1
|
Bakkerus L, Subtil B, Bontkes HJ, Gootjes EC, Reijm M, Vullings M, Verrijp K, Bokhorst JM, Woortman C, Nagtegaal ID, Jonker MA, van der Vliet HJ, Verhoef C, Gorris MA, de Vries IJM, de Gruijl TD, Verheul HM, Buffart TE, Tauriello DVF. Exploring immune status in peripheral blood and tumor tissue in association with survival in patients with multi-organ metastatic colorectal cancer. Oncoimmunology 2024; 13:2361971. [PMID: 38868078 PMCID: PMC11168219 DOI: 10.1080/2162402x.2024.2361971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 05/27/2024] [Indexed: 06/14/2024] Open
Abstract
Colorectal cancer (CRC) raises considerable clinical challenges, including a high mortality rate once the tumor spreads to distant sites. At this advanced stage, more accurate prediction of prognosis and treatment outcome is urgently needed. The role of cancer immunity in metastatic CRC (mCRC) is poorly understood. Here, we explore cellular immune cell status in patients with multi-organ mCRC. We analyzed T cell infiltration in primary tumor sections, surveyed the lymphocytic landscape of liver metastases, and assessed circulating mononuclear immune cells. Besides asking whether immune cells are associated with survival at this stage of the disease, we investigated correlations between the different tissue types; as this could indicate a dominant immune phenotype. Taken together, our analyses corroborate previous observations that higher levels of CD8+ T lymphocytes link to better survival outcomes. Our findings therefore extend evidence from earlier stages of CRC to indicate an important role for cancer immunity in disease control even after metastatic spreading to multiple organs. This finding may help to improve predicting outcome of patients with mCRC and suggests a future role for immunotherapeutic strategies.
Collapse
Affiliation(s)
- Lotte Bakkerus
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Beatriz Subtil
- Department of Medical BioSciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Hetty J. Bontkes
- Department Laboratory Medicine, LGDO, Section Medical Immunology, Amsterdam, The Netherlands
| | - Elske C. Gootjes
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Martine Reijm
- Department Laboratory Medicine, LGDO, Section Medical Immunology, Amsterdam, The Netherlands
| | - Manon Vullings
- Department of Medical BioSciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Kiek Verrijp
- Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - John-Melle Bokhorst
- Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Carmen Woortman
- Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Iris D. Nagtegaal
- Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Marianne A. Jonker
- Department of IQ Health, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Hans J. van der Vliet
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, Location VUmc, Amsterdam, The Netherlands
| | - Cornelis Verhoef
- Department of Surgery, ErasmusMC Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Mark A.J. Gorris
- Department of Medical BioSciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - I. Jolanda M. de Vries
- Department of Medical BioSciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Tanja D. de Gruijl
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, Location VUmc, Amsterdam, The Netherlands
| | - Henk M.W. Verheul
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Medical Oncology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, The Netherlands
| | - Tineke E. Buffart
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, Location VUmc, Amsterdam, The Netherlands
| | - Daniele V. F. Tauriello
- Department of Medical BioSciences, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Medical Oncology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, The Netherlands
| |
Collapse
|
2
|
Li N, Zhang X, Zhang Y, Yang F, Zhou F. Study of PD-1 Customization and Autoimmune T Cells for Advanced Colorectal Cancer with High MSI Expression. CONTRAST MEDIA & MOLECULAR IMAGING 2022; 2022:6390924. [PMID: 35965619 PMCID: PMC9357754 DOI: 10.1155/2022/6390924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 06/11/2022] [Accepted: 06/17/2022] [Indexed: 11/17/2022]
Abstract
Objective To evaluate the significance of PD-1 customization and autoimmune T-cell therapy for advanced colorectal cancer with high MSI expression. Methods One hundred and eight patients with advanced colorectal cancer with high MSI expression admitted to our hospital between August 2019 and January 2022 were divided into control and study groups, and PD-1 customization and autoimmune T-cell therapy were administered to the two groups, respectively. Trends in immune indexes, PD-1 exposure, and survival rates were studied in both groups. Results The treatment efficiency of the study group was 90.74%, which was higher than that of the control group (61.11%) (P < 0. 05); after treatment, the presence of CDl07a, perforin, and GranB cells was significantly higher in both groups compared with that before treatment, but the expression of PD-1 was more pronounced in the study group (P < 0. 05); that is, the expression of PD-1 in peripheral T lymphocytes in the study group compared with that of the control group was higher in patients with grade III-IV, and peripheral T lymphocytes were also higher in patients with grade III-IV compared with patients with grade I-II (P < 0. 05). Conclusion PD-1 customization combined with autoimmune T-cell therapy is a novel therapeutic modality that can substantially improve.
Collapse
Affiliation(s)
- Na Li
- Hengshui City People's Hospital, Hengshui 053000, Hebei, China
| | - Xiaojie Zhang
- Hengshui City People's Hospital, Hengshui 053000, Hebei, China
| | - Yinsong Zhang
- Hengshui City People's Hospital, Hengshui 053000, Hebei, China
| | - Fang Yang
- Hengshui City People's Hospital, Hengshui 053000, Hebei, China
| | - Fengju Zhou
- Hengshui City People's Hospital, Hengshui 053000, Hebei, China
| |
Collapse
|
3
|
Bayless NL, Bluestone JA, Bucktrout S, Butterfield LH, Jaffee EM, Koch CA, Roep BO, Sharpe AH, Murphy WJ, Villani AC, Walunas TL. Development of preclinical and clinical models for immune-related adverse events following checkpoint immunotherapy: a perspective from SITC and AACR. J Immunother Cancer 2021; 9:e002627. [PMID: 34479924 PMCID: PMC8420733 DOI: 10.1136/jitc-2021-002627] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/14/2021] [Indexed: 12/17/2022] Open
Abstract
Recent advances in cancer immunotherapy have completely revolutionized cancer treatment strategies. Nonetheless, the increasing incidence of immune-related adverse events (irAEs) is now limiting the overall benefits of these treatments. irAEs are well-recognized side effects of some of the most effective cancer immunotherapy agents, including antibody blockade of the cytotoxic T-lymphocyte-associated protein 4 and programmed death protein 1/programmed-death ligand 1 pathways. To develop an action plan on the key elements needed to unravel and understand the key mechanisms driving irAEs, the Society for Immunotherapy for Cancer and the American Association for Cancer Research partnered to bring together research and clinical experts in cancer immunotherapy, autoimmunity, immune regulation, genetics and informatics who are investigating irAEs using animal models, clinical data and patient specimens to discuss current strategies and identify the critical next steps needed to create breakthroughs in our understanding of these toxicities. The genetic and environmental risk factors, immune cell subsets and other key immunological mediators and the unique clinical presentations of irAEs across the different organ systems were the foundation for identifying key opportunities and future directions described in this report. These include the pressing need for significantly improved preclinical model systems, broader collection of biospecimens with standardized collection and clinical annotation made available for research and integration of electronic health record and multiomic data with harmonized and standardized methods, definitions and terminologies to further our understanding of irAE pathogenesis. Based on these needs, this report makes a set of recommendations to advance our understanding of irAE mechanisms, which will be crucial to prevent their occurrence and improve their treatment.
Collapse
Affiliation(s)
- Nicholas L Bayless
- Parker Institute for Cancer Immunotherapy, San Francisco, California, USA
| | - Jeffrey A Bluestone
- Diabetes Center, University of California San Francisco, San Francisco, California, USA
| | - Samantha Bucktrout
- Parker Institute for Cancer Immunotherapy, San Francisco, California, USA
| | - Lisa H Butterfield
- Parker Institute for Cancer Immunotherapy, San Francisco, California, USA
- Microbiology and Immunology, University of California San Francisco, San Francisco, California, USA
| | - Elizabeth M Jaffee
- Johns Hopkins Medicine Sidney Kimmel Comprehensive Cancer Center, Baltimore, Maryland, USA
| | | | - Bart O Roep
- Department of Diabetes Immunology, Diabetes & Metabolism Research Institute at the Beckman Research Institute, City of Hope National Medical Center, Duarte, California, USA
| | - Arlene H Sharpe
- Department of Immunology, Blavatnik Institute, Harvard Medical School and Evergrande Center for Immunologic Diseases, Harvard Medical School, Boston, Massachusetts, USA
| | - William J Murphy
- Department of Dermatology, Institute for Regenerative Cures, University of California Davis, Sacramento, California, USA
| | - Alexandra-Chloé Villani
- Center for Cancer Research, Center for Immunology and Inflammatory Diseases, Department of Medicine, Harvard Medical School, Massachusetts General Hospital, Boston, Massachusetts, USA
- Broad Institute, Cambridge, Massachusetts, USA
| | - Theresa L Walunas
- Department of Medicine and Center for Health Information Partnerships, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| |
Collapse
|
4
|
Li J, Shi Z, Liu F, Fang X, Cao K, Meng Y, Zhang H, Yu J, Feng X, Li Q, Liu Y, Wang L, Jiang H, Lu J, Shao C, Bian Y. XGBoost Classifier Based on Computed Tomography Radiomics for Prediction of Tumor-Infiltrating CD8 + T-Cells in Patients With Pancreatic Ductal Adenocarcinoma. Front Oncol 2021; 11:671333. [PMID: 34094971 PMCID: PMC8170309 DOI: 10.3389/fonc.2021.671333] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 04/26/2021] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVES This study constructed and validated a machine learning model to predict CD8+ tumor-infiltrating lymphocyte expression levels in patients with pancreatic ductal adenocarcinoma (PDAC) using computed tomography (CT) radiomic features. MATERIALS AND METHODS In this retrospective study, 184 PDAC patients were randomly assigned to a training dataset (n =137) and validation dataset (n =47). All patients were divided into CD8+ T-high and -low groups using X-tile plots. A total of 1409 radiomics features were extracted from the segmentation of regions of interest, based on preoperative CT images of each patient. The LASSO algorithm was applied to reduce the dimensionality of the data and select features. The extreme gradient boosting classifier (XGBoost) was developed using a training set consisting of 137 consecutive patients admitted between January 2017 and December 2017. The model was validated in 47 consecutive patients admitted between January 2018 and April 2018. The performance of the XGBoost classifier was determined by its discriminative ability, calibration, and clinical usefulness. RESULTS The cut-off value of the CD8+ T-cell level was 18.69%, as determined by the X-tile program. A Kaplan-Meier analysis indicated a correlation between higher CD8+ T-cell levels and better overall survival (p = 0.001). The XGBoost classifier showed good discrimination in the training set (area under curve [AUC], 0.75; 95% confidence interval [CI]: 0.67-0.83) and validation set (AUC, 0.67; 95% CI: 0.51-0.83). Moreover, it showed a good calibration. The sensitivity, specificity, accuracy, positive and negative predictive values were 80.65%, 60.00%, 0.69, 0.63, and 0.79, respectively, for the training set, and 80.95%, 57.69%, 0.68, 0.61, and 0.79, respectively, for the validation set. CONCLUSIONS We developed a CT-based XGBoost classifier to extrapolate the infiltration levels of CD8+ T-cells in patients with PDAC. This method could be useful in identifying potential patients who can benefit from immunotherapies.
Collapse
Affiliation(s)
- Jing Li
- Department of Radiology, Changhai Hospital, Navy Medical University, Shanghai, China
| | - Zhang Shi
- Department of Radiology, Changhai Hospital, Navy Medical University, Shanghai, China
| | - Fang Liu
- Department of Radiology, Changhai Hospital, Navy Medical University, Shanghai, China
| | - Xu Fang
- Department of Radiology, Changhai Hospital, Navy Medical University, Shanghai, China
| | - Kai Cao
- Department of Radiology, Changhai Hospital, Navy Medical University, Shanghai, China
| | - Yinghao Meng
- Department of Radiology, Changhai Hospital, Navy Medical University, Shanghai, China
| | - Hao Zhang
- Department of Radiology, Changhai Hospital, Navy Medical University, Shanghai, China
| | - Jieyu Yu
- Department of Radiology, Changhai Hospital, Navy Medical University, Shanghai, China
| | - Xiaochen Feng
- Department of Radiology, Changhai Hospital, Navy Medical University, Shanghai, China
| | - Qi Li
- Department of Radiology, Changhai Hospital, Navy Medical University, Shanghai, China
| | - Yanfang Liu
- Department of Pathology, Changhai Hospital, Navy Medical University, Shanghai, China
| | - Li Wang
- Department of Radiology, Changhai Hospital, Navy Medical University, Shanghai, China
| | - Hui Jiang
- Department of Pathology, Changhai Hospital, Navy Medical University, Shanghai, China
| | - Jianping Lu
- Department of Radiology, Changhai Hospital, Navy Medical University, Shanghai, China
| | - Chengwei Shao
- Department of Radiology, Changhai Hospital, Navy Medical University, Shanghai, China
| | - Yun Bian
- Department of Radiology, Changhai Hospital, Navy Medical University, Shanghai, China
| |
Collapse
|
5
|
Hu-Lieskovan S, Bhaumik S, Dhodapkar K, Grivel JCJB, Gupta S, Hanks BA, Janetzki S, Kleen TO, Koguchi Y, Lund AW, Maccalli C, Mahnke YD, Novosiadly RD, Selvan SR, Sims T, Zhao Y, Maecker HT. SITC cancer immunotherapy resource document: a compass in the land of biomarker discovery. J Immunother Cancer 2020; 8:e000705. [PMID: 33268350 PMCID: PMC7713206 DOI: 10.1136/jitc-2020-000705] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/15/2020] [Indexed: 02/07/2023] Open
Abstract
Since the publication of the Society for Immunotherapy of Cancer's (SITC) original cancer immunotherapy biomarkers resource document, there have been remarkable breakthroughs in cancer immunotherapy, in particular the development and approval of immune checkpoint inhibitors, engineered cellular therapies, and tumor vaccines to unleash antitumor immune activity. The most notable feature of these breakthroughs is the achievement of durable clinical responses in some patients, enabling long-term survival. These durable responses have been noted in tumor types that were not previously considered immunotherapy-sensitive, suggesting that all patients with cancer may have the potential to benefit from immunotherapy. However, a persistent challenge in the field is the fact that only a minority of patients respond to immunotherapy, especially those therapies that rely on endogenous immune activation such as checkpoint inhibitors and vaccination due to the complex and heterogeneous immune escape mechanisms which can develop in each patient. Therefore, the development of robust biomarkers for each immunotherapy strategy, enabling rational patient selection and the design of precise combination therapies, is key for the continued success and improvement of immunotherapy. In this document, we summarize and update established biomarkers, guidelines, and regulatory considerations for clinical immune biomarker development, discuss well-known and novel technologies for biomarker discovery and validation, and provide tools and resources that can be used by the biomarker research community to facilitate the continued development of immuno-oncology and aid in the goal of durable responses in all patients.
Collapse
Affiliation(s)
- Siwen Hu-Lieskovan
- Huntsman Cancer Institute, Salt Lake City, UT, USA
- University of Utah School of Medicine, Salt Lake City, UT, USA
| | | | - Kavita Dhodapkar
- Department of Pediatrics, Emory University, Atlanta, Georgia, USA
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, Georgia, USA
| | | | - Sumati Gupta
- Huntsman Cancer Institute, Salt Lake City, Utah, USA
| | - Brent A Hanks
- Duke University Medical Center, Durham, North Carolina, USA
| | | | | | - Yoshinobu Koguchi
- Earle A Chiles Research Institute, Providence Cancer Institute, Portland, Oregon, USA
| | - Amanda W Lund
- Oregon Health and Science University, Portland, Oregon, USA
| | | | | | | | | | - Tasha Sims
- Regeneron Pharmaceuticals Inc, Tarrytown, New York, USA
| | | | | |
Collapse
|
6
|
Swann JR, Rajilic-Stojanovic M, Salonen A, Sakwinska O, Gill C, Meynier A, Fança-Berthon P, Schelkle B, Segata N, Shortt C, Tuohy K, Hasselwander O. Considerations for the design and conduct of human gut microbiota intervention studies relating to foods. Eur J Nutr 2020; 59:3347-3368. [PMID: 32246263 PMCID: PMC7669793 DOI: 10.1007/s00394-020-02232-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Accepted: 03/18/2020] [Indexed: 12/15/2022]
Abstract
With the growing appreciation for the influence of the intestinal microbiota on human health, there is increasing motivation to design and refine interventions to promote favorable shifts in the microbiota and their interactions with the host. Technological advances have improved our understanding and ability to measure this indigenous population and the impact of such interventions. However, the rapid growth and evolution of the field, as well as the diversity of methods used, parameters measured and populations studied, make it difficult to interpret the significance of the findings and translate their outcomes to the wider population. This can prevent comparisons across studies and hinder the drawing of appropriate conclusions. This review outlines considerations to facilitate the design, implementation and interpretation of human gut microbiota intervention studies relating to foods based upon our current understanding of the intestinal microbiota, its functionality and interactions with the human host. This includes parameters associated with study design, eligibility criteria, statistical considerations, characterization of products and the measurement of compliance. Methodologies and markers to assess compositional and functional changes in the microbiota, following interventions are discussed in addition to approaches to assess changes in microbiota-host interactions and host responses. Last, EU legislative aspects in relation to foods and health claims are presented. While it is appreciated that the field of gastrointestinal microbiology is rapidly evolving, such guidance will assist in the design and interpretation of human gut microbiota interventional studies relating to foods.
Collapse
Affiliation(s)
- J. R. Swann
- Division of Integrative Systems Medicine and Digestive Diseases, Imperial College London, London, UK
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK
| | - M. Rajilic-Stojanovic
- Department for Biochemical Engineering and Biotechnology, Faculty of Technology and Metallurgy, University of Belgrade, Belgrade, Serbia
| | - A. Salonen
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - O. Sakwinska
- Société Des Produits Nestlé S.A, Nestlé Research, Lausanne, Switzerland
| | - C. Gill
- Nutrition Innovation Centre for Food and Health, Centre for Molecular Biosciences, Ulster University, Londonderry, Northern Ireland, UK
| | | | | | | | - N. Segata
- Department CIBIO, University of Trento, Trento, Italy
| | - C. Shortt
- Johnson & Johnson Consumer Services EAME Ltd., Foundation Park, Maidenhead, UK
| | - K. Tuohy
- Department of Food Quality and Nutrition, Research and Innovation Centre, Fondazione Edmund Mach, Trento, Italy
| | - O. Hasselwander
- DuPont Nutrition and Biosciences, c/o Danisco (UK) Limited, Reigate, UK
| |
Collapse
|
7
|
Repurposing Food and Drug Administration-Approved Drugs to Promote Antitumor Immunity. ACTA ACUST UNITED AC 2020; 25:88-99. [PMID: 30896530 DOI: 10.1097/ppo.0000000000000368] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
There has been a major resurgence of interest in immune-based approaches to treat cancer, based largely on the success of checkpoint inhibitors (anti-cytotoxic T-lymphocyte-associated antigen 4, anti-programmed cell death 1, and anti-programmed cell death ligand 1 antibodies) in several malignancies. However, not all tumors respond to checkpoint therapy, and there is clearly a need for additional approaches for enhancing tumor immunity. We summarize the critical elements necessary for mounting an efficacious T-cell response to a tumor. We cite drugs approved by the Food and Drug Administration for no-cancer indications that could be repurposed and used as part of an antitumor immune cocktail. We also list cancer drugs not initially intended to impact tumor immunity (soft repurposing) but that have been found to modulate the immune system. We highlight those drugs that might be used in combination with checkpoint inhibitors to increase response rates and survival of cancer patients. Our focus will be on drugs for which there are limited but existing human data. We cite supporting mechanistic mouse data as well. Repurposing drugs to modulate antitumor immunity is an opportunity to rapidly bring new, effective, and affordable treatments to cancer patients.
Collapse
|
8
|
Huang S, Zhang B, Fan W, Zhao Q, Yang L, Xin W, Fu D. Identification of prognostic genes in the acute myeloid leukemia microenvironment. Aging (Albany NY) 2019; 11:10557-10580. [PMID: 31740623 PMCID: PMC6914404 DOI: 10.18632/aging.102477] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Accepted: 11/08/2019] [Indexed: 12/27/2022]
Abstract
The tumor microenvironment (TME) has a strong influence on the progression, therapeutic response, and clinical outcome of acute myeloid leukemia (AML), one of the most common hematopoietic malignancies in adults. In this study, we identified TME-related genes associated with AML prognosis. Gene expression profiles from AML patients were downloaded from TCGA database, and immune and stromal scores were calculated using the ESTIMATE algorithm. Immune scores were correlated with clinical features such as FAB subtypes and patient's age. After categorizing AML cases into high and low score groups, an association between several differentially expressed genes (DEGs) and overall survival was identified. Functional enrichment analysis of the DEGs showed that they were primarily enriched in the immune response, inflammatory response, and cytokine activity, and were involved in signaling processes related to hematopoietic cell lineage, B cell receptor, and chemokine pathways. Two significant modules, dominated respectively by CCR5 and ITGAM nodes, were identified from the PPI network, and 20 hub genes were extracted. A total of 112 DEGs correlated with poor overall survival of AML patients, and 11 of those genes were validated in a separate TARGET-AML cohort. By identifying TME-associated genes, our findings may lead to improved prognoses and therapies for AML.
Collapse
Affiliation(s)
- Shaoxin Huang
- School of Basic Medicine, Jiujiang University, Jiujiang, Jiangxi 332005, China
| | - Biyu Zhang
- School of Pharmacy and Life Science, Jiujiang University, Jiujiang, Jiangxi 332005, China
| | - Wenyan Fan
- School of Basic Medicine, Jiujiang University, Jiujiang, Jiangxi 332005, China
| | - Qihan Zhao
- School of Basic Medicine, Jiujiang University, Jiujiang, Jiangxi 332005, China
| | - Lei Yang
- Key Laboratory of System Bio-medicine of Jiangxi Province, Jiujiang University, Jiujiang, Jiangxi 332000, China
| | - Wang Xin
- School of Basic Medicine, Jiujiang University, Jiujiang, Jiangxi 332005, China
| | - Denggang Fu
- School of Basic Medicine, Jiujiang University, Jiujiang, Jiangxi 332005, China
- Institute of Genomic and Personalized Medicine, School of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| |
Collapse
|
9
|
Caplan H, Olson SD, Kumar A, George M, Prabhakara KS, Wenzel P, Bedi S, Toledano-Furman NE, Triolo F, Kamhieh-Milz J, Moll G, Cox CS. Mesenchymal Stromal Cell Therapeutic Delivery: Translational Challenges to Clinical Application. Front Immunol 2019; 10:1645. [PMID: 31417542 PMCID: PMC6685059 DOI: 10.3389/fimmu.2019.01645] [Citation(s) in RCA: 209] [Impact Index Per Article: 34.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Accepted: 07/02/2019] [Indexed: 12/12/2022] Open
Abstract
For several decades, multipotent mesenchymal stromal cells (MSCs) have been extensively studied for their therapeutic potential across a wide range of diseases. In the preclinical setting, MSCs demonstrate consistent ability to promote tissue healing, down-regulate excessive inflammation and improve outcomes in animal models. Several proposed mechanisms of action have been posited and demonstrated across an array of in vitro models. However, translation into clinical practice has proven considerably more difficult. A number of prominent well-funded late-phase clinical trials have failed, thus calling out for new efforts to optimize product delivery in the clinical setting. In this review, we discuss novel topics critical to the successful translation of MSCs from pre-clinical to clinical applications. In particular, we focus on the major routes of cell delivery, aspects related to hemocompatibility, and potential safety concerns associated with MSC therapy in the different settings.
Collapse
Affiliation(s)
- Henry Caplan
- Department of Pediatric Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Scott D. Olson
- Department of Pediatric Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Akshita Kumar
- Department of Pediatric Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Mitchell George
- Department of Pediatric Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Karthik S. Prabhakara
- Department of Pediatric Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Pamela Wenzel
- Department of Pediatric Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Supinder Bedi
- Department of Pediatric Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Naama E. Toledano-Furman
- Department of Pediatric Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Fabio Triolo
- Department of Pediatric Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Julian Kamhieh-Milz
- Department of Transfusion Medicine, Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Guido Moll
- BIH Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Charles S. Cox
- Department of Pediatric Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| |
Collapse
|
10
|
Huang F, Wang K, Shen J. Lipoprotein-associated phospholipase A2: The story continues. Med Res Rev 2019; 40:79-134. [PMID: 31140638 PMCID: PMC6973114 DOI: 10.1002/med.21597] [Citation(s) in RCA: 113] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 04/20/2019] [Accepted: 04/30/2019] [Indexed: 12/15/2022]
Abstract
Inflammation is thought to play an important role in the pathogenesis of vascular diseases. Lipoprotein-associated phospholipase A2 (Lp-PLA2) mediates vascular inflammation through the regulation of lipid metabolism in blood, thus, it has been extensively investigated to identify its role in vascular inflammation-related diseases, mainly atherosclerosis. Although darapladib, the most advanced Lp-PLA2 inhibitor, failed to meet the primary endpoints of two large phase III trials in atherosclerosis patients cotreated with standard medical care, the research on Lp-PLA2 has not been terminated. Novel pathogenic, epidemiologic, genetic, and crystallographic studies regarding Lp-PLA2 have been reported recently, while novel inhibitors were identified through a fragment-based lead discovery strategy. More strikingly, recent clinical and preclinical studies revealed that Lp-PLA2 inhibition showed promising therapeutic effects in diabetic macular edema and Alzheimer's disease. In this review, we not only summarized the knowledge of Lp-PLA2 established in the past decades but also emphasized new findings in recent years. We hope this review could be valuable for helping researchers acquire a much deeper insight into the nature of Lp-PLA2, identify more potent and selective Lp-PLA2 inhibitors, and discover the potential indications of Lp-PLA2 inhibitors.
Collapse
Affiliation(s)
- Fubao Huang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences, Shanghai, China.,School of Pharmacy, University of Chinese Academy of Sciences, Beijing, China
| | - Kai Wang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences, Shanghai, China
| | - Jianhua Shen
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
11
|
Maio M, Coukos G, Ferrone S, Fox BA, Fridman WH, Garcia PL, Lahn M, Provendier O, Russo V, Rüttinger D, Shalabi A, Trajanoski Z, Viallet J, Wolchok JD, Ibrahim R. Addressing current challenges and future directions in immuno-oncology: expert perspectives from the 2017 NIBIT Foundation Think Tank, Siena, Italy. Cancer Immunol Immunother 2019; 68:1-9. [PMID: 30564889 PMCID: PMC11028087 DOI: 10.1007/s00262-018-2285-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Accepted: 12/08/2018] [Indexed: 11/30/2022]
Abstract
A collaborative think tank involving panellists from immuno-oncology networks, clinical/translational investigators and the pharmaceutical industry was held in Siena, Italy, in October 2017 to discuss the evolving immune-oncology landscape, identify selected key challenges, and provide a perspective on the next steps required in the translation of current research and knowledge to clinical reality. While there is a trend of combining new agents (e.g., co-stimulator agonists) with a PD-1/PD-L1 treatment backbone, use of alternative combination therapy approaches should also be considered. While the rapid evolution in systems biology provides a deeper understanding of tumor and tumor microenvironment heterogeneity, there remains the need to identify and define genuinely predictive biomarkers to guide treatment and patient selection. Cross-specialty and cross-sector collaboration, along with a broader collective data-sharing approach are key to optimizing immuno-oncology therapy in clinical practice. Continued support of younger research-clinicians is essential for future success in clinical, translational and basic science investigations.
Collapse
Affiliation(s)
- Michele Maio
- Center for Immuno-Oncology, Medical Oncology and Immunotherapy, Istituto Toscano Tumori, University Hospital of Siena, V.le Bracci, 16, 53100, Siena, Italy.
- Italian Network for Tumor Bio-Immunotherapy Foundation, Center for Immuno-Oncology, Istituto Toscano Tumori, University Hospital of Siena, 53100, Siena, Italy.
| | - George Coukos
- Lausanne Branch, Ludwig Institute for Cancer Research, University of Lausanne, Epalinges, Switzerland
- Department of Oncology, University Hospital of Lausanne (CHUV), Lausanne, Switzerland
| | - Soldano Ferrone
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
| | - Bernard A Fox
- Earle A. Chiles Research Institute at the Robert W. Franz Cancer Center, Providence Cancer Institute, Providence Portland Medical Center, 4805 NE Glisan, Portland, OR, 97213, USA
| | - Wolf H Fridman
- Cancer, Immune Control and Escape Team, Cordeliers Research Center, INSERM UMRS 1138, Paris, France
| | - Patrick L Garcia
- Merck, ZI de l'Ouriettaz, 1170, Aubonne, Switzerland
- An Affiliate of Merck KGaA, Darmstadt, Germany
| | | | - Olivier Provendier
- Laboratoires Pierre Fabre, 45 Place Abel Gance, 92100, Boulogne-Billancourt, France
| | - Vincenzo Russo
- Italian Network for Tumor Bio-Immunotherapy Foundation, Center for Immuno-Oncology, Istituto Toscano Tumori, University Hospital of Siena, 53100, Siena, Italy
- Unit of Immuno-Biotherapy of Melanoma and Solid Tumors, Division of Experimental Oncology, San Raffaele Scientific Institute, Via Olgettina 58, Milan, Italy
| | - Dominik Rüttinger
- Roche Pharma Research and Early Development, Roche Innovation Center Munich, Nonnenwald 2, 82377, Penzberg, Germany
| | - Aiman Shalabi
- Cancer Research Institute, 29 Broadway, New York, NY, 10006-3111, USA
| | - Zlatko Trajanoski
- Biocenter, Division of Bioinformatics, Medical University of Innsbruck, 6020, Innsbruck, Austria
| | | | - Jedd D Wolchok
- Ludwig Center for Cancer Immunotherapy, Parker Institute for Cancer Immunotherapy, San Francisco, USA
- Department of Medicine, Memorial Sloan-Kettering Cancer Center, Weill Cornell Medical and Graduate Schools, New York, NY, 10065, USA
| | - Ramy Ibrahim
- Parker Institute for Cancer Immunotherapy, 1 Letterman Drive, San Francisco, CA, 94129, USA
| |
Collapse
|
12
|
Davidson-Moncada J, Viboch E, Church SE, Warren SE, Rutella S. Dissecting the Immune Landscape of Acute Myeloid Leukemia. Biomedicines 2018; 6:E110. [PMID: 30477280 PMCID: PMC6316310 DOI: 10.3390/biomedicines6040110] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 11/19/2018] [Accepted: 11/21/2018] [Indexed: 01/05/2023] Open
Abstract
Acute myeloid leukemia (AML) is a molecularly heterogeneous hematological malignancy with variable response to treatment. Recurring cytogenetic abnormalities and molecular lesions identify AML patient subgroups with different survival probabilities; however, 50⁻70% of AML cases harbor either normal or risk-indeterminate karyotypes. The discovery of better biomarkers of clinical success and failure is therefore necessary to inform tailored therapeutic decisions. Harnessing the immune system against cancer with programmed death-1 (PD-1)-directed immune checkpoint blockade (ICB) and other immunotherapy agents is an effective therapeutic option for several advanced malignancies. However, durable responses have been observed in only a minority of patients, highlighting the need to gain insights into the molecular features that predict response and to also develop more effective and rational combination therapies that address mechanisms of immune evasion and resistance. We will review the state of knowledge of the immune landscape of AML and identify the broad opportunity to further explore this incompletely characterized space. Multiplexed, spatially-resolved immunohistochemistry, flow cytometry/mass cytometry, proteomic and transcriptomic approaches are advancing our understanding of the complexity of AML-immune interactions and are expected to support the design and expedite the delivery of personalized immunotherapy clinical trials.
Collapse
Affiliation(s)
| | - Elena Viboch
- NanoString Technologies Inc., Seattle, WA 98109, USA.
| | | | | | - Sergio Rutella
- John van Geest Cancer Research Center, School of Science and Technology, Nottingham Trent University, Nottingham NG11 8NS, UK.
| |
Collapse
|
13
|
Kondou R, Iizuka A, Nonomura C, Miyata H, Ashizawa T, Nagashima T, Ohshima K, Urakami K, Kusuhara M, Yamaguchi K, Akiyama Y. Classification of tumor microenvironment immune types based on immune response-associated gene expression. Int J Oncol 2018; 54:219-228. [PMID: 30387832 DOI: 10.3892/ijo.2018.4617] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Accepted: 09/27/2018] [Indexed: 11/06/2022] Open
Abstract
In 2014, the Shizuoka Cancer Center launched project High‑tech Omics‑based Patient Evaluation (HOPE), which features whole exome sequencing (WES) and gene expression profiling (GEP) of fresh surgical specimens from cancer patients. With the development of clinical trials of programmed death‑1 (PD‑1)/PD‑ligand 1 (PD‑L1) blockade, PD‑L1 expression and a high tumor mutation burden become possible biomarkers that could be used to predict immune responses. In this study, based on WES and GEP data from 1,734 tumors from the HOPE project, we established a tumor microenvironment (TME) immune‑type classification consisting of 4 types to evaluate the immunological status of cancer patients and analyze immunological pathways specific for immune types. Project HOPE was conducted in accordance with the Ethical Guidelines for Human Genome and Genetic Analysis Research with the approval of the Institutional Review Board. Based on the expression level of the PD‑L1 and CD8B genes, the immunological status was divided into 4 types as follows: A, PD‑L1+CD8B+; B, PD‑L1+CD8B‑; C, PD‑L1‑CD8B‑; and D, PD‑L1‑CD8B+. Type A, with PD‑L1+ and CD8B+, exhibited an upregulation of cytotoxic T lymphocyte (CTL) killing‑associated genes, T‑cell activation genes, antigen‑presentation and dendritic cell (DC) maturation genes, and T‑cell‑attracting chemokine genes, which promoted Th1 antitumor responses. By contrast, type C, with PD‑L1‑ and CD8B‑, exhibited a low expression of T‑cell‑activating genes and an upregulation of cancer driver gene signaling, which suggested an immune‑suppressive status. With regard to hypermutator tumors, PD‑L1+ hypermutator cases exhibited a specific upregulation of the IL6 gene compared with the PD‑L1‑ cases. On the whole, our data indicate that the classification of the TME immune types may prove to be a useful tool for evaluating the immunological status and predicting antitumor responses and prognosis.
Collapse
Affiliation(s)
- Ryota Kondou
- Immunotherapy Division, Shizuoka Cancer Center Research Institute, Shizuoka 411-8777, Japan
| | - Akira Iizuka
- Immunotherapy Division, Shizuoka Cancer Center Research Institute, Shizuoka 411-8777, Japan
| | - Chizu Nonomura
- Immunotherapy Division, Shizuoka Cancer Center Research Institute, Shizuoka 411-8777, Japan
| | - Haruo Miyata
- Immunotherapy Division, Shizuoka Cancer Center Research Institute, Shizuoka 411-8777, Japan
| | - Tadashi Ashizawa
- Immunotherapy Division, Shizuoka Cancer Center Research Institute, Shizuoka 411-8777, Japan
| | | | - Keiichi Ohshima
- Medical Genetics Division, Shizuoka Cancer Center Research Institute, Shizuoka Cancer Center Hospital, Shizuoka 411-8777, Japan
| | - Kenichi Urakami
- Cancer Diagnostic Research Division, Shizuoka Cancer Center Research Institute, Shizuoka Cancer Center Hospital, Shizuoka 411-8777, Japan
| | - Masatoshi Kusuhara
- Regional Resources Division, Shizuoka Cancer Center Research Institute, Shizuoka Cancer Center Hospital, Shizuoka 411-8777, Japan
| | - Ken Yamaguchi
- Office of the President, Shizuoka Cancer Center Hospital, Shizuoka 411-8777, Japan
| | - Yasuto Akiyama
- Immunotherapy Division, Shizuoka Cancer Center Research Institute, Shizuoka 411-8777, Japan
| |
Collapse
|
14
|
Bilgrau AE, Brøndum RF, Eriksen PS, Dybkær K, Bøgsted M. Estimating a common covariance matrix for network meta-analysis of gene expression datasets in diffuse large B-cell lymphoma. Ann Appl Stat 2018. [DOI: 10.1214/18-aoas1136] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
15
|
Choi J, Maeng HG, Lee SJ, Kim YJ, Kim DW, Lee HN, Namgung JH, Oh HM, Kim TJ, Jeong JE, Park SJ, Choi YM, Kang YW, Yoon SG, Lee JK. Diagnostic value of peripheral blood immune profiling in colorectal cancer. Ann Surg Treat Res 2018; 94:312-321. [PMID: 29854709 PMCID: PMC5976572 DOI: 10.4174/astr.2018.94.6.312] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Revised: 10/25/2017] [Accepted: 11/06/2017] [Indexed: 01/05/2023] Open
Abstract
PURPOSE Little is known about the clinical value of peripheral blood immune profiling. Here, we aimed to identify colorectal cancer (CRC)-related peripheral blood immune cells and develop liquid biopsy-based immune profiling models for CRC diagnosis. METHODS Peripheral blood from 131 preoperative patients with CRC and 174 healthy controls was analyzed by flow cytometry and automated hematology. CRC-related immune factors were identified by comparing the mean values of immune cell percentages and counts. Subsequently, CRC diagnostic algorithms were constructed using binary logistic regression. RESULTS Significant differences were observed in percentages and counts of white blood cells, lymphocytes, neutrophils, regulatory T cells, and myeloid-derived suppressor cells (MDSCs) of patients and controls. The neutrophil/lymphocyte and Th1/Th2 ratios were also significantly different. Likewise, the percentages and counts of peripheral blood programed death 1, cytotoxic T lymphocyte antigen 4, B-and T-lymphocyte attenuator, and lymphocyte activation gene-3 were higher in patients with CRC. The binary logistic regression model included 12 variables, age, CD3+%, NK%, CD4+CD279+%, CD4+CD25+%, CD4+CD152+%, CD3+CD366+%, CD3+CD272+%, CD3+CD223+%, CD158b-CD314+CD3-CD56+%, Th2%, and MDSCs cells/µL, for the prediction of cancer. Results of retrospective and prospective evaluation of the area under the curve, sensitivity, and specificity were 0.980 and 0.940, 91.53% and 85.80%, and 93.50% and 86.20%, respectively. CONCLUSION Peripheral blood immune profiling may be valuable in evaluating the immunity of CRC patients. Our liquid biopsy-based immune diagnostic method and its algorithms may serve as a novel tool for CRC diagnosis. Future largescale studies are needed for better characterization of its diagnostic value and potential for clinical application.
Collapse
Affiliation(s)
- Joungbum Choi
- Immunology Laboratory, Seoul Song Do Colorectal Hospital, Seoul, Korea
| | - Hyung Gun Maeng
- Immunology Laboratory, Seoul Song Do Colorectal Hospital, Seoul, Korea
| | - Su Jin Lee
- Immunology Laboratory, Seoul Song Do Colorectal Hospital, Seoul, Korea
| | - Young Joo Kim
- Immunology Laboratory, Seoul Song Do Colorectal Hospital, Seoul, Korea
| | - Da Woon Kim
- Immunology Laboratory, Seoul Song Do Colorectal Hospital, Seoul, Korea
| | - Ha Na Lee
- Immunology Laboratory, Seoul Song Do Colorectal Hospital, Seoul, Korea
| | - Ji Hyeon Namgung
- Immunology Laboratory, Seoul Song Do Colorectal Hospital, Seoul, Korea
| | - Hyun-Mee Oh
- Immunology Laboratory, Seoul Song Do Colorectal Hospital, Seoul, Korea
| | - Tae Joo Kim
- Immunology Laboratory, Seoul Song Do Colorectal Hospital, Seoul, Korea
| | - Ji Eun Jeong
- Cancer Immune Clinic, Seoul Song Do Colorectal Hospital, Seoul, Korea
| | - Sang Jean Park
- Cancer Immune Clinic, Seoul Song Do Colorectal Hospital, Seoul, Korea
| | - Yong Man Choi
- Cancer Immune Clinic, Seoul Song Do Colorectal Hospital, Seoul, Korea
| | - Yong Won Kang
- Department of Surgical Oncology, Seoul Song Do Colorectal Hospital, Seoul, Korea
| | - Seo Gue Yoon
- Department of Surgical Oncology, Seoul Song Do Colorectal Hospital, Seoul, Korea
| | - Jong Kyun Lee
- Immunology Laboratory, Seoul Song Do Colorectal Hospital, Seoul, Korea
- Cancer Immune Clinic, Seoul Song Do Colorectal Hospital, Seoul, Korea
| |
Collapse
|
16
|
Pockley AG, Lindsay JO, Foulds GA, Rutella S, Gribben JG, Alexander T, Snowden JA. Immune Reconstitution After Autologous Hematopoietic Stem Cell Transplantation in Crohn's Disease: Current Status and Future Directions. A Review on Behalf of the EBMT Autoimmune Diseases Working Party and the Autologous Stem Cell Transplantation In Refractory CD-Low Intensity Therapy Evaluation Study Investigators. Front Immunol 2018; 9:646. [PMID: 29670622 PMCID: PMC5893785 DOI: 10.3389/fimmu.2018.00646] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2017] [Accepted: 03/15/2018] [Indexed: 12/13/2022] Open
Abstract
Patients with treatment refractory Crohn's disease (CD) suffer debilitating symptoms, poor quality of life, and reduced work productivity. Surgery to resect inflamed and fibrotic intestine may mandate creation of a stoma and is often declined by patients. Such patients continue to be exposed to medical therapy that is ineffective, often expensive and still associated with a burden of adverse effects. Over the last two decades, autologous hematopoietic stem cell transplantation (auto-HSCT) has emerged as a promising treatment option for patients with severe autoimmune diseases (ADs). Mechanistic studies have provided proof of concept that auto-HSCT can restore immunological tolerance in chronic autoimmunity via the eradication of pathological immune responses and a profound reconfiguration of the immune system. Herein, we review current experience of auto-HSCT for the treatment of CD as well as approaches that have been used to monitor immune reconstitution following auto-HSCT in patients with ADs, including CD. We also detail immune reconstitution studies that have been integrated into the randomized controlled Autologous Stem cell Transplantation In refractory CD-Low Intensity Therapy Evaluation trial, which is designed to test the hypothesis that auto-HSCT using reduced intensity mobilization and conditioning regimens will be a safe and effective means of inducing sustained control in refractory CD compared to standard of care. Immunological profiling will generate insight into the pathogenesis of the disease, restoration of responsiveness to anti-TNF therapy in patients with recurrence of endoscopic disease and immunological events that precede the onset of disease in patients that relapse after auto-HSCT.
Collapse
Affiliation(s)
- Alan Graham Pockley
- John van Geest Cancer Research Centre, School of Science and Technology, Nottingham Trent University, Nottingham, United Kingdom
| | - James O Lindsay
- Centre for Immunobiology, Barts and the London School of Medicine and Dentistry, Blizard Institute, Queen Mary University of London, London, United Kingdom
| | - Gemma A Foulds
- John van Geest Cancer Research Centre, School of Science and Technology, Nottingham Trent University, Nottingham, United Kingdom
| | - Sergio Rutella
- John van Geest Cancer Research Centre, School of Science and Technology, Nottingham Trent University, Nottingham, United Kingdom
| | - John G Gribben
- Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Tobias Alexander
- Department of Rheumatology and Clinical Immunology, Charité - University Medicine, Berlin, Germany.,German Rheumatism Research Center Berlin (DRFZ) - a Leibniz Institute, Berlin, Germany
| | - John A Snowden
- Department of Haematology, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, United Kingdom
| |
Collapse
|
17
|
Zafeiris D, Vadakekolathu J, Wagner S, Pockley AG, Ball GR, Rutella S. Discovery and application of immune biomarkers for hematological malignancies. Expert Rev Mol Diagn 2017; 17:983-1000. [PMID: 28927305 DOI: 10.1080/14737159.2017.1381560] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Hematological malignancies originate and progress in primary and secondary lymphoid organs, where they establish a uniquely immune-suppressive tumour microenvironment. Although high-throughput transcriptomic and proteomic approaches are being employed to interrogate immune surveillance and escape mechanisms in patients with solid tumours, and to identify actionable targets for immunotherapy, our knowledge of the immunological landscape of hematological malignancies, as well as our understanding of the molecular circuits that underpin the establishment of immune tolerance, is not comprehensive. Areas covered: This article will discuss how multiplexed immunohistochemistry, flow cytometry/mass cytometry, proteomic and genomic techniques can be used to dynamically capture the complexity of tumour-immune interactions. Moreover, the analysis of multi-dimensional, clinically annotated data sets obtained from public repositories such as Array Express, TCGA and GEO is crucial to identify immune biomarkers, to inform the rational design of immune therapies and to predict clinical benefit in individual patients. We will also highlight how artificial neural network models and alternative methodologies integrating other algorithms can support the identification of key molecular drivers of immune dysfunction. Expert commentary: High-dimensional technologies have the potential to enhance our understanding of immune-cancer interactions and will support clinical decision making and the prediction of therapeutic benefit from immune-based interventions.
Collapse
Affiliation(s)
- Dimitrios Zafeiris
- a John van Geest Cancer Research Centre, College of Science and Technology , Nottingham Trent University , Nottingham , United Kingdom
| | - Jayakumar Vadakekolathu
- a John van Geest Cancer Research Centre, College of Science and Technology , Nottingham Trent University , Nottingham , United Kingdom
| | - Sarah Wagner
- a John van Geest Cancer Research Centre, College of Science and Technology , Nottingham Trent University , Nottingham , United Kingdom
| | - Alan Graham Pockley
- a John van Geest Cancer Research Centre, College of Science and Technology , Nottingham Trent University , Nottingham , United Kingdom
| | - Graham Roy Ball
- a John van Geest Cancer Research Centre, College of Science and Technology , Nottingham Trent University , Nottingham , United Kingdom
| | - Sergio Rutella
- a John van Geest Cancer Research Centre, College of Science and Technology , Nottingham Trent University , Nottingham , United Kingdom
| |
Collapse
|
18
|
Stern PL. Is immunity in cancer the key to improving clinical outcome?: Report on the International Symposium on Immunotherapy, The Royal Society, London, UK, 12-13 May 2017. THERAPEUTIC ADVANCES IN VACCINES 2017; 5:55-68. [PMID: 28794878 DOI: 10.1177/2051013617720659] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Accepted: 06/19/2017] [Indexed: 12/14/2022]
Affiliation(s)
- Peter L Stern
- Division of Molecular & Clinical Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Paterson Building, Wilmslow Road, Manchester M20 4BX, UK
| |
Collapse
|
19
|
Gulley JL, Berzofsky JA, Butler MO, Cesano A, Fox BA, Gnjatic S, Janetzki S, Kalavar S, Karanikas V, Khleif SN, Kirsch I, Lee PP, Maccalli C, Maecker H, Schlom J, Seliger B, Siebert J, Stroncek DF, Thurin M, Yuan J, Butterfield LH. Immunotherapy biomarkers 2016: overcoming the barriers. J Immunother Cancer 2017; 5:29. [PMID: 28653584 PMCID: PMC5359902 DOI: 10.1186/s40425-017-0225-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Accepted: 02/13/2017] [Indexed: 12/18/2022] Open
Abstract
This report summarizes the symposium, 'Immunotherapy Biomarkers 2016: Overcoming the Barriers', which was held on April 1, 2016 at the National Institutes of Health in Bethesda, Maryland. The symposium, cosponsored by the Society for Immunotherapy of Cancer (SITC) and the National Cancer Institute (NCI), focused on emerging immunotherapy biomarkers, new technologies, current hurdles to further progress, and recommendations for advancing the field of biomarker development.
Collapse
Affiliation(s)
- James L Gulley
- Genitourinary Malignancies Branch, Center for Cancer Research, NCI, 10 Center Dr., 13 N240, Bethesda, MD, 20892, USA
| | - Jay A Berzofsky
- Vaccine Branch, Center for Cancer Research, 41 Medlars Dr, Bldg 41 Rm D702D, Bethesda, MD, 20892, USA
| | - Marcus O Butler
- Princess Margaret Cancer Center/Ontario Cancer Institute, RM 9-622, 610 University Ave, Toronto, ON, Canada
| | - Alessandra Cesano
- NanoString, Inc., 500 Fairview Avenue North, Seattle, WA, 98109, USA
| | - Bernard A Fox
- Earle A. Chiles Research Institute, Providence Cancer Center, 4805 NE Glisan Street, Portland, OR, 97213, USA
| | - Sacha Gnjatic
- Department of Hematology/Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, S5-105, 1470 Madison Avenue, Box 1128, New York, NY, 10029, USA
| | - Sylvia Janetzki
- ZellNet Consulting, Inc., 555 North Avenue, Fort Lee, NJ, 07024, USA
| | - Shyam Kalavar
- Center for Devices and Radiological Health, U.S. Food and Drug Administration, 1401 Rockville Pike, Rockville, MD, 20852, USA
| | - Vaios Karanikas
- Roche Innovation Center Zurich, Wagistrasse 18, Schlieren, Switzerland
| | - Samir N Khleif
- Georgia Cancer Center, Augusta University, 1120 15th Street, CN-2101A, Augusta, GA, 30912, USA
| | - Ilan Kirsch
- Adaptive Biotechnologies, Inc., 1551 Eastlake Ave. E., Seattle, WA, 98102, USA
| | - Peter P Lee
- Department of Immuno-oncology, City of Hope, 1500 East Duarte Road, Duarte, CA, 91010, USA
| | - Cristina Maccalli
- Department of Translational Medicine, Sidra Medical and Research Center, Doha, Qatar
| | - Holden Maecker
- Stanford University Medical Center, 299 Campus Drive, Stanford, CA, 94303, USA
| | - Jeffrey Schlom
- National Cancer Institute, National Institutes of Health, 10 Center Drive, Bldg. 10, Room 8B09, Bethesda, MD, 20892, USA
| | - Barbara Seliger
- Institute of Medical Immunology, Martin Luther University Halle-Wittenberg, Magdeburger Str. 2, Halle, Germany
| | - Janet Siebert
- CytoAnalytics, 3500 South Albion Street, Cherry Hills Village, CO, 80113, USA
| | - David F Stroncek
- Department of Transfusion Medicine, National Institutes of Health, 10 Center Drive, Building 10, Room 3C720, Bethesda, MD, 20892, USA
| | - Magdalena Thurin
- National Cancer Institute, Cancer Diagnosis Program, DCTD, National Institutes of Health, 9609 Medical Center Drive, Bethesda, 20892, MD, USA
| | - Jianda Yuan
- Early Clinical Oncology Development, Merck Research Laboratories, Rahway, NJ, 07065, USA
| | - Lisa H Butterfield
- Department of Medicine, Surgery and Immunology, University of Pittsburgh Cancer Institute, 5117 Centre Avenue, Pittsburgh, PA, 15213, USA.
| |
Collapse
|