jinhong Dong
- Clinical Immunology and Immunotherapeutics, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Anup Sood
- Diagnostics, Imaging and Biomedical Technologies, GE Global Research Center, Niskayuna, New York, United States of America
| | - Alberto Santamaria-Pang
- Diagnostics, Imaging and Biomedical Technologies, GE Global Research Center, Niskayuna, New York, United States of America
| | - Aaron S. Mansfield
- Division of Medical Oncology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Chrystal Chadwick
- Diagnostics, Imaging and Biomedical Technologies, GE Global Research Center, Niskayuna, New York, United States of America
| | - Rong Zhang
- Diagnostics, Imaging and Biomedical Technologies, GE Global Research Center, Niskayuna, New York, United States of America
| | - Wendy K. Nevala
- Division of Hematology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Thomas J. Flotte
- Division of Anatomic Pathology and Division of Dermatopathology and Cutaneous Immunopathology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Fiona Ginty
- Diagnostics, Imaging and Biomedical Technologies, GE Global Research Center, Niskayuna, New York, United States of America
| | - Svetomir N. Markovic
- Division of Medical Oncology, Mayo Clinic, Rochester, Minnesota, United States of America
- * E-mail:
| |
Collapse
|
17
|
McQuade JL, Homsi J, Torres-Cabala CA, Bassett R, Popuri RM, James ML, Vence LM, Hwu WJ. A phase II trial of recombinant MAGE-A3 protein with immunostimulant AS15 in combination with high-dose Interleukin-2 (HDIL2) induction therapy in metastatic melanoma. BMC Cancer 2018; 18:1274. [PMID: 30567529 PMCID: PMC6300080 DOI: 10.1186/s12885-018-5193-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Accepted: 12/06/2018] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND HDIL-2 is approved for advanced melanoma based on its durable antitumor activity. MAGE-A3 cancer immunotherapeutic (MAGE-A3 CI) is a recombinant MAGE-A3 protein combined with an immunostimulant adjuvant system and has shown antitumor activity in melanoma. We assessed the safety and anti-tumor activity of HDIL-2 combined with MAGE-A3 CI in advanced melanoma. METHODS Patients with unresectable Stage III or Stage IV MAGE-A3-positive melanoma were enrolled in this phase II study. Treatment included an induction phase of MAGE-A3 CI plus HDIL-2 for 8 cycles followed by a maintenance phase of MAGE-A3 CI monotherapy. The primary endpoints were safety and objective response assessed per RECIST v1.1. Immune biomarker and correlative studies on tumor and peripheral blood were performed. RESULTS Eighteen patients were enrolled. Seventeen patients were evaluable for safety and sixteen for response. Responses occurred in 4/16 (25%) patients with 3 complete responses, and stable disease in 6/16 (38%) patients with a disease control rate of 63%. The median duration of response was not reached at median follow-up of 36.8 months. Induction therapy of HDIL-2 + MAGE-A3 CI had similar toxicities to those reported with HDIL-2 alone. Maintenance MAGE-A3 monotherapy was well-tolerated. Increased immune checkpoint receptor expression by circulating T regulatory cells was associated with poor clinical outcomes; and responders tended to have increased tumor infiltrating T cells in the baseline tumor samples. CONCLUSIONS The safety profile of HDIL-2 + MAGE-A3 CI was similar to HDIL-2 monotherapy. Maintenance MAGE-A3 CI provides robust anti-tumor activity in patients who achieved disease control with induction therapy. Immune monitoring data suggest that MAGE-A3 CI plus checkpoint inhibitors could be a promising treatment for MAGE-A3-positive melanoma. TRIAL REGISTRATION ClinicalTrials.gov, NCT01266603 . Registered 12/24/2010, https://clinicaltrials.gov/ct2/show/NCT01266603.
Collapse
Affiliation(s)
- Jennifer L. McQuade
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe, Unit 0430, Houston, TX 77030 USA
| | - Jade Homsi
- Division of Hematology/Oncology, University of Texas Southwestern Medical Center, Dallas, TX USA
| | | | - Roland Bassett
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Rashmi Murthy Popuri
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe, Unit 0430, Houston, TX 77030 USA
| | - Marihella L. James
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe, Unit 0430, Houston, TX 77030 USA
| | - Luis M. Vence
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Wen-Jen Hwu
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe, Unit 0430, Houston, TX 77030 USA
| |
Collapse
|
18
|
Katagiri T. A potential novel option for cancer immunotherapy - TLR7 stimulation inhibits malignant melanoma bone invasion. Oncotarget 2018; 9:31792. [PMID: 30159120 PMCID: PMC6112761 DOI: 10.18632/oncotarget.25872] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Accepted: 07/23/2018] [Indexed: 01/22/2023] Open
Affiliation(s)
- Takenobu Katagiri
- Takenobu Katagiri: Division of Pathophysiology, Research Center for Genomic Medicine, Saitama Medical University, Saitama, Japan
| |
Collapse
|
19
|
Jian W, Li X, Kang J, Lei Y, Bai Y, Xue Y. Antitumor effect of recombinant Mycobacterium smegmatis expressing MAGEA3 and SSX2 fusion proteins. Exp Ther Med 2018; 16:2160-2166. [PMID: 30186454 DOI: 10.3892/etm.2018.6425] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Accepted: 06/08/2018] [Indexed: 12/17/2022] Open
Abstract
Mycobacterium smegmatis (M. smegmatis), which is a nonpathogenic and fast-growing mycobacterium, is a potential vaccine vector capable of expressing heterologous antigens. Spontaneous humoral and cellular immune responses have been demonstrated against cancer/testis antigens (CTA), including melanoma-associated antigen A (MAGEA) and SSX. In the present study, recombinant plasmids expressing MAGEA3 and SSX2 were constructed. The recombinant plasmids were transferred into M. smegmatis to generate the novel antitumor DNA vaccine. As MAGEA3 and SSX2 were in different ligation sequences, the two DNA vaccines were recombinant M. smegmatis MAGEA3-SSX2 (rM.S-MS) and recombinant M. smegmatis SSX2-MAGEA3 (rM.S-SM), respectively. The expression levels of Fusion proteins were assessed by western blotting. BALB/c mice were immunized with rM.S and western blot analysis was used to determine whether antibodies against MAGEA3 or SSX2 were produced in immunized mice. EC9706 cells were inoculated into BALB/c nude mice and the mice were maintained until an obvious visible tumor appeared on the back. Subsequently, the blood from the rM.S immunized BALB/c mice was injected into the BALB/c nude mice via the tail vein. In order to evaluate the antitumor effect of the vaccines, tumor volume and weight were measured 5 to 21 days after injection. Mice were euthanized on day 21 of tumor growth, and the tumor was dissected and weighed. The two fusion proteins were expressed in the rM.S and the specific fusion protein antibodies were expressed in the blood of immunized BALB/c mice. The tumor volumes and weight in the recombinant M. smegmatis MAGEA3 (rM.S-M) and recombinant M. smegmatis SSX2 (rM.S-S) groups were significantly reduced compared with the control group. Furthermore, the decrease in tumor volumes and weight in the rM.S-MS and rM.S-SM groups was more severe than in the rM.S-M or rM.S-S groups. There was no significant difference in the antitumor effect of the rM.S-MS and rM.S-SM groups. The present findings suggest that this rM.S may be a potential candidate therapeutic vaccine for the treatment of cancer.
Collapse
Affiliation(s)
- Wen Jian
- Department of Respiratory Medicine, The First Affiliated Hospital of The Fourth Military Medical University, Xi'an, Shaanxi 710038, P.R. China
| | - Xin Li
- Medical Oncology Center, Dongguan Kanghua Hospital, Dongguan, Guangdong 523080, P.R. China
| | - Jian Kang
- Department of Basic Microbiology, The Fourth Military Medical University, Xi'an, Shaanxi 710038, P.R. China
| | - Yingfeng Lei
- Department of Basic Microbiology, The Fourth Military Medical University, Xi'an, Shaanxi 710038, P.R. China
| | - Yinlan Bai
- Department of Basic Microbiology, The Fourth Military Medical University, Xi'an, Shaanxi 710038, P.R. China
| | - Ying Xue
- Medical Oncology Center, Dongguan Kanghua Hospital, Dongguan, Guangdong 523080, P.R. China.,Department of Radiation Oncology, The First Affiliated Hospital of The Fourth Military Medical University, Xi'an, Shaanxi 710038, P.R. China
| |
Collapse
|
20
|
Hoang NT, Acevedo LA, Mann MJ, Tolani B. A review of soft-tissue sarcomas: translation of biological advances into treatment measures. Cancer Manag Res 2018; 10:1089-1114. [PMID: 29785138 PMCID: PMC5955018 DOI: 10.2147/cmar.s159641] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Soft-tissue sarcomas are rare malignant tumors arising from connective tissues and have an overall incidence of about five per 100,000 per year. While this diverse family of malignancies comprises over 100 histological subtypes and many molecular aberrations are prevalent within specific sarcomas, very few are therapeutically targeted. Instead of utilizing molecular signatures, first-line sarcoma treatment options are still limited to traditional surgery and chemotherapy, and many of the latter remain largely ineffective and are plagued by disease resistance. Currently, the mechanism of sarcoma oncogenesis remains largely unknown, thus necessitating a better understanding of pathogenesis. Although substantial progress has not occurred with molecularly targeted therapies over the past 30 years, increased knowledge about sarcoma biology could lead to new and more effective treatment strategies to move the field forward. Here, we discuss biological advances in the core molecular determinants in some of the most common soft-tissue sarcomas - liposarcoma, angiosarcoma, leiomyosarcoma, rhabdomyosarcoma, Ewing's sarcoma, and synovial sarcoma - with an emphasis on emerging genomic and molecular pathway targets and immunotherapeutic treatment strategies to combat this confounding disease.
Collapse
Affiliation(s)
- Ngoc T Hoang
- Thoracic Oncology Program, Department of Surgery, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA
| | - Luis A Acevedo
- Thoracic Oncology Program, Department of Surgery, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA
| | - Michael J Mann
- Thoracic Oncology Program, Department of Surgery, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA
| | - Bhairavi Tolani
- Thoracic Oncology Program, Department of Surgery, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA
| |
Collapse
|
21
|
Martins MR, Santos RLD, Jatahy KDN, Matta MCD, Batista TP, Júnior JIC, Begnami MDFS, Torres LC. Could OX40 agonist antibody promote activation of the anti-tumor immune response in gastric cancer? J Surg Oncol 2018. [PMID: 29529339 DOI: 10.1002/jso.25001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND AND OBJECTIVES OX40, a membrane-bound molecule of the tumor-necrosis-factor-receptor superfamily, is a critical costimulatory receptor during the immune response, especially to T cells, but studies described their presence of OX-40 on neutrophils and monocytes, suggesting a potential role in the activation of immune response. Our aim was to characterize costimulatory receptors OX40 expression on circulating leukocytes in gastric cancer to identify novel targets for immunotherapy. METHODS Peripheral blood mononuclear cells were isolated from 24 gastric cancer patients and 34 healthy controls and the expression of costimulatory (OX40) receptors were analyzed on T cells, neutrophil and monocyte using monoclonal antibodies by flow cytometry. RESULTS We found that the higher levels of OX40 + T cells, monocytes/OX40+ and neutrophils/OX40+ from gastric cancer patients when compared to controls (P < 0.0001), and also higher levels of OX40+ T cells when compared to stages III and IV (P = 0.02). Percentage levels of total T cells were similar between patients and controls. CONCLUSIONS OX40 as a therapeutic agent has been investigated in many preclinical tumor models. Our findings suggest that of levels of costimulatory in T cells in GC will direct future studies on the role that costimulatory receptors play in the failure of T cell-mediated immunity.
Collapse
Affiliation(s)
- Mário R Martins
- Oncology Surgical Department, Sociedade Pernambucana de Combate ao Câncer-Hospital do Cancer de Pernambuco (SPCC-HCP), Recife, Brazil
| | - Rogério L D Santos
- Oncology Surgical Department, Sociedade Pernambucana de Combate ao Câncer-Hospital do Cancer de Pernambuco (SPCC-HCP), Recife, Brazil
| | - Kleber D N Jatahy
- Translational Research Laboratory Prof. C. A. Hart, Instituto de Medicina Integral Prof. Fernando Figueira (IMIP), Recife, Brazil
| | - Marina C D Matta
- Translational Research Laboratory Prof. C. A. Hart, Instituto de Medicina Integral Prof. Fernando Figueira (IMIP), Recife, Brazil
| | - Thales P Batista
- Surgical Department, Instituto de Medicina Integral Prof. Fernando Figueira (IMIP), Recife, Brazil
| | - José Iran C Júnior
- Oncology Surgical Department, Sociedade Pernambucana de Combate ao Câncer-Hospital do Cancer de Pernambuco (SPCC-HCP), Recife, Brazil.,Translational Research Laboratory Prof. C. A. Hart, Instituto de Medicina Integral Prof. Fernando Figueira (IMIP), Recife, Brazil
| | - Maria D F S Begnami
- Anatomic Pathology Department, A.C. Camargo Cancer Center, São Paulo, Brazil
| | - Leuridan C Torres
- Oncology Surgical Department, Sociedade Pernambucana de Combate ao Câncer-Hospital do Cancer de Pernambuco (SPCC-HCP), Recife, Brazil.,Translational Research Laboratory Prof. C. A. Hart, Instituto de Medicina Integral Prof. Fernando Figueira (IMIP), Recife, Brazil
| |
Collapse
|
22
|
Grah JJ, Katalinic D, Juretic A, Santek F, Samarzija M. Clinical Significance of Immunohistochemical Expression of Cancer/Testis Tumor-associated Antigens (MAGE-A1, MAGE-A3/4, NY-ESO-1) in Patients with Non-small Cell Lung Cancer. TUMORI JOURNAL 2018. [DOI: 10.1177/1430.15817] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Affiliation(s)
- Josip Joachim Grah
- Department of Oncology, University Hospital Center (KBC Zagreb), University of Zagreb School of Medicine, Zagreb, Croatia
| | - Darko Katalinic
- Department of Oncology, University Hospital Center (KBC Zagreb), University of Zagreb School of Medicine, Zagreb, Croatia
| | - Antonio Juretic
- Department of Oncology, University Hospital Center (KBC Zagreb), University of Zagreb School of Medicine, Zagreb, Croatia
| | - Fedor Santek
- Department of Oncology, University Hospital Center (KBC Zagreb), University of Zagreb School of Medicine, Zagreb, Croatia
| | - Miroslav Samarzija
- Department of Pulmonary Medicine, University Hospital Center (KBC Zagreb), University of Zagreb School of Medicine, Zagreb, Croatia
| |
Collapse
|
23
|
Gearing up T-cell immunotherapy in cervical cancer. Curr Probl Cancer 2018; 42:175-188. [DOI: 10.1016/j.currproblcancer.2018.04.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Accepted: 04/04/2018] [Indexed: 01/08/2023]
|
24
|
Fouquet G, Marcq I, Debuysscher V, Bayry J, Rabbind Singh A, Bengrine A, Nguyen-Khac E, Naassila M, Bouhlal H. Signaling lymphocytic activation molecules Slam and cancers: friends or foes? Oncotarget 2018; 9:16248-16262. [PMID: 29662641 PMCID: PMC5882332 DOI: 10.18632/oncotarget.24575] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Accepted: 12/03/2017] [Indexed: 01/01/2023] Open
Abstract
Signaling Lymphocytic Activation Molecules (SLAM) family receptors are initially described in immune cells. These receptors recruit both activating and inhibitory SH2 domain containing proteins through their Immunoreceptor Tyrosine based Switch Motifs (ITSMs). Accumulating evidence suggest that the members of this family are intimately involved in different physiological and pathophysiological events such as regulation of immune responses and entry pathways of certain viruses. Recently, other functions of SLAM, principally in the pathophysiology of neoplastic transformations have also been deciphered. These new findings may prompt SLAM to be considered as new tumor markers, diagnostic tools or potential therapeutic targets for controlling the tumor progression. In this review, we summarize the major observations describing the implications and features of SLAM in oncology and discuss the therapeutic potential attributed to these molecules.
Collapse
Affiliation(s)
- Gregory Fouquet
- INSERM 1247-GRAP, Centre Universitaire de Recherche en Santé CURS, Université de Picardie Jules Verne, CHU Sud, Amiens, France
| | - Ingrid Marcq
- INSERM 1247-GRAP, Centre Universitaire de Recherche en Santé CURS, Université de Picardie Jules Verne, CHU Sud, Amiens, France
| | - Véronique Debuysscher
- INSERM 1247-GRAP, Centre Universitaire de Recherche en Santé CURS, Université de Picardie Jules Verne, CHU Sud, Amiens, France
| | - Jagadeesh Bayry
- INSERM UMRS 1138, Centre de Recherche des Cordeliers-Paris, Paris, France
| | | | | | - Eric Nguyen-Khac
- INSERM 1247-GRAP, Centre Universitaire de Recherche en Santé CURS, Université de Picardie Jules Verne, CHU Sud, Amiens, France.,Service Hepato-Gastroenterologie, Centre Hospitalier Universitaire Sud, Amiens, France
| | - Mickael Naassila
- INSERM 1247-GRAP, Centre Universitaire de Recherche en Santé CURS, Université de Picardie Jules Verne, CHU Sud, Amiens, France
| | - Hicham Bouhlal
- INSERM 1247-GRAP, Centre Universitaire de Recherche en Santé CURS, Université de Picardie Jules Verne, CHU Sud, Amiens, France
| |
Collapse
|
25
|
Zhu J, Li R, Tiselius E, Roudi R, Teghararian O, Suo C, Song H. Immunotherapy (excluding checkpoint inhibitors) for stage I to III non-small cell lung cancer treated with surgery or radiotherapy with curative intent. Cochrane Database Syst Rev 2017; 12:CD011300. [PMID: 29247502 PMCID: PMC6486009 DOI: 10.1002/14651858.cd011300.pub2] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
BACKGROUND Non-small cell lung cancer (NSCLC) is the most common lung cancer, accounting for approximately 80% to 85% of all cases. For patients with localised NSCLC (stages I to III), it has been speculated that immunotherapy may be helpful for reducing postoperative recurrence rates, or improving the clinical outcomes of current treatment for unresectable tumours. While several new agents have now entered phase III clinical trials, we felt a systematic review was needed to address the question of the effectiveness and safety of immunotherapy in patients with stages I to III NSCLC. OBJECTIVES To evaluate the effectiveness and safety of immunotherapy (excluding checkpoint inhibitors) in patients with localised NSCLC (stages I to III) who received surgery or radiotherapy with curative intent. SEARCH METHODS We searched the following databases (from inception to 20 January 2017): CENTRAL, MEDLINE, Embase, and CINAHL, and five trial registers. We also manually checked abstracts or reports from relevant conference proceedings and the reference lists of included trials. SELECTION CRITERIA We searched for randomised controlled trials (RCTs) in adults (≥ 18 years) with histologically-confirmed early-stage (stages I to III) NSCLC after surgical resection, and those with unresectable locally advanced stage III NSCLC who had received radiotherapy with curative intent. For patients who had received primary surgical treatment, postoperative radiotherapy or chemoradiotherapy was allowed if it was used for both experimental and control groups. DATA COLLECTION AND ANALYSIS Two review authors independently selected eligible trials, assessed risk of bias, and extracted data. We used survival analysis to pool time-to-event data, expressing the intervention effect as a hazard ratio (HR). We calculated risk ratios (RR) for dichotomous data, and mean differences for continuous data, with 95% confidence intervals (CI). Due to clinical heterogeneity (immunotherapeutic agents with different underlying mechanisms), we used random-effects models for our meta-analyses. MAIN RESULTS We identified nine eligible trials that randomised 4940 participants, who had received surgical resection or curative radiotherapy, to either an immunotherapy group or a control group. Included immunological interventions were active immunotherapy (i.e. Bacillus Calmette-Guérin (BCG)), adoptive cell transfer (i.e. transfer factor (TF), tumour-infiltrating lymphocytes (TIL), dendritic cell-cytokine induced killer (DC-CIK), and antigen-specific cancer vaccines (melanoma-associated antigen 3 (MAGE-A3) and L-BLP25). Except for one small trial, which provided insufficient information for risk assessment, we assessed five studies at high risk of bias for at least one of the seven biases studied; we considered the risk of bias in the other three trials to be low. We included data from seven of the nine trials in the meta-analyses (4695 participants). We pooled data from 3693 participants from the three high quality RCTs to evaluate overall survival (OS) and progression-free survival (PFS). We found a small, but not statistically significant, improvement in OS (HR 0.94, 95% CI 0.83 to 1.06; P = 0.35), and PFS (HR 0.93, 95% CI 0.81 to 1.07; P = 0.19; high-quality evidence). The addition of immunotherapy resulted in a small, but not statistically significant, increased risk of having any adverse event (RR 1.15, 95% CI 0.97 to 1.37; P = 0.11, three trials, 3955 evaluated participants, moderate-quality evidence), or severe adverse events (RR 1.10, 95% CI 0.88 to 1.39; four trials, 4362 evaluated participants; low-quality evidence).We analysed data from six studies for one-, two-, and three-year survival rates (4265 participants), and from six studies for five-year survival rates (4234 participants). We observed no clear between-group differences (low-quality evidence for one- and two-year survival rates, and moderate-quality evidence for three- and five-year survival rate).No trial reported the overall response rates; only one trial provided health-related quality of life results. AUTHORS' CONCLUSIONS The current literature does not provide evidence that suggests a survival benefit from adding immunotherapy (excluding checkpoint inhibitors) to conventional curative surgery or radiotherapy, for patients with localised NSCLC (stages I to III). The addition of vaccine-based immunotherapy might increase the risk of adverse events. Several ongoing trials with immune checkpoints inhibitors (PD-1/PD-L1) might bring new insights for role of immunotherapy for patients with stages I to III NSCLC.
Collapse
Affiliation(s)
- Jianwei Zhu
- Shandong Provincial Hospital Affiliated to Shandong UniversityDepartment of OrthopaedicsNo.324, Jingwu RoadJinanChina250021
| | - Rui Li
- Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan UniversityThoracic OncologyChengduChina
| | | | - Raheleh Roudi
- Iran University of Medical SciencesOncopathology Research CenterHemmat Street, Next to Milad TowerTeheranIran14496‐14530
| | | | - Chen Suo
- Fudan UniversityCollaborative Innovative Center for Genetic and Development, School of Life SciencesSonghu road 2005ShanghaiChina200438
| | | |
Collapse
|
26
|
Olsen LR, Tongchusak S, Lin H, Reinherz EL, Brusic V, Zhang GL. TANTIGEN: a comprehensive database of tumor T cell antigens. Cancer Immunol Immunother 2017; 66:731-735. [PMID: 28280852 PMCID: PMC11028736 DOI: 10.1007/s00262-017-1978-y] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Accepted: 02/14/2017] [Indexed: 02/04/2023]
Abstract
Tumor T cell antigens are both diagnostically and therapeutically valuable molecules. A large number of new peptides are examined as potential tumor epitopes each year, yet there is no infrastructure for storing and accessing the results of these experiments. We have retroactively cataloged more than 1000 tumor peptides from 368 different proteins, and implemented a web-accessible infrastructure for storing and accessing these experimental results. All peptides in TANTIGEN are labeled as one of the four categories: (1) peptides measured in vitro to bind the HLA, but not reported to elicit either in vivo or in vitro T cell response, (2) peptides found to bind the HLA and to elicit an in vitro T cell response, (3) peptides shown to elicit in vivo tumor rejection, and (4) peptides processed and naturally presented as defined by physical detection. In addition to T cell response, we also annotate peptides that are naturally processed HLA binders, e.g., peptides eluted from HLA in mass spectrometry studies. TANTIGEN provides a rich data resource for tumor-associated epitope and neoepitope discovery studies and is freely available at http://cvc.dfci.harvard.edu/tantigen/ or http://projects.met-hilab.org/tadb (mirror).
Collapse
Affiliation(s)
- Lars Rønn Olsen
- Cancer Vaccine Center, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA, 02115, USA
- Department of Bio and Health Informatics, Technical University of Denmark, Lyngby, 2800, Denmark
| | - Songsak Tongchusak
- Cancer Vaccine Center, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA, 02115, USA
| | - Honghuang Lin
- Cancer Vaccine Center, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA, 02115, USA
- Section of Computational Biomedicine, Department of Medicine, Boston University School of Medicine, 72 E. Concord Street, B-616, Boston, MA, 02118, USA
| | - Ellis L Reinherz
- Cancer Vaccine Center, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA, 02115, USA
- Department of Medicine, Harvard Medical School, 25 Shattuck Street, Boston, MA, 02115, USA
- Laboratory of Immunobiology, Dana-Farber Cancer Institute, 77 Avenue Louis Pasteur, Boston, MA, 02115, USA
| | - Vladimir Brusic
- Cancer Vaccine Center, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA, 02115, USA
- School of Medicine and Bioinformatics Center, Nazarbayev University, Astana, Kazakhstan
- Department of Computer Science, Metropolitan College, Boston University, Room 254808 Commonwealth Ave, Boston, MA, 02215, USA
| | - Guang Lan Zhang
- Cancer Vaccine Center, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA, 02115, USA.
- Department of Computer Science, Metropolitan College, Boston University, Room 254808 Commonwealth Ave, Boston, MA, 02215, USA.
| |
Collapse
|
27
|
Abstract
Historically, immune-based therapies have played a leading role in the treatment of hematologic malignancies, with the efficacy of stem cell transplantation largely attributable to donor immunity against malignant cells. As new and more targeted immunotherapies have developed, their role in the treatment of hematologic malignancies is evolving and expanding. Herein, we discuss approaches for antigen discovery and review known and novel tumor antigens in hematologic malignancies. We further explore the role of established and investigational immunotherapies in hematologic malignancies, with a focus on personalization of treatment modalities such as cancer vaccines and adoptive cell therapy. Finally, we identify areas of active investigation and development. Immunotherapy is at an exciting crossroads for the treatment of hematologic malignancies, with further investigation aimed at producing effective, targeted immune therapies that maximize antitumor effects while minimizing toxicity.
Collapse
Affiliation(s)
- David A. Braun
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts, USA
| | - Catherine J. Wu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| |
Collapse
|
28
|
Mendonça BDS, Agostini M, Aquino IG, Dias WB, Bastos DC, Rumjanek FD. Suppression of MAGE-A10 alters the metastatic phenotype of tongue squamous cell carcinoma cells. Biochem Biophys Rep 2017; 10:267-275. [PMID: 28955754 PMCID: PMC5614724 DOI: 10.1016/j.bbrep.2017.04.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Revised: 04/13/2017] [Accepted: 04/18/2017] [Indexed: 12/21/2022] Open
Abstract
MAGE-A10 is a member of the MAGE protein family (melanoma associated antigen) which is overexpressed in cancer cells. Although MAGE-A10 has been characterized for some time and is generally associated to metastasis its function remains unknown. Here we describe experiments using as models oral squamous cell carcinoma (OSCC) cell lines displaying increasing metastatic potential (LN1 and LN2). These cell lines were transduced with lentivirus particles coding for short hairpin against MAGE-A10 mRNA. Repression of MAGE-A10 expression in LN2 cells altered their morphology and impaired growth of LN1 and LN2 cell lines. Furthermore, repression of MAGE-A10 expression increased cell-cell and cell matrix adhesion. Furthermore shMAGEA10 cells were shown to assemble aberrantly on a 3D culture system (microspheroids) when compared to cells transduced with the control scrambled construct. Cell migration was inhibited in knocked down cells as revealed by two different migration assays, wound healing and a phagokinetic track motility assay. In vitro invasion assay using a leiomyoma tissue derived matrix (myogel) showed that shMAGEA10 LN1 and shMAGEA10 LN2 cells displayed a significantly diminished ability to penetrate the matrices. Concomitantly, the expression of E-cadherin, N-cadherin and vimentin genes was analyzed. shMAGEA10 activated the expression of E-cadherin and repression N-cadherin and vimentin transcription. Taken together the results indicate that MAGE-A10 exerts its effects at the level of the epithelial-mesenchymal transition (EMT) presumably by regulating the expression of adhesion molecules.
Collapse
Affiliation(s)
- Bruna dos Santos Mendonça
- Instituto de Bioquímica Médica Leopoldo de Meis, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro Ilha do Fundão CEP 21941-902 Rio de Janeiro, Brazil
| | - Michelle Agostini
- Departamento de Patologia e Diagnóstico Oral - Faculdade de Odontologia, Universidade Federal do Rio de Janeiro, Brazil
| | - Iara Gonçalves Aquino
- Departamento de Patologia e Diagnóstico Oral - Faculdade de Odontologia, Universidade Federal do Rio de Janeiro, Brazil
| | - Wagner Barbosa Dias
- Laboratório de Glicobiologia Estrutural e Funcional Instituto de Biofísica-Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Débora Campanella Bastos
- Faculdade de Odontologia de Piracicaba, Universidade Estadual de Campinas, Piracicaba, SP, Brazil
| | - Franklin D. Rumjanek
- Instituto de Bioquímica Médica Leopoldo de Meis, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro Ilha do Fundão CEP 21941-902 Rio de Janeiro, Brazil
- Corresponding author.
| |
Collapse
|
29
|
Zhou Z, Lyu X, Wu J, Yang X, Wu S, Zhou J, Gu X, Su Z, Chen S. TSNAD: an integrated software for cancer somatic mutation and tumour-specific neoantigen detection. ROYAL SOCIETY OPEN SCIENCE 2017; 4:170050. [PMID: 28484631 PMCID: PMC5414268 DOI: 10.1098/rsos.170050] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 03/06/2017] [Indexed: 06/02/2023]
Abstract
Tumour antigens have attracted much attention because of their importance to cancer diagnosis, prognosis and targeted therapy. With the development of cancer genomics, the identification of tumour-specific neoantigens became possible, which is a crucial step for cancer immunotherapy. In this study, we developed software called the tumour-specific neoantigen detector for detecting cancer somatic mutations following the best practices of the genome analysis toolkit and predicting potential tumour-specific neoantigens, which could be either extracellular mutations of membrane proteins or mutated peptides presented by class I major histocompatibility complex molecules. This pipeline was beneficial to the biologist with little programmatic background. We also applied the software to the somatic mutations from the International Cancer Genome Consortium database to predict numerous potential tumour-specific neoantigens. This software is freely available from https://github.com/jiujiezz/tsnad.
Collapse
Affiliation(s)
- Zhan Zhou
- Zhejiang Provincial Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, People's Republic of China
| | - Xingzheng Lyu
- College of Computer Science and Technology, Zhejiang University, Hangzhou 310013, People's Republic of China
| | - Jingcheng Wu
- Zhejiang Provincial Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, People's Republic of China
| | - Xiaoyue Yang
- Zhejiang Provincial Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, People's Republic of China
| | - Shanshan Wu
- Zhejiang Provincial Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, People's Republic of China
| | - Jie Zhou
- Zhejiang Provincial Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, People's Republic of China
| | - Xun Gu
- Department of Genetics, Development and Cell Biology, Program of Bioinformatics and Computational Biology, Iowa State University, Ames, IA 50010, USA
| | - Zhixi Su
- State Key Laboratory of Genetic Engineering and MOE Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai 200438, People's Republic of China
| | - Shuqing Chen
- Zhejiang Provincial Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, People's Republic of China
| |
Collapse
|
30
|
Saiag P, Gutzmer R, Ascierto PA, Maio M, Grob JJ, Murawa P, Dreno B, Ross M, Weber J, Hauschild A, Rutkowski P, Testori A, Levchenko E, Enk A, Misery L, Vanden Abeele C, Vojtek I, Peeters O, Brichard VG, Therasse P. Prospective assessment of a gene signature potentially predictive of clinical benefit in metastatic melanoma patients following MAGE-A3 immunotherapeutic (PREDICT). Ann Oncol 2016; 27:1947-53. [PMID: 27502712 PMCID: PMC5035794 DOI: 10.1093/annonc/mdw291] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2016] [Revised: 05/26/2016] [Accepted: 07/20/2016] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Genomic profiling of tumor tissue may aid in identifying predictive or prognostic gene signatures (GS) in some cancers. Retrospective gene expression profiling of melanoma and non-small-cell lung cancer led to the characterization of a GS associated with clinical benefit, including improved overall survival (OS), following immunization with the MAGE-A3 immunotherapeutic. The goal of the present study was to prospectively evaluate the predictive value of the previously characterized GS. PATIENTS AND METHODS An open-label prospective phase II trial ('PREDICT') in patients with MAGE-A3-positive unresectable stage IIIB-C/IV-M1a melanoma. RESULTS Of 123 subjects who received the MAGE-A3 immunotherapeutic, 71 (58.7%) displayed the predictive GS (GS+). The 1-year OS rate was 83.1%/83.3% in the GS+/GS- populations. The rate of progression-free survival at 12 months was 5.8%/4.1% in GS+/GS- patients. The median time-to-treatment failure was 2.7/2.4 months (GS+/GS-). There was one complete response (GS-) and two partial responses (GS+). The MAGE-A3 immunotherapeutic was similarly immunogenic in both populations and had a clinically acceptable safety profile. CONCLUSION Treatment of patients with MAGE-A3-positive unresectable stage IIIB-C/IV-M1a melanoma with the MAGE-A3 immunotherapeutic demonstrated an overall 1-year OS rate of 83.5%. GS- and GS+ patients had similar 1-year OS rates, indicating that in this study, GS was not predictive of outcome. Unexpectedly, the objective response rate was lower in this study than in other studies carried out in the same setting with the MAGE-A3 immunotherapeutic. Investigation of a GS to predict clinical benefit to adjuvant MAGE-A3 immunotherapeutic treatment is ongoing in another melanoma study.This study is registered at www.clinicatrials.gov NCT00942162.
Collapse
Affiliation(s)
- P Saiag
- General Dermatology and Oncology Service, Ambroise-Paré Hospital, AP-HP, University of Versailles-Saint-Quentin-en-Yvelines, Boulogne, France
| | - R Gutzmer
- Skin Cancer Center Hannover, Hannover Medical School, Hannover, Germany
| | - P A Ascierto
- National Institute for Tumors Foundation 'G. Pascale', Napoli
| | - M Maio
- Medical Oncology and Immunotherapy, Department of Oncology, University Hospital of Siena, Istituto Toscano Tumori, Siena, Italy
| | - J-J Grob
- Department of Dermatology and Skin Cancers, La Timone APHM Hospital, Aix-Marseille University, Marseille, France
| | - P Murawa
- Department of Biochemistry and Molecular Biology, Poznań University of Medical Sciences, Poznań, Poland
| | - B Dreno
- Dermatology Clinic, Hôtel-Dieu Hospital, CHU Nantes, Nantes, France
| | - M Ross
- Department of Surgical Oncology, UTMD Anderson Cancer Center, Houston
| | - J Weber
- Moffitt Cancer Center, Tampa, USA
| | - A Hauschild
- Department of Dermatology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - P Rutkowski
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie Memorial Center and Institute of Oncology, Warsaw, Poland
| | - A Testori
- Melanoma and Soft Tissue Sarcoma Division, European Institute of Oncology, Milan, Italy
| | - E Levchenko
- Petrov Research Institute of Oncology, St Petersburg, Russian Federation
| | - A Enk
- Department of Dermatology, University of Heidelberg, Heidelberg, Germany
| | - L Misery
- Department of Dermatology, University Hospital of Brest, Brest, France
| | | | - I Vojtek
- GSK Vaccines, Rixensart, Belgium
| | | | | | | |
Collapse
|
31
|
Novel Immunotherapeutic Approaches for Head and Neck Squamous Cell Carcinoma. Cancers (Basel) 2016; 8:cancers8100087. [PMID: 27669306 PMCID: PMC5082377 DOI: 10.3390/cancers8100087] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Revised: 08/16/2016] [Accepted: 09/08/2016] [Indexed: 12/11/2022] Open
Abstract
The immune system plays a key role in preventing tumor formation by recognizing and destroying malignant cells. For over a century, researchers have attempted to harness the immune response as a cancer treatment, although this approach has only recently achieved clinical success. Head and neck squamous cell carcinoma (HNSCC) is the sixth most common cancer worldwide and is associated with cigarette smoking, alcohol consumption, betel nut use, and human papillomavirus infection. Unfortunately, worldwide mortality from HNSCC remains high, partially due to limits on therapy secondary to the significant morbidity associated with current treatments. Therefore, immunotherapeutic approaches to HNSCC treatment are attractive for their potential to reduce morbidity while improving survival. However, the application of immunotherapies to this disease has been challenging because HNSCC is profoundly immunosuppressive, resulting in decreased absolute lymphocyte counts, impaired natural killer cell function, reduced antigen-presenting cell function, and a tumor-permissive cytokine profile. Despite these challenges, numerous clinical trials testing the safety and efficacy of immunotherapeutic approaches to HNSCC treatment are currently underway, many of which have produced promising results. This review will summarize immunotherapeutic approaches to HNSCC that are currently undergoing clinical trials.
Collapse
|
32
|
Jaravine V, Raffegerst S, Schendel DJ, Frishman D. Assessment of cancer and virus antigens for cross-reactivity in human tissues. Bioinformatics 2016; 33:104-111. [DOI: 10.1093/bioinformatics/btw567] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Revised: 08/03/2016] [Accepted: 08/26/2016] [Indexed: 12/17/2022] Open
|
33
|
Gato M, Blanco-Luquin I, Zudaire M, de Morentin XM, Perez-Valderrama E, Zabaleta A, Kochan G, Escors D, Fernandez-Irigoyen J, Santamaría E. Drafting the proteome landscape of myeloid-derived suppressor cells. Proteomics 2015; 16:367-78. [PMID: 26403437 DOI: 10.1002/pmic.201500229] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Revised: 08/18/2015] [Accepted: 09/21/2015] [Indexed: 01/12/2023]
Abstract
Myeloid-derived suppressor cells (MDSCs) are a heterogeneous population of cells that are defined by their myeloid origin, immature state, and ability to potently suppress T-cell responses. They regulate immune responses and the population significantly increases in the tumor microenvironment of patients with glioma and other malignant tumors. For their study, MDSCs are usually isolated from the spleen or directly of tumors from a large number of tumor-bearing mice although promising ex vivo differentiated MDSC production systems have been recently developed. During the last years, proteomics has emerged as a powerful approach to analyze MDSCs proteomes using shotgun-based mass spectrometry (MS), providing functional information about cellular homeostasis and metabolic state at a global level. Here, we will revise recent proteome profiling studies performed in MDSCs from different origins. Moreover, we will perform an integrative functional analysis of the protein compilation derived from these large-scale proteomic studies in order to obtain a comprehensive view of MDSCs biology. Finally, we will also discuss the potential application of high-throughput proteomic approaches to study global proteome dynamics and post-translational modifications (PTMs) during the differentiation process of MDSCs that will greatly boost the identification of novel MDSC-specific therapeutic targets to apply in cancer immunotherapy.
Collapse
Affiliation(s)
- María Gato
- Immunomodulation Laboratory, Navarrabiomed, Fundación Miguel Servet, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Idoia Blanco-Luquin
- Immunomodulation Laboratory, Navarrabiomed, Fundación Miguel Servet, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Maribel Zudaire
- Immunomodulation Laboratory, Navarrabiomed, Fundación Miguel Servet, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Xabier Martínez de Morentin
- Proteomics Unit, Navarrabiomed, Fundación Miguel Servet, ProteoRed-ISCIII, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Estela Perez-Valderrama
- Proteomics Unit, Navarrabiomed, Fundación Miguel Servet, ProteoRed-ISCIII, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Aintzane Zabaleta
- Biofunctional Nanomaterials Laboratory, CIC Biomagune, San Sebastian, Spain
| | - Grazyna Kochan
- Immunomodulation Laboratory, Navarrabiomed, Fundación Miguel Servet, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - David Escors
- Immunomodulation Laboratory, Navarrabiomed, Fundación Miguel Servet, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Joaquín Fernandez-Irigoyen
- Proteomics Unit, Navarrabiomed, Fundación Miguel Servet, ProteoRed-ISCIII, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Enrique Santamaría
- Proteomics Unit, Navarrabiomed, Fundación Miguel Servet, ProteoRed-ISCIII, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| |
Collapse
|
34
|
Anzengruber F, Avci P, de Freitas LF, Hamblin MR. T-cell mediated anti-tumor immunity after photodynamic therapy: why does it not always work and how can we improve it? Photochem Photobiol Sci 2015; 14:1492-1509. [PMID: 26062987 PMCID: PMC4547550 DOI: 10.1039/c4pp00455h] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Photodynamic therapy (PDT) uses the combination of non-toxic photosensitizers and harmless light to generate reactive oxygen species that destroy tumors by a combination of direct tumor cell killing, vascular shutdown, and activation of the immune system. It has been shown in some animal models that mice that have been cured of cancer by PDT, may exhibit resistance to rechallenge. The cured mice can also possess tumor specific T-cells that recognize defined tumor antigens, destroy tumor cells in vitro, and can be adoptively transferred to protect naïve mice from cancer. However, these beneficial outcomes are the exception rather than the rule. The reasons for this lack of consistency lie in the ability of many tumors to suppress the host immune system and to actively evade immune attack. The presence of an appropriate tumor rejection antigen in the particular tumor cell line is a requisite for T-cell mediated immunity. Regulatory T-cells (CD25+, Foxp3+) are potent inhibitors of anti-tumor immunity, and their removal by low dose cyclophosphamide can potentiate the PDT-induced immune response. Treatments that stimulate dendritic cells (DC) such as CpG oligonucleotide can overcome tumor-induced DC dysfunction and improve PDT outcome. Epigenetic reversal agents can increase tumor expression of MHC class I and also simultaneously increase expression of tumor antigens. A few clinical reports have shown that anti-tumor immunity can be generated by PDT in patients, and it is hoped that these combination approaches may increase tumor cures in patients.
Collapse
Affiliation(s)
- Florian Anzengruber
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Dermatology, Harvard Medical School, Boston, MA, USA
| | - Pinar Avci
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Dermatology, Harvard Medical School, Boston, MA, USA
- Department of Dermatology, Dermatooncology and Venerology, Semmelweis University School of Medicine, Budapest, 1085, Hungary
| | - Lucas Freitas de Freitas
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Dermatology, Harvard Medical School, Boston, MA, USA
- Programa de Pos Graduacao Interunidades Bioengenharia – USP – Sao Carlos, Brazil
| | - Michael R Hamblin
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Dermatology, Harvard Medical School, Boston, MA, USA
- Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA, USA
- Correspondence to: Michael R Hamblin, PhD, Wellman Center for Photomedicine, Massachusetts General Hospital, 50 Blossom Street, Boston, MA 02114, USA.
| |
Collapse
|
35
|
Yang XN, Huang L, Chen Y, An SJ, Zhang XC, Liao RQ, Su J, Wu YL. Single nucleotide polymorphisms of MAGE-A3 gene and its clinical implications in Chinese patients with non-small cell lung cancer (NSCLC). Chin J Cancer Res 2015; 27:301-8. [PMID: 26157327 DOI: 10.3978/j.issn.1000-9604.2014.11.03] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Accepted: 01/15/2015] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Available study revealed advanced tumors have a higher expression rate of MAGE-A3 gene which has a lot of single nucleotide polymorphism (SNP) loci with polymorphisms. This study aimed to analyze the allele frequency of SNP loci in MAGE-A3 gene and investigate the relationship between MAGE-A3 gene polymorphisms and clinical factors. METHODS Tumor samples of a cohort of 191 NSCLC patients were collected. EGFR mRNA expression were detected by qRT-PCR. SNPs in whole length of MAGE-A3 gene were detected by direct sequencing. Frequencies of the SNPs were correlated to gene expression, mutation status of EGFR and clinical factors. RESULTS Sequencing analysis confirmed that allele frequencies of genotypes on SNP loci rs5970360, rs5925210, rs5970361, rs5925211 and rs35123853 were CC (0.681)/CT (0.319), CC (0.660)/CG (0.340), CC (0.681)/CA (0.319), AA (0.984)/AT (0.016) and GG (1.000)/GA (0.000), respectively, which were different from the frequencies and genotypes of MAGE-A3 in SNP database. Chi-square tests showed the EGFR mRNA expression level had significant correlation with the genotypes of SNP loci rs5970360 and rs5925210. But all frequencies of each MAGE-A3 SNPs were not found significantly different between EGFR mutant and wild type patients. MAGE-A3 gene polymorphisms had no significant effects on survival of NSCLC patients. CONCLUSIONS Chinese patients with NSCLC had different SNP patterns of MAGE-A3 in comparison with those in international SNP database. These MAGE-A3 SNP loci might have not prognostic significance. MAGE-A3 SNP loci rs5970360 and rs5925210 might be predictive for EGFR mRNA expression levels and helpful to the selection of patients for epidermal growth factor receptor (EGFR) targeted immunotherapy.
Collapse
Affiliation(s)
- Xue-Ning Yang
- Guangdong Lung Cancer Institute, Guangdong Provincial Key Laboratory of Translational Medicine in Lung Cancer, Guangdong General Hospital & Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Ling Huang
- Guangdong Lung Cancer Institute, Guangdong Provincial Key Laboratory of Translational Medicine in Lung Cancer, Guangdong General Hospital & Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Yu Chen
- Guangdong Lung Cancer Institute, Guangdong Provincial Key Laboratory of Translational Medicine in Lung Cancer, Guangdong General Hospital & Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - She-Juan An
- Guangdong Lung Cancer Institute, Guangdong Provincial Key Laboratory of Translational Medicine in Lung Cancer, Guangdong General Hospital & Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Xu-Chao Zhang
- Guangdong Lung Cancer Institute, Guangdong Provincial Key Laboratory of Translational Medicine in Lung Cancer, Guangdong General Hospital & Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Ri-Qiang Liao
- Guangdong Lung Cancer Institute, Guangdong Provincial Key Laboratory of Translational Medicine in Lung Cancer, Guangdong General Hospital & Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Jian Su
- Guangdong Lung Cancer Institute, Guangdong Provincial Key Laboratory of Translational Medicine in Lung Cancer, Guangdong General Hospital & Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Yi-Long Wu
- Guangdong Lung Cancer Institute, Guangdong Provincial Key Laboratory of Translational Medicine in Lung Cancer, Guangdong General Hospital & Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| |
Collapse
|
36
|
Immune evasion in cancer: Mechanistic basis and therapeutic strategies. Semin Cancer Biol 2015; 35 Suppl:S185-S198. [PMID: 25818339 DOI: 10.1016/j.semcancer.2015.03.004] [Citation(s) in RCA: 1092] [Impact Index Per Article: 109.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Revised: 03/10/2015] [Accepted: 03/13/2015] [Indexed: 12/27/2022]
Abstract
Cancer immune evasion is a major stumbling block in designing effective anticancer therapeutic strategies. Although considerable progress has been made in understanding how cancers evade destructive immunity, measures to counteract tumor escape have not kept pace. There are a number of factors that contribute to tumor persistence despite having a normal host immune system. Immune editing is one of the key aspects why tumors evade surveillance causing the tumors to lie dormant in patients for years through "equilibrium" and "senescence" before re-emerging. In addition, tumors exploit several immunological processes such as targeting the regulatory T cell function or their secretions, antigen presentation, modifying the production of immune suppressive mediators, tolerance and immune deviation. Besides these, tumor heterogeneity and metastasis also play a critical role in tumor growth. A number of potential targets like promoting Th1, NK cell, γδ T cell responses, inhibiting Treg functionality, induction of IL-12, use of drugs including phytochemicals have been designed to counter tumor progression with much success. Some natural agents and phytochemicals merit further study. For example, use of certain key polysaccharide components from mushrooms and plants have shown to possess therapeutic impact on tumor-imposed genetic instability, anti-growth signaling, replicative immortality, dysregulated metabolism etc. In this review, we will discuss the advances made toward understanding the basis of cancer immune evasion and summarize the efficacy of various therapeutic measures and targets that have been developed or are being investigated to enhance tumor rejection.
Collapse
|
37
|
Destexhe E, Stannard D, Wilby OK, Grosdidier E, Baudson N, Forster R, Gérard CM, Garçon N, Segal L. Nonclinical reproductive and developmental safety evaluation of the MAGE-A3 Cancer Immunotherapeutic, a therapeutic vaccine for cancer treatment. Reprod Toxicol 2014; 51:90-105. [PMID: 25530039 DOI: 10.1016/j.reprotox.2014.12.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Revised: 12/01/2014] [Accepted: 12/11/2014] [Indexed: 11/25/2022]
Abstract
We assessed potential toxic effects of the MAGE-A3 Cancer Immunotherapeutic on female fertility and embryo-fetal, pre- and post-natal development in rats and on male fertility in rats and monkeys. Three groups of 48 female (Study 1) or 22 male (Study 2) CD rats received 5 or 3 injections of 100μL of saline, AS15 immunostimulant, or MAGE-A3 Cancer Immunotherapeutic (MAGE-A3 recombinant protein combined with AS15) at various timepoints pre- or post-mating. Male Cynomolgus monkeys (Study 3) received 8 injections of 500μL of saline (n=2) or the MAGE-A3 Cancer Immunotherapeutic (n=6) every 2 weeks. Rats were sacrificed on gestation day 20 or lactation day 25 (Study 1) or 9 weeks after first injection (Study 2) and monkeys, 3 days or 8 weeks after last injection. Injections were well tolerated. Female rat mating performance or fertility, pre- and post-natal survival, offspring development up to 25 days of age, and male mating performance (rats) or fertility parameters (rats and monkeys) were unaffected.
Collapse
Affiliation(s)
- E Destexhe
- GSK Vaccines, Rue de l'Institut 89, 1330 Rixensart, Belgium
| | - D Stannard
- Huntingdon Life Sciences, Eye Research Centre, Eye, Suffolk IP23 7PX, UK
| | - O K Wilby
- Huntingdon Life Sciences, Eye Research Centre, Eye, Suffolk IP23 7PX, UK
| | | | - N Baudson
- GSK Vaccines, Rue de l'Institut 89, 1330 Rixensart, Belgium
| | - R Forster
- CiToxLAB France, BP 563, 27005 Evreux, France
| | - C M Gérard
- GSK Vaccines, Rue de l'Institut 89, 1330 Rixensart, Belgium
| | - N Garçon
- Bioaster, 321 Avenue Jean Jaurès, 69007 Lyon, France
| | - L Segal
- GSK Vaccines, Parc de la Noire Epine, Rue Fleming 20, 1300 Wavre, Belgium.
| |
Collapse
|
38
|
Abstract
Newer immunotherapy agents may break the barrier that tumors create to evade the attack from the immune system. Dendritic cell vaccination has shown encouraging clinical activity and a favorable safety profile in advanced tumor stages. However, optimal cell maturation status, choice of tumor antigens and route of administration have not been established. Single or multiple peptides derived from tumor-associated antigens may also be used for cancer vaccination. Intratumoral delivery of oncolytic viruses expressing immunostimulating cytokines like GM-CSF have produced stimulating clinical results that need further verification. But it is probably T-cell checkpoint modulation with monoclonal antibodies that has attracted the highest expectations. Promising activity has been reported for tremelimumab, a CTLA-4 inhibitor, and a clinical trial testing the PD-1 antibody nivolumab is underway. Future progress will probably come from a better understanding of the mechanisms of cancer-related immunosuppression, improvement in agents and strategies and combination of the available therapeutic tools.
Collapse
Affiliation(s)
- Bruno Sangro
- Liver Unit, Clínica Universidad de Navarra, and Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD); Avda. Pio XII 36. 31008-Pamplona, Spain.,Liver Unit, Clínica Universidad de Navarra, and Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD); Avda. Pio XII 36. 31008-Pamplona, Spain
| | - Daniel Palmer
- The Department of Molecular & Clinical Cancer Medicine, University of Liverpool, Liverpool, L69 3GA, UK.,The Department of Molecular & Clinical Cancer Medicine, University of Liverpool, Liverpool, L69 3GA, UK
| | - Ignacio Melero
- Departments of Oncology & Immunology, Centro de Investigación Médica Aplicada y Clínica Universidad de Navarra. Avda. Pio XII, 55. 31008-Pamplona, Spain.,Departments of Oncology & Immunology, Centro de Investigación Médica Aplicada y Clínica Universidad de Navarra. Avda. Pio XII, 55. 31008-Pamplona, Spain
| |
Collapse
|
39
|
Abstract
Background The majority of genetic biomarkers for human cancers are defined by statistical screening of high-throughput genomics data. While a large number of genetic biomarkers have been proposed for diagnostic and prognostic applications, only a small number have been applied in the clinic. Similarly, the use of proteomics methods for the discovery of cancer biomarkers is increasing. The emerging field of proteogenomics seeks to enrich the value of genomics and proteomics approaches by studying the intersection of genomics and proteomics data. This task is challenging due to the complex nature of transcriptional and translation regulatory mechanisms and the disparities between genomic and proteomic data from the same samples. In this study, we have examined tumor antigens as potential biomarkers for breast cancer using genomics and proteomics data from previously reported laser capture microdissected ER+ tumor samples. Results We applied proteogenomic analyses to study the genetic aberrations of 32 tumor antigens determined in the proteomic data. We found that tumor antigens that are aberrantly expressed at the genetic level and expressed at the protein level, are likely involved in perturbing pathways directly linked to the hallmarks of cancer. The results found by proteogenomic analysis of the 32 tumor antigens studied here, capture largely the same pathway irregularities as those elucidated from large-scale screening of genomics analyses, where several thousands of genes are often found to be perturbed. Conclusion Tumor antigens are a group of proteins recognized by the cells of the immune system. Specifically, they are recognized in tumor cells where they are present in larger than usual amounts, or are physiochemically altered to a degree at which they no longer resemble native human proteins. This proteogenomic analysis of 32 tumor antigens suggests that tumor antigens have the potential to be highly specific biomarkers for different cancers.
Collapse
|
40
|
Olsen LR, Campos B, Barnkob MS, Winther O, Brusic V, Andersen MH. Bioinformatics for cancer immunotherapy target discovery. Cancer Immunol Immunother 2014; 63:1235-49. [PMID: 25344903 PMCID: PMC11029190 DOI: 10.1007/s00262-014-1627-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Accepted: 10/08/2014] [Indexed: 12/13/2022]
Abstract
The mechanisms of immune response to cancer have been studied extensively and great effort has been invested into harnessing the therapeutic potential of the immune system. Immunotherapies have seen significant advances in the past 20 years, but the full potential of protective and therapeutic cancer immunotherapies has yet to be fulfilled. The insufficient efficacy of existing treatments can be attributed to a number of biological and technical issues. In this review, we detail the current limitations of immunotherapy target selection and design, and review computational methods to streamline therapy target discovery in a bioinformatics analysis pipeline. We describe specialized bioinformatics tools and databases for three main bottlenecks in immunotherapy target discovery: the cataloging of potentially antigenic proteins, the identification of potential HLA binders, and the selection epitopes and co-targets for single-epitope and multi-epitope strategies. We provide examples of application to the well-known tumor antigen HER2 and suggest bioinformatics methods to ameliorate therapy resistance and ensure efficient and lasting control of tumors.
Collapse
Affiliation(s)
- Lars Rønn Olsen
- Department of Biology, Bioinformatics Centre, University of Copenhagen, Ole Maaløes Vej 5, 2200, Copenhagen, Denmark,
| | | | | | | | | | | |
Collapse
|
41
|
Xia Y, Gupta GK, Castano AP, Mroz P, Avci P, Hamblin MR. CpG oligodeoxynucleotide as immune adjuvant enhances photodynamic therapy response in murine metastatic breast cancer. JOURNAL OF BIOPHOTONICS 2014; 7:897-905. [PMID: 23922221 PMCID: PMC3917974 DOI: 10.1002/jbio.201300072] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2013] [Revised: 06/28/2013] [Accepted: 07/06/2013] [Indexed: 05/07/2023]
Abstract
Breast cancer is the most common cause of cancer death in women. The side effects and complications following current breast cancer therapy can be devastating. Moreover, the prognosis in late stages of the diseases is usually poor. Photodynamic therapy (PDT) is a promising cancer treatment modality that is capable of both local tumor destruction and immune stimulation. However, treatment with PDT alone is often non-curative due to tumor-induced immune cell dysfunction and immune suppression. This phenomenon has motivated a new approach by combining immunostimulants with PDT to enhance anti-tumor immunity. In the present study, we investigated PDT mediated by verteporfin and 690 nm light delivered 15 min later, in combination with an immunomodulation approach using CpG oligodeoxynucleotide for the treatment of 4T1 metastatic breast cancer in a BALB/c immunocompetent mouse model. In vitro, CpG primed immature dendritic cells (DC) via toll like receptor 9 to phagocytose PDT killed tumor cells leading to DC maturation and activation. Peritumoral injection of CpG after PDT in mice gave improved local tumor control and a survival advantage compared to either treatment alone (p < 0.05). CpG may be a valuable dendritic cell targeted immunoadjuvant to combine with PDT.
Collapse
Affiliation(s)
- Yumin Xia
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Dermatology, Harvard Medical School, Boston, MA, USA
| | - Gaurav K. Gupta
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Dermatology, Harvard Medical School, Boston, MA, USA
| | - Ana P. Castano
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Dermatology, Harvard Medical School, Boston, MA, USA
| | - Pawel Mroz
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Dermatology, Harvard Medical School, Boston, MA, USA
| | - Pinar Avci
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Dermatology, Harvard Medical School, Boston, MA, USA
- Department of Dermatology, Dermatooncology and Venerology, Semmelweis University School of Medicine, Budapest, 1085, Hungary
| | - Michael R. Hamblin
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Dermatology, Harvard Medical School, Boston, MA, USA
- Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA, USA
| |
Collapse
|
42
|
van Duikeren S, Arens R. Predicting the efficacy of cancer vaccines by evaluating T-cell responses. Oncoimmunology 2014; 2:e22616. [PMID: 23482345 PMCID: PMC3583929 DOI: 10.4161/onci.22616] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
In a preclinical experimental model of human papilloma virus (HPV)-induced cervical carcinoma, we have shown that the efficacy of cancer vaccines can be predicted by the evaluation of vaccine-induced T-cell responses in healthy subjects. We argue that such knowledge can be used to screen candidates for vaccination, which in turn may accelerate the development and increase the overall efficacy of cancer vaccines.
Collapse
Affiliation(s)
- Suzanne van Duikeren
- Department of Immunohematology and Blood Transfusion; Leiden University Medical Center; Leiden, The Netherlands
| | | |
Collapse
|
43
|
Martín J, Pinazo I, Mateo J, Escandell I, Jordá E, Monteagudo C. Evaluación de la regresión en melanomas primarios sucesivos. ACTAS DERMO-SIFILIOGRAFICAS 2014; 105:768-73. [DOI: 10.1016/j.ad.2014.01.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2013] [Revised: 12/03/2013] [Accepted: 01/07/2014] [Indexed: 10/25/2022] Open
|
44
|
Assessment of Regression in Successive Primary Melanomas. ACTAS DERMO-SIFILIOGRAFICAS 2014. [DOI: 10.1016/j.adengl.2014.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
45
|
Destexhe E, Grosdidier E, Baudson N, Forster R, Gerard C, Garçon N, Segal L. Non‐clinical safety evaluation of single and repeated intramuscular administrations of MAGE‐A3 Cancer Immunotherapeutic in rabbits and cynomolgus monkeys. J Appl Toxicol 2014; 35:717-28. [DOI: 10.1002/jat.3025] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Revised: 03/14/2014] [Accepted: 04/02/2014] [Indexed: 12/24/2022]
Affiliation(s)
- Eric Destexhe
- GlaxoSmithKline Vaccines 1330 Rixensart/Wavre Belgium
| | | | | | | | | | | | | |
Collapse
|
46
|
Song H, Zhu J, Suo C, Lu D. Immunotherapy for stage I-III non-small cell lung cancer treated with surgery or radiotherapy with curative intent. Hippokratia 2014. [DOI: 10.1002/14651858.cd011300] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Huan Song
- Karolinska Institutet; Department of Medical Epidemiology and Biostatistics; Box 281 Stockholm Sweden SE-17177
| | - Jianwei Zhu
- Shandong Provincial Hospital Affiliated to Shandong University; Department of Orthopaedics; No.324, Jingwu Road Jinan Shandong China 250021
| | - Chen Suo
- Karolinska Institutet; Department of Medical Epidemiology and Biostatistics; Box 281 Stockholm Sweden SE-17177
| | - DongHao Lu
- Karolinska Institutet; Department of Medical Epidemiology and Biostatistics; Box 281 Stockholm Sweden SE-17177
| |
Collapse
|
47
|
Tao Y, Lin F, Li T, Xie J, Shen C, Zhu Z. Epigenetically Modified Pancreatic Carcinoma PANC-1 Cells Can Act as Cancer Vaccine to Enhance Antitumor Immune Response in Mice. Oncol Res 2014; 21:307-16. [DOI: 10.3727/096504014x13983417587320] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
|
48
|
Big data analytics in immunology: a knowledge-based approach. BIOMED RESEARCH INTERNATIONAL 2014; 2014:437987. [PMID: 25045677 PMCID: PMC4090507 DOI: 10.1155/2014/437987] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Accepted: 05/07/2014] [Indexed: 01/27/2023]
Abstract
With the vast amount of immunological data available, immunology research is entering the big data era. These data vary in granularity, quality, and complexity and are stored in various formats, including publications, technical reports, and databases. The challenge is to make the transition from data to actionable knowledge and wisdom and bridge the knowledge gap and application gap. We report a knowledge-based approach based on a framework called KB-builder that facilitates data mining by enabling fast development and deployment of web-accessible immunological data knowledge warehouses. Immunological knowledge discovery relies heavily on both the availability of accurate, up-to-date, and well-organized data and the proper analytics tools. We propose the use of knowledge-based approaches by developing knowledgebases combining well-annotated data with specialized analytical tools and integrating them into analytical workflow. A set of well-defined workflow types with rich summarization and visualization capacity facilitates the transformation from data to critical information and knowledge. By using KB-builder, we enabled streamlining of normally time-consuming processes of database development. The knowledgebases built using KB-builder will speed up rational vaccine design by providing accurate and well-annotated data coupled with tailored computational analysis tools and workflow.
Collapse
|
49
|
Mount DW, Putnam CW, Centouri SM, Manziello AM, Pandey R, Garland LL, Martinez JD. Using logistic regression to improve the prognostic value of microarray gene expression data sets: application to early-stage squamous cell carcinoma of the lung and triple negative breast carcinoma. BMC Med Genomics 2014; 7:33. [PMID: 24916928 PMCID: PMC4110620 DOI: 10.1186/1755-8794-7-33] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2013] [Accepted: 05/27/2014] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Numerous microarray-based prognostic gene expression signatures of primary neoplasms have been published but often with little concurrence between studies, thus limiting their clinical utility. We describe a methodology using logistic regression, which circumvents limitations of conventional Kaplan Meier analysis. We applied this approach to a thrice-analyzed and published squamous cell carcinoma (SQCC) of the lung data set, with the objective of identifying gene expressions predictive of early death versus long survival in early-stage disease. A similar analysis was applied to a data set of triple negative breast carcinoma cases, which present similar clinical challenges. METHODS Important to our approach is the selection of homogenous patient groups for comparison. In the lung study, we selected two groups (including only stages I and II), equal in size, of earliest deaths and longest survivors. Genes varying at least four-fold were tested by logistic regression for accuracy of prediction (area under a ROC plot). The gene list was refined by applying two sliding-window analyses and by validations using a leave-one-out approach and model building with validation subsets. In the breast study, a similar logistic regression analysis was used after selecting appropriate cases for comparison. RESULTS A total of 8594 variable genes were tested for accuracy in predicting earliest deaths versus longest survivors in SQCC. After applying the two sliding window and the leave-one-out analyses, 24 prognostic genes were identified; most of them were B-cell related. When the same data set of stage I and II cases was analyzed using a conventional Kaplan Meier (KM) approach, we identified fewer immune-related genes among the most statistically significant hits; when stage III cases were included, most of the prognostic genes were missed. Interestingly, logistic regression analysis of the breast cancer data set identified many immune-related genes predictive of clinical outcome. CONCLUSIONS Stratification of cases based on clinical data, careful selection of two groups for comparison, and the application of logistic regression analysis substantially improved predictive accuracy in comparison to conventional KM approaches. B cell-related genes dominated the list of prognostic genes in early stage SQCC of the lung and triple negative breast cancer.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Jesse D Martinez
- Department of Cellular and Molecular Medicine, Arizona Health Sciences Center, The University of Arizona, Tucson, Arizona 85735, USA.
| |
Collapse
|
50
|
Molecular targeted therapy for early-stage non-small-cell lung cancer: Will it increase the cure rate? Lung Cancer 2014; 84:97-100. [DOI: 10.1016/j.lungcan.2014.01.018] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2014] [Accepted: 01/21/2014] [Indexed: 11/22/2022]
|