1
|
Schett G, Müller F, Taubmann J, Mackensen A, Wang W, Furie RA, Gold R, Haghikia A, Merkel PA, Caricchio R, D'Agostino MA, Locatelli F, June CH, Mougiakakos D. Advancements and challenges in CAR T cell therapy in autoimmune diseases. Nat Rev Rheumatol 2024; 20:531-544. [PMID: 39107407 DOI: 10.1038/s41584-024-01139-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/28/2024] [Indexed: 08/29/2024]
Abstract
Chimeric antigen receptor (CAR) T cells are highly effective at targeting and eliminating cells of the B cell lineage. CAR T cell therapy has become a standard-of-care treatment for patients with relapsed or refractory B cell malignancies. In addition, the administration of genetically modified T cells with the capacity to deplete B cells and/or plasma cells has tremendous therapeutic potential in autoimmune diseases. In the past few years, CD19-based and B cell maturation antigen (BCMA)-based CAR T cell therapies have been applied to various B cell-mediated autoimmune diseases including systemic lupus erythematosus, idiopathic inflammatory myopathy, systemic sclerosis, neuromyelitis optica spectrum disorder, myasthenia gravis and multiple sclerosis. The scientific rationale behind this approach is that deep depletion of B cells, including autoreactive B cell clones, could restore normal immune function, referred to as an immune reset. In this Review, we discuss important aspects of CAR T cell therapy in autoimmune disease, including considerations relating to patient selection, safety, efficacy and medical management. These considerations are based on the early experiences of CAR T cell therapy in autoimmune diseases, and as the field of CAR T cell therapy in autoimmune diseases continues to rapidly evolve, these issues will remain subject to ongoing refinement and adaptation.
Collapse
Affiliation(s)
- Georg Schett
- Department of Internal Medicine 3 - Rheumatology and Immunology, FAU Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany.
- Deutsches Zentrum Immuntherapie, Universitätsklinikum Erlangen, Friedrich-Alexander University (FAU) Erlangen-Nürnberg, Erlangen, Germany.
| | - Fabian Müller
- Deutsches Zentrum Immuntherapie, Universitätsklinikum Erlangen, Friedrich-Alexander University (FAU) Erlangen-Nürnberg, Erlangen, Germany
- Department of Internal Medicine 5 - Hematology and Oncology, FAU Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Jule Taubmann
- Department of Internal Medicine 3 - Rheumatology and Immunology, FAU Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie, Universitätsklinikum Erlangen, Friedrich-Alexander University (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Andreas Mackensen
- Deutsches Zentrum Immuntherapie, Universitätsklinikum Erlangen, Friedrich-Alexander University (FAU) Erlangen-Nürnberg, Erlangen, Germany
- Department of Internal Medicine 5 - Hematology and Oncology, FAU Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Wei Wang
- Department of Neurology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Rich A Furie
- Department of Rheumatology, Northwell Health, Great Neck, New York, NJ, USA
| | - Ralf Gold
- Department of Neurology, St. Josef-Hospital Bochum, Ruhr-University Bochum, Bochum, Germany
| | - Aiden Haghikia
- Department of Neurology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Peter A Merkel
- Division of Rheumatology, Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Division of Epidemiology, Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania, Philadelphia, PA, USA
| | | | | | - Franco Locatelli
- Department of Paediatric Hematology and Oncology, Bambino Gesù Children's Hospital, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) and Catholic University of the Sacred Heart, Rome, Italy
| | - Carl H June
- Center for Cellular Immunology, Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Dimitrios Mougiakakos
- Department of Hematology, Oncology, and Cell Therapy, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
- Health Campus Immunology, Infectiology and Inflammation, Medical Center, Otto-von-Guericke University, Magdeburg, Germany
| |
Collapse
|
2
|
Rogers S, Charles A, Thomas RM. The Prospect of Harnessing the Microbiome to Improve Immunotherapeutic Response in Pancreatic Cancer. Cancers (Basel) 2023; 15:5708. [PMID: 38136254 PMCID: PMC10741649 DOI: 10.3390/cancers15245708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 11/24/2023] [Accepted: 12/02/2023] [Indexed: 12/24/2023] Open
Abstract
Pancreatic ductal adenocarcinoma cancer (PDAC) is projected to become the second leading cause of cancer-related death in the United States by 2030. Patients are often diagnosed with advanced disease, which explains the dismal 5-year median overall survival rate of ~12%. Immunotherapy has been successful in improving outcomes in the past decade for a variety of malignancies, including gastrointestinal cancers. However, PDAC is historically an immunologically "cold" tumor, one with an immunosuppressive environment and with restricted entry of immune cells that have limited the success of immunotherapy in these tumors. The microbiome, the intricate community of microorganisms present on and within humans, has been shown to contribute to many cancers, including PDAC. Recently, its role in tumor immunology and response to immunotherapy has generated much interest. Herein, the current state of the interaction of the microbiome and immunotherapy in PDAC is discussed with a focus on needed areas of study in order to harness the immune system to combat pancreatic cancer.
Collapse
Affiliation(s)
- Sherise Rogers
- Department of Medicine, Division of Hematology and Oncology, University of Florida College of Medicine, Gainesville, FL 32610, USA;
| | - Angel Charles
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL 32610, USA;
| | - Ryan M. Thomas
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL 32610, USA;
- Department of Molecular Genetics and Microbiology, University of Florida College of Medicine, Gainesville, FL 32603, USA
| |
Collapse
|
3
|
Bhattacharjee P, Karim KA, Khan Z. Harnessing the Microbiome: A Comprehensive Review on Advancing Therapeutic Strategies for Rheumatic Diseases. Cureus 2023; 15:e50964. [PMID: 38249228 PMCID: PMC10800157 DOI: 10.7759/cureus.50964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/22/2023] [Indexed: 01/23/2024] Open
Abstract
Rheumatic diseases are a group of disorders that affect the joints, muscles, and bones. These diseases, such as rheumatoid arthritis, lupus, and psoriatic arthritis, can cause pain, stiffness, and swelling, leading to reduced mobility and disability. Recent studies have identified the microbiome, the diverse community of microorganisms that live in and on the human body, as a potential factor in the development and progression of rheumatic diseases. Harnessing the microbiome offers a promising new avenue for developing therapeutic strategies for these debilitating conditions. There is growing interest in the role of oral and gut microbiomes in the management of rheumatoid arthritis and other autoimmune disease. Microbial metabolites have immunomodulatory properties that could be exploited for rheumatic disorders. A wide range of microorganisms are present in the oral cavity and are found to be vulnerable to the effects of the environment. The physiology and ecology of the microbiota become intimately connected with those of the host, and they critically influence the promotion of health or progression toward disease. This article aims to provide a comprehensive overview of the current state of knowledge on oral and gut microbiome and its potential future role in the management of rheumatic diseases. This article will also discuss newer treatment strategies such as bioinformatic analyses and fecal transplantation.
Collapse
Affiliation(s)
- Priyadarshini Bhattacharjee
- Acute Medicine, Cambridge University Hospital NHS Foundation Trust, Cambridge, GBR
- School of Clinical Medicine, University of Cambridge, Cambridge, GBR
| | - Karim Arif Karim
- Medicine and Surgery, Kamuzu University of Health Sciences, Blantyre, MWI
| | - Zahid Khan
- Acute Medicine, Mid and South Essex NHS Foundation Trust, Southend-on-Sea, GBR
- Cardiology, Bart's Heart Centre, London, GBR
- Cardiology and General Medicine, Barking, Havering and Redbridge University Hospitals NHS Trust, London, GBR
- Cardiology, Royal Free Hospital, London, GBR
| |
Collapse
|
4
|
Xu X, Zhang M, Liu X, Chai M, Diao L, Ma L, Nie S, Xu M, Wang Y, Mo F, Liu M. Probiotics formulation and cancer nanovaccines show synergistic effect in immunotherapy and prevention of colon cancer. iScience 2023; 26:107167. [PMID: 37456845 PMCID: PMC10338235 DOI: 10.1016/j.isci.2023.107167] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 04/26/2023] [Accepted: 06/13/2023] [Indexed: 07/18/2023] Open
Abstract
Probiotics play essential roles in immune modulation. Combining probiotics with cancer vaccines potentially can achieve a synergistic effect. To maximize the efficacy of probiotics, proper probiotics formulation is necessary. Herein, Lactobacillus rhamnosus and Bifidobacterium longum are coated with lipid membrane to achieve the goal of losing less activity and bettering colonization in colon. In the subcutaneous transplanted colon cancer mouse model, probiotics formulation showed potent preventive and therapeutic efficacy, and the efficacy could be further improved by combining with cancer nanovaccines. Probiotics formulation can perform as immune adjuvants to enhance the innate immune response or as in-situ cancer vaccines. In the study of preventing chemical-induced orthotopic colon cancer model, probiotics formulation alone efficiently reduced tumor number in colon and the efficacy is improved by combining with cancer nanovaccines. All in all, the studies demonstrated that probiotics formulation can assist to maximize the efficacy of cancer nanovaccines.
Collapse
Affiliation(s)
- Xiangxiang Xu
- College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, People’s Republic of China
- Kunshan Hospital of Traditional Chinese Medicine, Kunshan, Jiangsu 215300, People’s Republic of China
- Suzhou Ersheng Biopharmaceutical Co., Ltd, Suzhou 215123, People’s Republic of China
| | - Meng Zhang
- College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, People’s Republic of China
| | - Xiaoyan Liu
- College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, People’s Republic of China
| | - Mingze Chai
- College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, People’s Republic of China
| | - Lu Diao
- College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, People’s Republic of China
- Kunshan Hospital of Traditional Chinese Medicine, Kunshan, Jiangsu 215300, People’s Republic of China
- Suzhou Ersheng Biopharmaceutical Co., Ltd, Suzhou 215123, People’s Republic of China
| | - Lin Ma
- Kunshan Hospital of Traditional Chinese Medicine, Kunshan, Jiangsu 215300, People’s Republic of China
| | - Shuang Nie
- Department of Nutrition and Food Hygiene, Faculty of Naval Medicine, Naval Medical University, 800 Xiangyin Rd, Shanghai 200433, People’s Republic of China
| | - Minghao Xu
- College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, People’s Republic of China
- Suzhou Ersheng Biopharmaceutical Co., Ltd, Suzhou 215123, People’s Republic of China
| | - Yipeng Wang
- College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, People’s Republic of China
| | - Fengfeng Mo
- Department of Nutrition and Food Hygiene, Faculty of Naval Medicine, Naval Medical University, 800 Xiangyin Rd, Shanghai 200433, People’s Republic of China
| | - Mi Liu
- College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, People’s Republic of China
- Kunshan Hospital of Traditional Chinese Medicine, Kunshan, Jiangsu 215300, People’s Republic of China
- Suzhou Ersheng Biopharmaceutical Co., Ltd, Suzhou 215123, People’s Republic of China
| |
Collapse
|
5
|
Grigorievskaya ZV, Petukhova IN, Bagirova NS, Aginova VV, Kononets PV. Role of the microbiota in oncogenesis. SIBERIAN JOURNAL OF ONCOLOGY 2023; 22:129-142. [DOI: 10.21294/1814-4861-2023-22-2-129-142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
Objective. To conduct a systematic analysis of data on the results of studies published in scientific journals on the pro-carcinogenic and anticarcinogenic role of microbiota, as well as on the therapeutic potential of microorganisms in oncogenesis.Material and Methods. The articles were searched using the Web of Science, Scopus, PubMed, Medline, and eLIBRARY databases. More than 150 sources dedicated to the study of the carcinogenic function of the microbiota and the possible influence of its species and quantitative composition on the efficacy and toxicity of antitumor therapy were found. Data from 71 articles were included in the review.Results. The relationship between the gut microbiota and cancer is multifactorial and bilateral: pro-carcinogenic on the one hand and anti-carcinogenic on the other hand. Microorganisms can induce tumor growth and cancer development through DNA damage and induction of mutagenesis, trigger oncogenic signals, disruption of barrier function, as well as immune response system disruption. Depletion of microbiota, the development of dysbiosis and induction of chronic inflammatory state are negative factors in the development of cancer. The anticancer effect of microorganisms is presumably based on the production of tumor-suppressive metabolites that function through multiple immune reactions. Maintenance of barrier function, competitive exclusion of pathogenic bacteria, and direct action on immune cells to prevent inflammation are also important protective factors. The presence of intratumor microorganisms in various tumors has been noted. Changes in species and quantitative composition of cancer patients’ microbiota are influenced by diet, taking antibacterial drugs, chemo-, immuno- and radiation therapy. In turn, the microbiota can affect the ongoing treatment. Numerous studies on the influence of the gut microbiota on the efficacy of immunotherapy, particularly in disseminated melanoma, have been conducted. It has been suggested that primary resistance to immunotherapy may be related to the abnormal composition of the gut microbiota. The level of gut microfora composition diversity and the number of Faecalibacterium or Bacteroidales in the fecal microbiota have been suggested to be the predictor of response to anti-PD-1 therapy. To change the composition and activity of the gut microbiota, several therapeutic methods, such as the administration of prebiotics, probiotics, synbiotics, postbiotics, fecal microbiota transplantation, as well as the change in the microbiota composition through a specific diet, are available.
Collapse
Affiliation(s)
- Z. V. Grigorievskaya
- N.N. Blokhin National Medical Research Center of Oncology of the Ministry of Health of Russia
| | - I. N. Petukhova
- N.N. Blokhin National Medical Research Center of Oncology of the Ministry of Health of Russia
| | - N. S. Bagirova
- N.N. Blokhin National Medical Research Center of Oncology of the Ministry of Health of Russia
| | - V. V. Aginova
- N.N. Blokhin National Medical Research Center of Oncology of the Ministry of Health of Russia
| | - P. V. Kononets
- N.N. Blokhin National Medical Research Center of Oncology of the Ministry of Health of Russia
| |
Collapse
|
6
|
Thippabhotla S, Liu B, Podgorny A, Yooseph S, Yang Y, Zhang J, Zhong C. Integrated de novo gene prediction and peptide assembly of metagenomic sequencing data. NAR Genom Bioinform 2023; 5:lqad023. [PMID: 36915411 PMCID: PMC10006731 DOI: 10.1093/nargab/lqad023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 12/03/2022] [Accepted: 02/18/2023] [Indexed: 03/16/2023] Open
Abstract
Metagenomics is the study of all genomic content contained in given microbial communities. Metagenomic functional analysis aims to quantify protein families and reconstruct metabolic pathways from the metagenome. It plays a central role in understanding the interaction between the microbial community and its host or environment. De novo functional analysis, which allows the discovery of novel protein families, remains challenging for high-complexity communities. There are currently three main approaches for recovering novel genes or proteins: de novo nucleotide assembly, gene calling and peptide assembly. Unfortunately, their information dependency has been overlooked, and each has been formulated as an independent problem. In this work, we develop a sophisticated workflow called integrated Metagenomic Protein Predictor (iMPP), which leverages the information dependencies for better de novo functional analysis. iMPP contains three novel modules: a hybrid assembly graph generation module, a graph-based gene calling module, and a peptide assembly-based refinement module. iMPP significantly improved the existing gene calling sensitivity on unassembled metagenomic reads, achieving a 92-97% recall rate at a high precision level (>85%). iMPP further allowed for more sensitive and accurate peptide assembly, recovering more reference proteins and delivering more hypothetical protein sequences. The high performance of iMPP can provide a more comprehensive and unbiased view of the microbial communities under investigation. iMPP is freely available from https://github.com/Sirisha-t/iMPP.
Collapse
Affiliation(s)
- Sirisha Thippabhotla
- Department of Electrical Engineering and Computer Science, The University of Kansas, Lawrence, KS 66045, USA
| | - Ben Liu
- Department of Electrical Engineering and Computer Science, The University of Kansas, Lawrence, KS 66045, USA
| | - Adam Podgorny
- Center for Computational Biology, The University of Kansas, Lawrence, KS 66045, USA
| | - Shibu Yooseph
- Department of Computer Science, Genomics and Bioinformatics Cluster, University of Central Florida, Orlando, FL 32816, USA
| | - Youngik Yang
- National Marine Biodiversity Institute of Korea, 101-75, Jangsan-ro, Janghang-eup, Seochun-gun, Chungchungnam-do, 33662, South Korea
| | - Jun Zhang
- Division of Medical Oncology, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA.,Department of Cancer Biology, University of Kansas Cancer Center; Kansas City, KS 66160, USA
| | - Cuncong Zhong
- Department of Electrical Engineering and Computer Science, The University of Kansas, Lawrence, KS 66045, USA
| |
Collapse
|
7
|
Bhat MH, Hajam YA, Neelam, Kumar R, Diksha. Microbial Diversity and Their Role in Human Health and Diseases. ROLE OF MICROBES IN SUSTAINABLE DEVELOPMENT 2023:1-33. [DOI: 10.1007/978-981-99-3126-2_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
8
|
Small Molecules as Toll-like Receptor 4 Modulators Drug and In-House Computational Repurposing. Biomedicines 2022; 10:biomedicines10092326. [PMID: 36140427 PMCID: PMC9496124 DOI: 10.3390/biomedicines10092326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 09/08/2022] [Accepted: 09/09/2022] [Indexed: 12/05/2022] Open
Abstract
The innate immunity toll-like receptor 4 (TLR4) system is a receptor of paramount importance as a therapeutic target. Virtual screening following a “computer-aided drug repurposing” approach was applied to the discovery of novel TLR4 modulators with a non-lipopolysaccharide-like structure. We screened almost 29,000 approved drugs and drug-like molecules from commercial, public, and in-house academia chemical libraries and, after biological assays, identified several compounds with TLR4 antagonist activity. Our computational protocol showed to be a robust approach for the identification of hits with drug-like scaffolds as possible inhibitors of the TLR4 innate immune pathways. Our collaborative work broadens the chemical diversity for inspiration of new classes of TLR4 modulators.
Collapse
|
9
|
Burcher KM, Burcher JT, Inscore L, Bloomer CH, Furdui CM, Porosnicu M. A Review of the Role of Oral Microbiome in the Development, Detection, and Management of Head and Neck Squamous Cell Cancers. Cancers (Basel) 2022; 14:4116. [PMID: 36077651 PMCID: PMC9454796 DOI: 10.3390/cancers14174116] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 08/19/2022] [Accepted: 08/22/2022] [Indexed: 11/16/2022] Open
Abstract
The role of the microbiome in the development and propagation of head and neck squamous cell cancer (HNSCC) is largely unknown and the surrounding knowledge lags behind what has been discovered related to the microbiome and other malignancies. In this review, the authors performed a structured analysis of the available literature from several databases. The authors discuss the merits and detriments of several studies discussing the microbiome of the structures of the aerodigestive system throughout the development of HNSCC, the role of the microbiome in the development of malignancies (generally and in HNSCC) and clinical applications of the microbiome in HNSCC. Further studies will be needed to adequately describe the relationship between HNSCC and the microbiome, and to push this relationship into a space where it is clinically relevant outside of a research environment.
Collapse
Affiliation(s)
| | | | - Logan Inscore
- Wake Forest Baptist Medical Center, Winston-Salem, NC 27157, USA
| | | | | | | |
Collapse
|
10
|
Golčić M, Simetić L, Majnarić T, Golčić G, Herceg D. Could fecal microbial transplantation offer a new potential in the treatment of metastatic pancreatic ductal adenocarcinoma? Med Hypotheses 2022. [DOI: 10.1016/j.mehy.2022.110801] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
|
11
|
Viswanath DI, Liu HC, Capuani S, Vander Pol RS, Saunders SZ, Chua CYX, Grattoni A. Engineered implantable vaccine platform for continuous antigen-specific immunomodulation. Biomaterials 2022; 281:121374. [PMID: 35066287 PMCID: PMC8865051 DOI: 10.1016/j.biomaterials.2022.121374] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 01/03/2022] [Accepted: 01/12/2022] [Indexed: 02/03/2023]
Abstract
Cancer vaccines harness the host immune system to generate antigen-specific antitumor immunity for long-term tumor elimination with durable immunomodulation. Commonly investigated strategies reintroduce ex vivo autologous dendritic cells (DCs) but have limited clinical adoption due to difficulty in manufacturing, delivery and low clinical efficacy. To combat this, we designed the "NanoLymph", an implantable subcutaneous device for antigen-specific antitumor immunomodulation. The NanoLymph consists of a dual-reservoir platform for sustained release of immune stimulants via a nanoporous membrane and hydrogel-encapsulated antigens for local immune cell recruitment and activation, respectively. Here, we present the development and characterization of the NanoLymph as well as efficacy validation for immunomodulation in an immunocompetent murine model. Specifically, we established the NanoLymph biocompatibility and mechanical stability. Further, we demonstrated minimally invasive transcutaneous refilling of the drug reservoir in vivo for prolonging drug release duration. Importantly, our study demonstrated that local elution of two drugs (GMCSF and Resiquimod) generates an immune stimulatory microenvironment capable of local DC recruitment and activation and generation of antigen-specific T lymphocytes within 14 days. In summary, the NanoLymph approach can achieve in situ immunomodulation, presenting a viable strategy for therapeutic cancer vaccines.
Collapse
Affiliation(s)
- Dixita Ishani Viswanath
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, USA; Texas A&M University College of Medicine, Bryan & Houston, TX, USA
| | - Hsuan-Chen Liu
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, USA
| | - Simone Capuani
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, USA; University of Chinese Academy of Science (UCAS), Shijingshan, 19 Yuquan Road, Beijing, 100049, China
| | | | | | | | - Alessandro Grattoni
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, USA; Department of Surgery, Houston Methodist Hospital, Houston, TX, USA; Department of Radiation Oncology, Houston Methodist Hospital, Houston, TX, USA.
| |
Collapse
|
12
|
Li M, Wang X, Wang Y, Bao S, Chang Q, Liu L, Zhang S, Sun L. Strategies for Remodeling the Tumor Microenvironment Using Active Ingredients of Ginseng-A Promising Approach for Cancer Therapy. Front Pharmacol 2022; 12:797634. [PMID: 35002732 PMCID: PMC8727883 DOI: 10.3389/fphar.2021.797634] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 12/06/2021] [Indexed: 12/21/2022] Open
Abstract
The tumor microenvironment (TME) plays a key role in promoting the initiation and progression of tumors, leading to chemoradiotherapy resistance and immunotherapy failure. Targeting of the TME is a novel anti-tumor therapeutic approach and is currently a focus of anti-tumor research. Panax ginseng C. A. Meyer (ginseng), an ingredient of well-known traditional Asia medicines, exerts beneficial anti-tumor effects and can regulate the TME. Here, we present a systematic review that describes the current status of research efforts to elucidate the functions and mechanisms of ginseng active components (including ginsenosides and ginseng polysaccharides) for achieving TME regulation. Ginsenosides have variety effects on TME, such as Rg3, Rd and Rk3 can inhibit tumor angiogenesis; Rg3, Rh2 and M4 can regulate the function of immune cells; Rg3, Rd and Rg5 can restrain the stemness of cancer stem cells. Ginseng polysaccharides (such as red ginseng acidic polysaccharides and polysaccharides extracted from ginseng berry and ginseng leaves) can regulate TME mainly by stimulating immune cells. In addition, we propose a potential mechanistic link between ginseng-associated restoration of gut microbiota and the tumor immune microenvironment. Finally, we describe recent advances for improving ginseng efficacy, including the development of a nano-drug delivery system. Taken together, this review provides novel perspectives on potential applications for ginseng active ingredients as anti-cancer adjuvants that achieve anti-cancer effects by reshaping the tumor microenvironment.
Collapse
Affiliation(s)
- Mo Li
- Department of Radiotherapy, The Second Hospital of Jilin University, Changchun, China.,Department of Thyroid Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Xin Wang
- Department of Neurology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Ying Wang
- Department of Breast Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Shunchao Bao
- Department of Radiotherapy, The Second Hospital of Jilin University, Changchun, China
| | - Qing Chang
- Department of Radiotherapy, The Second Hospital of Jilin University, Changchun, China
| | - Linlin Liu
- Department of Radiotherapy, The Second Hospital of Jilin University, Changchun, China
| | - Shuai Zhang
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, China
| | - Liwei Sun
- Research Center of Traditional Chinese Medicine, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, China
| |
Collapse
|
13
|
Shi L, Sheng J, Chen G, Zhu P, Shi C, Li B, Park C, Wang J, Zhang B, Liu Z, Yang X. Combining IL-2-based immunotherapy with commensal probiotics produces enhanced antitumor immune response and tumor clearance. J Immunother Cancer 2021; 8:jitc-2020-000973. [PMID: 33028692 PMCID: PMC7542661 DOI: 10.1136/jitc-2020-000973] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/20/2020] [Indexed: 12/11/2022] Open
Abstract
Background Interleukin-2 (IL-2) serves as a pioneer of immunotherapeutic agent in cancer treatment. However, there is a considerable proportion of patients who cannot benefit from this therapy due to the limited clinical responses and dose-limiting toxicities. Mounting evidence indicates that commensal microbiota shapes the outcome of cancer immunotherapies. In this study, we aim to investigate the enhancing effect of Akkermansia muciniphila (AKK), a beneficial commensal microbe receiving considerable attentions, on the antitumor efficacy of IL-2 and explore the underlying molecular mechanism. Methods Colorectal carcinoma patient-derived tumor tissues were used to evaluate the therapeutic efficacy of combination treatment. AKK was orally delivered to B16F10 and CT26 tumor-bearing mice along with systemic IL-2 treatment. Flow cytometry was carried out to analyze the tumor immune microenvironment. The molecular mechanism of the enhanced therapeutic efficacy was explored by RNA-seq and then verified in tumor-bearing mice. Results Combined treatment with IL-2 and AKK showed a stronger antitumor efficacy in colorectal cancer patient-derived tumor tissues. Meanwhile, the therapeutic outcome of IL-2 was significantly potentiated by oral administration of AKK in subcutaneous melanoma and colorectal tumor-bearing mice, resulting from the strengthened antitumor immune surveillance. Mechanistically, the antitumor immune response elicited by AKK was partially mediated by Amuc, derived from the outer membrane protein of AKK, through activating toll-like receptor 2 (TLR2) signaling pathway. Besides, oral supplementation with AKK protected gut barrier function and maintained mucosal homeostasis under systemic IL-2 treatment. Conclusion These findings propose that IL-2 combined with AKK is a novel therapeutic strategy with prospecting application for cancer treatment in clinical practice.
Collapse
Affiliation(s)
- Linlin Shi
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Jianyong Sheng
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Guozhong Chen
- Department of Biotechnology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Peng Zhu
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Changping Shi
- Department of Biotechnology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Bei Li
- Department of Biotechnology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Chaiwoo Park
- Department of Biotechnology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Jingyi Wang
- Department of Biology, St. Olaf College, Northfield, Minnesota, USA
| | - Bixiang Zhang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhi Liu
- Department of Biotechnology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Xiangliang Yang
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
14
|
The Role of DNA Damage Response in Dysbiosis-Induced Colorectal Cancer. Cells 2021; 10:cells10081934. [PMID: 34440703 PMCID: PMC8391204 DOI: 10.3390/cells10081934] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 07/23/2021] [Accepted: 07/23/2021] [Indexed: 12/16/2022] Open
Abstract
The high incidence of colorectal cancer (CRC) in developed countries indicates a predominant role of the environment as a causative factor. Natural gut microbiota provides multiple benefits to humans. Dysbiosis is characterized by an unbalanced microbiota and causes intestinal damage and inflammation. The latter is a common denominator in many cancers including CRC. Indeed, in an inflammation scenario, cellular growth is promoted and immune cells release Reactive Oxygen Species (ROS) and Reactive Nitrogen Species (RNS), which cause DNA damage. Apart from that, many metabolites from the diet are converted into DNA damaging agents by microbiota and some bacteria deliver DNA damaging toxins in dysbiosis conditions as well. The interactions between diet, microbiota, inflammation, and CRC are not the result of a straightforward relationship, but rather a network of multifactorial interactions that deserve deep consideration, as their consequences are not yet fully elucidated. In this paper, we will review the influence of dysbiosis in the induction of DNA damage and CRC.
Collapse
|
15
|
Szczyrek M, Bitkowska P, Chunowski P, Czuchryta P, Krawczyk P, Milanowski J. Diet, Microbiome, and Cancer Immunotherapy-A Comprehensive Review. Nutrients 2021; 13:2217. [PMID: 34203292 PMCID: PMC8308287 DOI: 10.3390/nu13072217] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 06/11/2021] [Accepted: 06/25/2021] [Indexed: 02/08/2023] Open
Abstract
The immune system plays a key role in cancer suppression. Immunotherapy is widely used as a treatment method in patients with various types of cancer. Immune checkpoint blockade using antibodies, such as anti-PD-1, anti-PD-L1, and anti-CTLA-4, is currently gaining popularity. A systematic literature search was executed, and all available data was summarized. This review shows that specific dietary patterns (such as, e.g., animal-based, vegetarian, or Mediterranean diet) alter the gut microbiome's composition. An appropriate intestinal microbiota structure might modulate the function of human immune system, which affects the bodily anti-cancer response. This paper shows also that specific bacteria species inhabiting the gastrointestinal tract can have a beneficial influence on the efficacy of immunotherapy. Antibiotics weaken gut bacteria and worsen the immune checkpoint blockers' efficacy, whereas a faecal microbiota transplant or probiotics supplementation may help restore bacterial balance in the intestine. Other factors (like vitamins, glucose, or BMI) change the cancer treatment response, as well. This review demonstrates that there is a strong association between one's diet, gut microbiome composition, and the outcome of immunotherapy. However, further investigation on this subject is required.
Collapse
Affiliation(s)
- Michał Szczyrek
- Department of Pneumology, Oncology and Allergology, Medical University of Lublin, 20-090 Lublin, Poland; (P.B.); (P.C.); (P.C.); (P.K.); (J.M.)
| | | | | | | | | | | |
Collapse
|
16
|
Li W, Deng X, Chen T. Exploring the Modulatory Effects of Gut Microbiota in Anti-Cancer Therapy. Front Oncol 2021; 11:644454. [PMID: 33928033 PMCID: PMC8076595 DOI: 10.3389/fonc.2021.644454] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 03/18/2021] [Indexed: 12/16/2022] Open
Abstract
In the recent decade, gut microbiota has received growing interest due to its role in human health and disease. On the one hand, by utilizing the signaling pathways of the host and interacting with the immune system, the gut microbiota is able to maintain the homeostasis in human body. This important role is mainly modulated by the composition of microbiota, as a normal microbiota composition is responsible for maintaining the homeostasis of human body, while an altered microbiota profile could contribute to several pathogenic conditions and may further lead to oncogenesis and tumor progression. Moreover, recent insights have especially focused on the important role of gut microbiota in current anticancer therapies, including chemotherapy, radiotherapy, immunotherapy and surgery. Research findings have indicated a bidirectional interplay between gut microbiota and these therapeutic methods, in which the implementation of different therapeutic methods could lead to different alterations in gut microbiota, and the presence of gut microbiota could in turn contribute to different therapeutic responses. As a result, manipulating the gut microbiota to reduce the therapy-induced toxicity may provide an adjuvant therapy to achieve a better therapeutic outcome. Given the complex role of gut microbiota in cancer treatment, this review summarizes the interactions between gut microbiota and anticancer therapies, and demonstrates the current strategies for reshaping gut microbiota community, aiming to provide possibilities for finding an alternative approach to lower the damage and improve the efficacy of cancer therapy.
Collapse
Affiliation(s)
- Wenyu Li
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Queen Mary School, Nanchang University, Nanchang, China
| | - Xiaorong Deng
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Tingtao Chen
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, The First Affiliated Hospital, Nanchang University, Nanchang, China
| |
Collapse
|
17
|
Oliva M, Schneeberger PHH, Rey V, Cho M, Taylor R, Hansen AR, Taylor K, Hosni A, Bayley A, Hope AJ, Bratman SV, Ringash J, Singh S, Weinreb I, Perez-Ordoñez B, Chepeha D, Waldron J, Xu W, Guttman D, Siu LL, Coburn B, Spreafico A. Transitions in oral and gut microbiome of HPV+ oropharyngeal squamous cell carcinoma following definitive chemoradiotherapy (ROMA LA-OPSCC study). Br J Cancer 2021; 124:1543-1551. [PMID: 33750907 PMCID: PMC8076306 DOI: 10.1038/s41416-020-01253-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 11/25/2020] [Accepted: 12/17/2020] [Indexed: 02/07/2023] Open
Abstract
Background Oral and gut microbiomes have emerged as potential biomarkers in cancer. We characterised the oral and gut microbiomes in a prospective observational cohort of HPV+ oropharyngeal squamous cell carcinoma (OPSCC) patients and evaluated the impact of chemoradiotherapy (CRT). Methods Saliva, oropharyngeal swabs over the tumour site and stool were collected at baseline and post-CRT. 16S RNA and shotgun metagenomic sequencing were used to generate taxonomic profiles, including relative abundance (RA), bacterial density, α-diversity and β-diversity. Results A total of 132 samples from 22 patients were analysed. Baseline saliva and swabs had similar taxonomic composition (R2 = 0.006; p = 0.827). Oropharyngeal swabs and stool taxonomic composition varied significantly by stage, with increased oral RA of Fusobacterium nucleatum observed in stage III disease (p < 0.05). CRT significantly reduced the species richness and increased the RA of gut-associated taxa in oropharyngeal swabs (p < 0.05), while it had no effect in stool samples. These findings remained significant when adjusted by stage, smoking status and antibiotic use. Conclusions Baseline oral and gut microbiomes differ by stage in this HPV+ cohort. CRT caused a shift towards a gut-like microbiome composition in oropharyngeal swabs. Stage-specific features and the transitions in oral microbiome might have prognostic and therapeutic implications.
Collapse
Affiliation(s)
- Marc Oliva
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, Canada.,Department of Medical Oncology, Catalan Institute of Oncology (ICO), Barcelona, Spain
| | - Pierre H H Schneeberger
- Division of Infectious Diseases and Toronto General Hospital Research Institute, University Health Network, Departments of Medicine, Immunology and Laboratory of Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Victor Rey
- Division of Infectious Diseases and Toronto General Hospital Research Institute, University Health Network, Departments of Medicine, Immunology and Laboratory of Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Matthew Cho
- Division of Infectious Diseases and Toronto General Hospital Research Institute, University Health Network, Departments of Medicine, Immunology and Laboratory of Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Rachel Taylor
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, Canada
| | - Aaron R Hansen
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, Canada
| | - Kirsty Taylor
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, Canada
| | - Ali Hosni
- Department of Radiation Oncology, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, Canada
| | - Andrew Bayley
- Department of Radiation Oncology, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, Canada
| | - Andrew J Hope
- Department of Radiation Oncology, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, Canada
| | - Scott V Bratman
- Department of Radiation Oncology, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, Canada
| | - Jolie Ringash
- Department of Radiation Oncology, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, Canada
| | - Simron Singh
- Department of Pathology, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, Canada
| | - Ilan Weinreb
- Department of Medical Oncology, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Bayardo Perez-Ordoñez
- Department of Medical Oncology, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Douglas Chepeha
- Department of Surgical Oncology, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, Canada
| | - John Waldron
- Department of Radiation Oncology, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, Canada
| | - Wei Xu
- Department of Biostatistics, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, Canada
| | - David Guttman
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada
| | - Lillian L Siu
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, Canada
| | - Bryan Coburn
- Division of Infectious Diseases and Toronto General Hospital Research Institute, University Health Network, Departments of Medicine, Immunology and Laboratory of Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Anna Spreafico
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
18
|
Oral delivery of bacteria: Basic principles and biomedical applications. J Control Release 2020; 327:801-833. [DOI: 10.1016/j.jconrel.2020.09.011] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Accepted: 09/05/2020] [Indexed: 12/18/2022]
|
19
|
Wang HC, Yeh TJ, Chan LP, Hsu CM, Cho SF. Exploration of Feasible Immune Biomarkers for Immune Checkpoint Inhibitors in Head and Neck Squamous Cell Carcinoma Treatment in Real World Clinical Practice. Int J Mol Sci 2020; 21:E7621. [PMID: 33076306 PMCID: PMC7589088 DOI: 10.3390/ijms21207621] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 10/12/2020] [Accepted: 10/13/2020] [Indexed: 02/06/2023] Open
Abstract
Recurrent locally advanced or metastatic head and neck squamous cell carcinoma (HNSCC) is associated with dismal prognosis because of its highly invasive behavior and resistance to conventional intensive chemotherapy. The combination of targeted therapy and conventional chemotherapy has significantly improved clinical outcomes. In recent years, the development of immunotherapies, such as immune checkpoint inhibitors (ICIs), has further increased treatment responses and prolonged survival. However, the limited response rate, risk of immunotherapy-related adverse effects and high cost of immunotherapy make the identification of predictive markers to optimize treatment efficacy a critical issue. Biomarkers are biological molecules that have been widely utilized to predict treatment response to certain treatments and clinical outcomes or to detect disease. An ideal biomarker should exhibit good predictive ability, which can guide healthcare professionals to achieve optimal treatment goals and bring clinical benefit to patients. In this review, we summarized the results of recent and important studies focused on HNSCC ICI immunotherapy and discussed potential biomarkers including their strengths and limitations, aiming to gain more insight into HNSCC immunotherapy in real world clinical practice.
Collapse
Affiliation(s)
- Hui-Ching Wang
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (H.-C.W.); (T.-J.Y.); (L.-P.C.)
- Division of Hematology and Oncology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
- Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Tsung-Jang Yeh
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (H.-C.W.); (T.-J.Y.); (L.-P.C.)
- Division of Hematology and Oncology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
| | - Leong-Perng Chan
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (H.-C.W.); (T.-J.Y.); (L.-P.C.)
- Department of Otolaryngology-Head and Neck Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Chin-Mu Hsu
- Division of Hematology and Oncology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
| | - Shih-Feng Cho
- Division of Hematology and Oncology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
- Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| |
Collapse
|
20
|
Deng X, Tian H, Yang R, Han Y, Wei K, Zheng C, Liu Z, Chen T. Oral Probiotics Alleviate Intestinal Dysbacteriosis for People Receiving Bowel Preparation. Front Med (Lausanne) 2020; 7:73. [PMID: 32181256 PMCID: PMC7059130 DOI: 10.3389/fmed.2020.00073] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Accepted: 02/19/2020] [Indexed: 12/13/2022] Open
Abstract
Background: Bowel preparation is necessary for successful colonoscopy, while it can seriously affect intestinal microbial composition and damage the intestinal mucosal barriers in humans. Methods: To figure out whether probiotics can sustain intestinal homeostasis and guard people's health, the probiotic drug of Bifidobacterium Tetragenous viable Bacteria Tablets (P group, n = 16) or placebo (C group, n = 16) was used for volunteers receiving bowel preparation, and high-throughput sequencing method was applied to monitor their intestinal microbial changes. Results: The present results suggested that bowel preparation obviously reduced the intestinal microbial diversity, while taking probiotics significantly restored it to normal level. In addition, probiotics sharply reduced the abundance of pathogenic Proteobacteria, and obviously lowered the ratio of Firmicutes/Bacteroidetes compared with control group at phylum level (P < 0.05). And probiotics markedly decreased the abundance of pathogenic Acinetobacter and Streptococcus, while greatly enriched the relative abundance of beneficial bacteria Bacteroides, Roseburia, Faecalibacterium, and Parabacteroides at genus level (P < 0.05). Conclusion: Probiotic drugs, e.g., Bifidobacterium Tetragenous viable Bacteria Tablets, can be used to restore intestinal dysbacteriosis caused by bowel preparation, and reduce side effects during colonoscopy.
Collapse
Affiliation(s)
- Xiaorong Deng
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Huakai Tian
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Rong Yang
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yiwen Han
- National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, China
| | - Kehong Wei
- National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, China
| | - Cihua Zheng
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Zhaoxia Liu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Tingtao Chen
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China.,National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, China
| |
Collapse
|
21
|
Zhang X, Liu Q, Liao Q, Zhao Y. Pancreatic Cancer, Gut Microbiota, and Therapeutic Efficacy. J Cancer 2020; 11:2749-2758. [PMID: 32226493 PMCID: PMC7086274 DOI: 10.7150/jca.37445] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2019] [Accepted: 01/04/2020] [Indexed: 01/18/2023] Open
Abstract
Pancreatic cancer remains one of the leading causes of cancer-related death worldwide and has a poor prognosis. Current treatment relies on surgical resection and adjuvant therapies. The gut microbiota plays important roles in metabolism and immunomodulation. Accumulating evidence has implied that the gut microbiota is involved in the metabolism of chemotherapeutic drugs and the tumor microenvironment (TME), which could affect the efficacy of both conventional chemotherapy and immunotherapy for pancreatic cancer. Herein, we comprehensively reviewed the history and highlights of the interactions among pancreatic cancer, the gut microbiota and therapeutic efficacy and showed the promising future of manipulating the gut microbiota to improve clinical outcomes of pancreatic cancer.
Collapse
Affiliation(s)
- Xiang Zhang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Qiaofei Liu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Quan Liao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Yupei Zhao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| |
Collapse
|
22
|
Shui L, Yang X, Li J, Yi C, Sun Q, Zhu H. Gut Microbiome as a Potential Factor for Modulating Resistance to Cancer Immunotherapy. Front Immunol 2020; 10:2989. [PMID: 32010123 PMCID: PMC6978681 DOI: 10.3389/fimmu.2019.02989] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 12/05/2019] [Indexed: 02/05/2023] Open
Abstract
Gut microbiota refers to the diverse community of more than 100 trillion microorganisms residing in our intestines. It is now known that any shift in the composition of gut microbiota from that present during the healthy state in an individual is associated with predisposition to multiple pathological conditions, such as diabetes, autoimmunity, and even cancer. Currently, therapies targeting programmed cell death protein 1/programmed cell death 1 ligand 1 or cytotoxic T-lymphocyte antigen-4 are the focus of cancer immunotherapy and are widely applied in clinical treatment of various tumors. Owing to relatively low overall response rate, however, it has been an ongoing research endeavor to identify the mechanisms or factors for improving the therapeutic efficacy of these immunotherapies. Other than causing mutations that affect gene expression, some gut bacteria may also activate or repress the host's response to immune checkpoint inhibitors. In this review, we have described recent advancements made in understanding the regulatory relationship between gut microbiome and cancer immunotherapy. We have also summarized the potential molecular mechanisms behind this interaction, which can serve as a basis for utilizing different kinds of gut bacteria as promising tools for reversing immunotherapy resistance in cancer.
Collapse
Affiliation(s)
- Lin Shui
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Xi Yang
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Jian Li
- Department of Pharmacy, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
| | - Cheng Yi
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Qin Sun
- Drug Research Center, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
| | - Hong Zhu
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
23
|
Wang L, Shang Q, Guo W, Wu X, Wu L, Wu L, Chen T. Evaluation of the hypoglycemic effect of probiotics via directly consuming glucose in intestines of STZ-induced diabetic mice and glucose water-induced diabetic mice. J Funct Foods 2020. [DOI: 10.1016/j.jff.2019.103614] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|
24
|
Yu Y, Dunaway S, Champer J, Kim J, Alikhan A. Changing our microbiome: probiotics in dermatology. Br J Dermatol 2019; 182:39-46. [PMID: 31049923 DOI: 10.1111/bjd.18088] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/26/2019] [Indexed: 12/13/2022]
Abstract
BACKGROUND Commensal bacteria are a major factor in human health and disease pathogenesis. Interest has recently expanded beyond the gastrointestinal microbiome to include the skin microbiome and its impact on various skin diseases. OBJECTIVES Here we present current data reviewing the role of the microbiome in dermatology, considering both the gut and skin microflora. Our objective was to evaluate whether the clinical data support the utility of oral and topical probiotics for certain dermatological diseases. METHODS The PubMed and ClinicalTrials.gov databases were searched for basic science, translational research and clinical studies that investigated differences in the cutaneous microbiome and the impact of probiotics in patients with atopic dermatitis, acne vulgaris, psoriasis, chronic wounds, seborrhoeic dermatitis and cutaneous neoplasms. RESULTS Few clinical trials exist that explore the utility of probiotics for the prevention and treatment of dermatological diseases, with the exception of atopic dermatitis. Most studies investigated oral probiotic interventions, and of those utilizing topical probiotics, few included skin commensals. In general, the available clinical trials yielded positive results with improvement of the skin conditions after probiotic intervention. CONCLUSIONS Oral and topical probiotics appear to be effective for the treatment of certain inflammatory skin diseases and demonstrate a promising role in wound healing and skin cancer. However, more studies are needed to confirm these results. What's already known about this topic? The microbiome plays a role in human health and disease pathogenesis. Probiotics can manipulate the host microbiome and may confer health benefits for patients. Research to date has already begun to explore the utility of oral and topical probiotics for certain dermatological diseases. What does this study add? This review presents basic science and clinical trial data to support the role of the gut and skin microbiome in dermatology. Current data are reviewed on the use of probiotics in the prevention and treatment of skin diseases, including atopic dermatitis, acne vulgaris, psoriasis, seborrhoeic dermatitis, chronic wounds and cutaneous neoplasms. Future probiotic interventions are proposed.
Collapse
Affiliation(s)
- Y Yu
- Department of Dermatology, University of Cincinnati, Cincinnati, OH, U.S.A
| | - S Dunaway
- Department of Dermatology, University of Cincinnati, Cincinnati, OH, U.S.A
| | - J Champer
- Department of Computational Biology and Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY, U.S.A
| | - J Kim
- Division of Dermatology and Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, U.S.A
| | - A Alikhan
- Sutter Medical Foundation, Sacramento, CA, U.S.A
| |
Collapse
|
25
|
Zhou H, Suo J, Zhu J. [Therapeutic Relevance of Human Microbiota and Lung Cancer]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2019; 22:464-469. [PMID: 31315786 PMCID: PMC6712272 DOI: 10.3779/j.issn.1009-3419.2019.07.09] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
人体菌群与人类健康状态密切相关,如人体菌群的失调可能导致糖尿病、胃肠道疾病、肥胖等疾病的发生。人体内微生物与约20%的恶性肿瘤有关,肺癌(lung cancer, LC)是目前最为常见的恶性肿瘤之一,我国男性LC发病率及死亡率高居所有恶性肿瘤之首。近来研究表明,人体菌群可能通过代谢、炎症或免疫等途径影响着LC的发生,同时影响LC对放化疗、基因治疗、免疫治疗等治疗方法的疗效,如免疫治疗,是用于治疗LC的一种极有前景的手段,但不同患者从中获益不一,包含以肺癌细胞株的实验表明肠道微生物群可通过与宿主免疫系统的相互作用调节对免疫治疗的反应。但针对肺癌患者,肠道菌群是否仍能对免疫治疗进行调节仍存在争议。本文就人体菌群与LC的治疗相关性的近来研究进展进行综述。
Collapse
Affiliation(s)
- Huijie Zhou
- Department of Thoracic Oncology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jiaojiao Suo
- Department of Thoracic Oncology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jiang Zhu
- Department of Thoracic Oncology, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
26
|
Shi L, Sheng J, Wang M, Luo H, Zhu J, Zhang B, Liu Z, Yang X. Combination Therapy of TGF-β Blockade and Commensal-derived Probiotics Provides Enhanced Antitumor Immune Response and Tumor Suppression. Am J Cancer Res 2019. [PMID: 31281535 DOI: 10.7150/thno.35131.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Galunisertib (Gal) is a transforming growth factor (TGF-β) blockade which is being investigated as a potential tumor immunotherapy candidate drug in clinical trials. However, primary or acquired resistance is often found in the recruited cancer patients, which limits its clinical application. Tumor immune microenvironment can be regulated by intestinal microbiota, leading to different therapeutic outcomes. It is hypothesized that manipulation of cancer patients' intestinal microbiome in the early stage of therapy may be a promising strategy to improve the therapeutic efficacy of Gal. Methods: 4T1 and H22 subcutaneous tumor bearing mice were used to evaluate the therapeutic effect. Escherichia coli strain Nissle 1917 (EcN), a widely used probiotic bacteria, was orally delivered to the tumor bearing mice daily along with Gal treatment. Antitumor effect of the combination therapy was evaluated by tumor volume, histological staining of tumor tissues. Furthermore, flow cytometry was performed to analyze the alteration of immune microenvironment in tumor bed after treatment. The suppressing effect of the combination therapy on tumor invasiveness and metastasis was evaluated in both mice and zebrafish xenografts models. Fecal sample 16S rRNA gene sequencing was conducted to analyze changes of intestinal microbial diversity. The effect of intestinal microbiota on tumor suppression after receiving EcN was further tested by fecal transplant. Results: The therapeutic outcomes in tumor growth inhibition and metastasis suppression of Gal were significantly potentiated by EcN, resulting from the strengthened antitumor immunity. EcN was able to relieve the immunosuppressive tumor microenvironment, which was evidenced by enhanced tumor-specific effector T cells infiltration and dendritic cells activation. Intestinal microbiota was modulated by EcN, illustrated by a shift of gut microbiome toward certain beneficial bacteria. Conclusion: These results suggested that Gal combined with EcN might be a novel therapeutic approach with great potential of clinical implications for cancer prevention or treatment.
Collapse
Affiliation(s)
- Linlin Shi
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Jianyong Sheng
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Mengli Wang
- Key Laboratory of Molecular Biophysics of Ministry of Education, Center for Human Genome Research, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Han Luo
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Jun Zhu
- Department of Biotechnology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China.,Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Bixiang Zhang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Zhi Liu
- Department of Biotechnology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Xiangliang Yang
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| |
Collapse
|
27
|
Shi L, Sheng J, Wang M, Luo H, Zhu J, Zhang B, Liu Z, Yang X. Combination Therapy of TGF-β Blockade and Commensal-derived Probiotics Provides Enhanced Antitumor Immune Response and Tumor Suppression. Theranostics 2019; 9:4115-4129. [PMID: 31281535 PMCID: PMC6592171 DOI: 10.7150/thno.35131] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 05/07/2019] [Indexed: 12/23/2022] Open
Abstract
Galunisertib (Gal) is a transforming growth factor (TGF-β) blockade which is being investigated as a potential tumor immunotherapy candidate drug in clinical trials. However, primary or acquired resistance is often found in the recruited cancer patients, which limits its clinical application. Tumor immune microenvironment can be regulated by intestinal microbiota, leading to different therapeutic outcomes. It is hypothesized that manipulation of cancer patients' intestinal microbiome in the early stage of therapy may be a promising strategy to improve the therapeutic efficacy of Gal. Methods: 4T1 and H22 subcutaneous tumor bearing mice were used to evaluate the therapeutic effect. Escherichia coli strain Nissle 1917 (EcN), a widely used probiotic bacteria, was orally delivered to the tumor bearing mice daily along with Gal treatment. Antitumor effect of the combination therapy was evaluated by tumor volume, histological staining of tumor tissues. Furthermore, flow cytometry was performed to analyze the alteration of immune microenvironment in tumor bed after treatment. The suppressing effect of the combination therapy on tumor invasiveness and metastasis was evaluated in both mice and zebrafish xenografts models. Fecal sample 16S rRNA gene sequencing was conducted to analyze changes of intestinal microbial diversity. The effect of intestinal microbiota on tumor suppression after receiving EcN was further tested by fecal transplant. Results: The therapeutic outcomes in tumor growth inhibition and metastasis suppression of Gal were significantly potentiated by EcN, resulting from the strengthened antitumor immunity. EcN was able to relieve the immunosuppressive tumor microenvironment, which was evidenced by enhanced tumor-specific effector T cells infiltration and dendritic cells activation. Intestinal microbiota was modulated by EcN, illustrated by a shift of gut microbiome toward certain beneficial bacteria. Conclusion: These results suggested that Gal combined with EcN might be a novel therapeutic approach with great potential of clinical implications for cancer prevention or treatment.
Collapse
Affiliation(s)
- Linlin Shi
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Jianyong Sheng
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Mengli Wang
- Key Laboratory of Molecular Biophysics of Ministry of Education, Center for Human Genome Research, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Han Luo
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Jun Zhu
- Department of Biotechnology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Bixiang Zhang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Zhi Liu
- Department of Biotechnology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Xiangliang Yang
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| |
Collapse
|
28
|
Abu-Sbeih H, Ali FS, Wang X, Mallepally N, Chen E, Altan M, Bresalier RS, Charabaty A, Dadu R, Jazaeri A, Lashner B, Wang Y. Early introduction of selective immunosuppressive therapy associated with favorable clinical outcomes in patients with immune checkpoint inhibitor-induced colitis. J Immunother Cancer 2019; 7:93. [PMID: 30940209 PMCID: PMC6444537 DOI: 10.1186/s40425-019-0577-1] [Citation(s) in RCA: 137] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 03/22/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Current treatment guidelines for immune-mediated colitis (IMC) recommend 4 to 6 weeks of steroids as first-line therapy, followed by selective immunosuppressive therapy (SIT) (infliximab or vedolizumab) in patients who do not respond to steroids. We assessed the effect of early SIT introduction and number of SIT infusions on clinical outcomes. METHODS We performed a retrospective review of patients with IMC who received SIT at The University of Texas MD Anderson Cancer Center between January and December 2018. Logistic regression analyses were used to assess associations between clinical outcomes and features of IMC. RESULTS Of the 1459 patients who received immune checkpoint inhibitors, 179 developed IMC of any grade; 84 of these 179 patients received SIT. Of the 84 patients who received SIT, 79% were males, and the mean age was 60 years (standard deviation, 14). Compared with patients who received SIT > 10 days after IMC onset, patients who received early SIT (≤10 days) required fewer hospitalizations (P = 0.03), experienced steroid taper failure less frequently (P = 0.03), had fewer steroid tapering attempts (P < 0.01), had a shorter course of steroid treatment (P = 0.09), and had a shorter duration of symptoms (P < 0.01). Patients who received one or two infusions of SIT achieved histologic remission less frequently (P = 0.09) and had higher fecal calprotectin levels after SIT (P = 0.01) compared with patients who received three or more infusions. Risk factors for IMC recurrence after weaning off steroids included: 1) needing multiple hospitalizations, 2) experiencing steroid taper failure after SIT, 3) receiving infliximab rather than vedolizumab, 4) receiving fewer than three infusions of SIT, 5) having higher fecal calprotectin levels after SIT, and 6) receiving a longer course of steroids, hospitalization and IMC symptoms. Unsuccessful weaning from steroids after SIT was associated with high IMC grades; multiple hospitalizations; steroid-resistant IMC; long interval from IMC to SIT initiation; and long duration of steroids, IMC symptoms, and hospitalization. CONCLUSION SIT should be introduced early in the disease course of IMC instead of waiting until failure of steroid therapy or steroid taper. Patients who received three or more infusions of SIT had more favorable clinical outcomes.
Collapse
Affiliation(s)
- Hamzah Abu-Sbeih
- Department of Gastroenterology, Hepatology and Nutrition, The University of Texas MD Anderson Cancer Center, Unit 1466, 1515 Holcombe Blvd, Houston, TX 77030 USA
| | - Faisal S. Ali
- Department of Gastroenterology, Hepatology and Nutrition, The University of Texas MD Anderson Cancer Center, Unit 1466, 1515 Holcombe Blvd, Houston, TX 77030 USA
| | - Xuemei Wang
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX USA
| | | | - Ellie Chen
- Department of Medicine, Baylor College of Medicine, Houston, TX USA
| | - Mehmet Altan
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Robert S. Bresalier
- Department of Gastroenterology, Hepatology and Nutrition, The University of Texas MD Anderson Cancer Center, Unit 1466, 1515 Holcombe Blvd, Houston, TX 77030 USA
| | - Aline Charabaty
- Department of Gastroenterology, Hepatology and Nutrition, MedStar-Georgetown University Hospital, Washington, DC USA
| | - Ramona Dadu
- Department of Endocrine Neoplasia and Hormonal Disorders, The University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Amir Jazaeri
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Bret Lashner
- Department of Gastroenterology, Hepatology and Nutrition, Cleveland Clinic Foundation, Cleveland, OH USA
| | - Yinghong Wang
- Department of Gastroenterology, Hepatology and Nutrition, The University of Texas MD Anderson Cancer Center, Unit 1466, 1515 Holcombe Blvd, Houston, TX 77030 USA
| |
Collapse
|
29
|
Analyses of the possible anti-tumor effect of yokukansan. J Nat Med 2019; 73:468-479. [PMID: 30739283 DOI: 10.1007/s11418-019-01283-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 01/28/2019] [Indexed: 02/08/2023]
Abstract
The Kampo medicine yokukansan (YKS) has a wide variety of properties such as anxiolytic, anti-inflammatory and analgesic effects, and is also thought to regulate tumor suppression. In this study, we investigated the anti-tumor effect of YKS. We used Lewis lung carcinoma (LLC)-bearing mice that were fed food pellets containing YKS and then performed a fecal microbiota analysis, a microarray analysis for microRNAs (miRNAs) and an in vitro anti-tumor assay. The fecal microbiota analysis revealed that treatment with YKS partly reversed changes in the microbiota composition due to LLC implantation. Furthermore, a miRNA array analysis using blood serum showed that treatment with YKS restored the levels of miR-133a-3p/133b-3p, miR-1a-3p and miR-342-3p following LLC implantation to normal levels. A TargetScan analysis revealed that the epidermal growth factor receptor 1 signaling pathway is one of the major target pathways for these miRNAs. Furthermore, treatment with YKS restored the levels of miR-200b-3p and miR-200c-3p, a recognized mediator of cancer progression and controller of emotion, in the hypothalamus of mice bearing LLC. An in vitro assay revealed that a mixture of pachymic acid, saikosaponins a and d and isoliquiritigenin, which are all contained in YKS, exerted direct and additive anti-tumor effects. The present findings constitute novel evidence that YKS may exert an anti-tumor effect by reversing changes in the fecal microbiota and miRNAs circulating in the blood serum and hypothalamus, and the compounds found in YKS could have direct and additive anti-tumor effects.
Collapse
|
30
|
Oliva M, Spreafico A, Taberna M, Alemany L, Coburn B, Mesia R, Siu LL. Immune biomarkers of response to immune-checkpoint inhibitors in head and neck squamous cell carcinoma. Ann Oncol 2019; 30:57-67. [PMID: 30462163 PMCID: PMC6336003 DOI: 10.1093/annonc/mdy507] [Citation(s) in RCA: 182] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Anti-programmed cell death protein 1 (PD-1) agents have become the standard of care for platinum-refractory recurrent/metastatic head and neck squamous cell carcinoma (HNSCC) and are currently being evaluated in various disease settings. However, despite the gain in overall survival seen in some of the clinical trials, the majority of patients display primary resistance and do not benefit from these agents. Taking into consideration the potentially severe immune-related toxicities and their high cost, the search for predictive biomarkers of response is crucial. Besides Programmed death ligand-1 (PD-L1) expression, other biomarkers such as immune infiltration, tumor mutational burden or immune-gene expression profiling have been explored, but none of them has been validated in this disease. Among these, the microbiota has recently garnered tremendous interest since it has proven to influence the efficacy of PD-1 blockade in some tumor types. With the accumulating evidence on the effect of the microbiota in HNSCC tumorigenesis and progression, the study of its potential role as a predictive immune biomarker is warranted. This review examines the available evidence on emerging immune predictive biomarkers of response to anti-PD-1/PD-L1 therapy in HNSCC, introducing the microbiota and its potential use as a predictive immune biomarker in this disease.
Collapse
Affiliation(s)
- M Oliva
- Division of Medical Oncology and Haematology, Princess Margaret Cancer Centre, Toronto; University of Toronto, Toronto, Canada
| | - A Spreafico
- Division of Medical Oncology and Haematology, Princess Margaret Cancer Centre, Toronto; University of Toronto, Toronto, Canada
| | - M Taberna
- Medical Oncology Department, Catalan Institute of Oncology (ICO), ONCOBELL-IDIBELL, L'Hospitalet de Llobregat, Barcelona; Barcelona University, Barcelona
| | - L Alemany
- Cancer Epidemiology Research Program, Catalan Institute of Oncology (ICO), IDIBELL, L'Hospitalet de Llobregat, Barcelona; CIBER in Epidemiology and Public Health (CIBERESP), Barcelona, Spain
| | - B Coburn
- Division of Infectious Diseases, University Health Network, Toronto; Departments of Medicine and Laboratory of Medicine and Pathobiology, University of Toronto, Toronto, Canada
| | - R Mesia
- Medical Oncology Department, B-ARGO Group, Catalan Institute of Oncology (ICO), Badalona, Spain
| | - L L Siu
- Division of Medical Oncology and Haematology, Princess Margaret Cancer Centre, Toronto; University of Toronto, Toronto, Canada.
| |
Collapse
|
31
|
Harnessing the immune system in glioblastoma. Br J Cancer 2018; 119:1171-1181. [PMID: 30393372 PMCID: PMC6251037 DOI: 10.1038/s41416-018-0258-8] [Citation(s) in RCA: 155] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2018] [Revised: 07/25/2018] [Accepted: 07/27/2018] [Indexed: 12/14/2022] Open
Abstract
Glioblastoma is the most common primary malignant brain tumour. Survival is poor and improved treatment options are urgently needed. Although immunotherapies have emerged as effective treatments for a number of cancers, translation of these through to brain tumours is a distinct challenge, particularly due to the blood-brain barrier and the unique immune tumour microenvironment afforded by CNS-specific cells. This review discusses the immune system within the CNS, mechanisms of immune escape employed by glioblastoma, and the immunological effects of conventional glioblastoma treatments. Novel therapies for glioblastoma that harness the immune system and their current clinical progress are outlined, including cancer vaccines, T-cell therapies and immune checkpoint modulators.
Collapse
|
32
|
Karabulutoglu M, Finnon R, Imaoka T, Friedl AA, Badie C. Influence of diet and metabolism on hematopoietic stem cells and leukemia development following ionizing radiation exposure. Int J Radiat Biol 2018; 95:452-479. [PMID: 29932783 DOI: 10.1080/09553002.2018.1490042] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE The review aims to discuss the prominence of dietary and metabolic regulators in maintaining hematopoietic stem cell (HSC) function, long-term self-renewal, and differentiation. RESULTS Most adult stem cells are preserved in a quiescent, nonmotile state in vivo which acts as a "protective state" for stem cells to reduce endogenous stress provoked by DNA replication and cellular respiration as well as exogenous environmental stress. The dynamic balance between quiescence, self-renewal and differentiation is critical for supporting a functional blood system throughout life of an organism. Stress-conditions, for example ionizing radiation exposure can trigger the blood forming HSCs to proliferate and migrate through extramedullary tissues to expand the number of HSCs and increase hematopoiesis. In addition, a wealth of investigation validated that deregulation of this balance plays a critical pathogenic role in various different hematopoietic diseases including the leukemia development. CONCLUSION The review summarizes the current knowledge on how alterations in dietary and metabolic factors could alter the risk of leukemia development following ionizing radiation exposure by inhibiting or even reversing the leukemic progression. Understanding the influence of diet, metabolism, and epigenetics on radiation-induced leukemogenesis may lead to the development of practical interventions to reduce the risk in exposed populations.
Collapse
Affiliation(s)
- Melis Karabulutoglu
- a Cancer Mechanisms and Biomarkers group, Biological Effects Department, Centre for Radiation, Chemical and Environmental Hazards , Public Health England , Didcot , UK.,b CRUK & MRC Oxford Institute for Radiation Oncology, Department of Oncology , University of Oxford , Oxford , UK
| | - Rosemary Finnon
- a Cancer Mechanisms and Biomarkers group, Biological Effects Department, Centre for Radiation, Chemical and Environmental Hazards , Public Health England , Didcot , UK
| | - Tatsuhiko Imaoka
- c Department of Radiation Effects Research, National Institute of Radiological Sciences , National Institutes for Quantum and Radiological Science and Technology , Chiba , Japan
| | - Anna A Friedl
- d Department of Radiation Oncology , University Hospital, LMU Munich , Munich , Germany
| | - Christophe Badie
- a Cancer Mechanisms and Biomarkers group, Biological Effects Department, Centre for Radiation, Chemical and Environmental Hazards , Public Health England , Didcot , UK
| |
Collapse
|
33
|
Chen H, Luo T, Chen T, Wang G. Seminal bacterial composition in patients with obstructive and non-obstructive azoospermia. Exp Ther Med 2018; 15:2884-2890. [PMID: 29456693 PMCID: PMC5795641 DOI: 10.3892/etm.2018.5778] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Accepted: 10/13/2017] [Indexed: 12/14/2022] Open
Abstract
A number of culture-dependent and -independent studies have reported that the number and significance of bacterial species in semen may have been underestimated. The aim of the present study was to profile the seminal microbiome in patients with obstructive or non-obstructive azoospermia. A high-throughput sequencing method was adopted to sequence genomic DNA extracted from the semen of healthy people (C group), patients with obstructive azoospermia (OA group) and patients with non-obstructive azoospermia (NOA group). The results revealed that Firmicutes, Proteobacteria, Bacteroidetes and Actinobacteria species comprised the majority of bacteria in the C (98.14%), OA (98.26%) and NOA (90.96%) groups. Patients in the OA and NOA groups exhibited an increase in Bacteroidetes and Firmicutes, whereas the number of Proteobacteria and Actinobacteria were decreased compared with the C group. A total of 398 common operational taxonomic units were identified, of which 27 belonged to the genus Lactobacillus. Furthermore, Phylogenetic Investigation of Communities by Reconstruction of Unobserved States analysis indicated that the pathogenic species and reduced biodiversity in the semen of patients with azoospermia may result in an increased risk of metabolic, infectious and immune diseases. In the present study, the seminal microbiome of patients with obstructive or non-obstructive azoospermia was explored, which may be useful for developing novel treatments against azoospermia as well as for its diagnosis.
Collapse
Affiliation(s)
- Houyang Chen
- Department of Urology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China.,Reproductive Medical Center, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, Jiangxi 330006, P.R. China
| | - Tao Luo
- Institute of Life Science, Nanchang University, Nanchang, Jiangxi 330031, P.R. China
| | - Tingtao Chen
- Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi 330031, P.R. China
| | - Gongxian Wang
- Department of Urology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
34
|
Kuczma MP, Ding ZC, Li T, Habtetsion T, Chen T, Hao Z, Bryan L, Singh N, Kochenderfer JN, Zhou G. The impact of antibiotic usage on the efficacy of chemoimmunotherapy is contingent on the source of tumor-reactive T cells. Oncotarget 2017; 8:111931-111942. [PMID: 29340102 PMCID: PMC5762370 DOI: 10.18632/oncotarget.22953] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 11/26/2017] [Indexed: 12/11/2022] Open
Abstract
In recent years the combined use of chemotherapy and immunotherapy, collectively termed chemoimmunotherapy, has emerged as a promising treatment option for patients with cancer. Antibiotics are commonly used to reduce infection-related complications in patients undergoing chemotherapy. Intriguingly, accumulating evidence has implicated gut microbiota as a critical determinant of host antitumor immune responses, raising the question as to whether the use of broad-spectrum antibiotics would invariably diminish tumor response to chemoimmunotherapies. We investigated the impact of antibiotics on the therapeutic outcomes of cyclophosphamide (CTX) chemotherapy and adoptive T-cell therapy (ACT) where CTX was used as the host-conditioning regimen in mice. We show that antibiotic prophylaxis dampened the endogenous T cell responses elicited by CTX, and reduced the efficacy of CTX against B-cell lymphoma. In the ACT setting, antibiotics administration impaired the therapeutic effects of adoptively transferred tumor-specific CD4+ T cells in mice with implanted colorectal tumors. In contrast, long-term antibiotic exposure did not affect the efficacy of ACT using CD19-targeting chimeric antigen receptor (CAR) T cells in mice with systemic B-cell lymphoma, although it correlated with prolonged CAR expression and sustained B-cell aplasia. Our study demonstrates that chemoimmunotherapies may have variable reliance on intestinal microbiota for T cell activation and function, and thus have different sensitivities to antibiotic prophylaxis. These findings may have implications for the judicial use of antibiotics in cancer patients receiving chemoimmunotherapies.
Collapse
Affiliation(s)
- Michal P Kuczma
- Georgia Cancer Center, Augusta University, Augusta, Georgia, USA.,Current/Present address: Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia, USA
| | - Zhi-Chun Ding
- Georgia Cancer Center, Augusta University, Augusta, Georgia, USA
| | - Tao Li
- Department of Oncology and Surgery, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Province, PR China
| | | | - Tingting Chen
- Georgia Cancer Center, Augusta University, Augusta, Georgia, USA
| | - Zhonglin Hao
- Georgia Cancer Center, Augusta University, Augusta, Georgia, USA
| | - Locke Bryan
- Hematology/Oncology, Georgia Cancer Center, Augusta University, Augusta, Georgia, USA
| | - Nagendra Singh
- Department of Biochemistry and Molecular Biology, Augusta University, Augusta, Georgia, USA
| | - James N Kochenderfer
- Experimental Transplantation and Immunology Branch, National Cancer Institute, Bethesda, Maryland, USA
| | - Gang Zhou
- Georgia Cancer Center, Augusta University, Augusta, Georgia, USA
| |
Collapse
|
35
|
Zhao X, Chen T, Meng F, Wang H, Tian P, Tang X, Wang X, Wang X, Xin H, Wei H. Therapeutic effect of herb residue fermentation supernatant on spleen‑deficient mice. Mol Med Rep 2017; 17:2764-2770. [PMID: 29207096 DOI: 10.3892/mmr.2017.8150] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 11/03/2017] [Indexed: 11/05/2022] Open
Abstract
To minimize the waste of active ingredients present in herb residues, in the present study, probiotics of Bacillus subtilis, Aspergillus oryzae and Lactobacillus plantarum M3 were selected to reuse herb residues from Jianweixiaoshi tablets, and the therapeutic effects of the herb residue fermentation supernatant were evaluated using a spleen‑deficient mouse model. The results of the present study indicated that the fermentation supernatant may effectively improve the immunity of mice, as measured by body weight, spleen and thymus index, and inflammatory cytokines, including interleukin (IL)‑2, IL‑4 and interferon‑γ. The viable cell count and denaturing gradient gel electrophoresis results indicated that the fermentation supernatant markedly enhanced bacterial diversity and the number of lactobacilli in mouse intestines. Therefore, the combination of the Jianweixiaoshi herb residue and probiotics provided a novel method to reuse herb residues and may in the future contribute to the treatment of spleen deficiency.
Collapse
Affiliation(s)
- Xiaoxiao Zhao
- Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi 330031, P.R. China
| | - Tingtao Chen
- Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi 330031, P.R. China
| | - Fanjing Meng
- Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi 330031, P.R. China
| | - Huan Wang
- School of Life Sciences, Nanchang University, Nanchang, Jiangxi 330047, P.R. China
| | - Puyuan Tian
- School of Life Sciences, Nanchang University, Nanchang, Jiangxi 330047, P.R. China
| | - Xianyao Tang
- Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi 330031, P.R. China
| | - Xin Wang
- School of Life Sciences, Nanchang University, Nanchang, Jiangxi 330047, P.R. China
| | - Xiaolei Wang
- Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi 330031, P.R. China
| | - Hongbo Xin
- Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi 330031, P.R. China
| | - Hua Wei
- Jiangxi‑OAI Joint Research Institute, Nanchang University, Nanchang, Jiangxi 330047, P.R. China
| |
Collapse
|
36
|
Bull JMC. A review of immune therapy in cancer and a question: can thermal therapy increase tumor response? Int J Hyperthermia 2017; 34:840-852. [PMID: 28974121 DOI: 10.1080/02656736.2017.1387938] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Immune therapy is a successful cancer treatment coming into its own. This is because checkpoint molecules, adoptive specific lymphocyte transfer and chimeric antigen T-cell (CAR-T) therapy are able to induce more durable responses in an increasing number of malignancies compared to chemotherapy. In addition, immune therapies are able to treat bulky disease, whereas standard cytotoxic therapies cannot treat large tumour burdens. Checkpoint inhibitor monoclonal antibodies are becoming widely used in the clinic and although more complex, adoptive lymphocyte transfer and CAR-T therapies show promise. We are learning that there are nuances to predicting the successful use of the checkpoint inhibitors as well as to specific-antigen adoptive and CAR-T therapies. We are also newly aware of a here-to-fore unrealised natural force, the status of the microbiome. However, despite better understanding of mechanisms of action of the new immune therapies, the best responses to the new immune therapies remain 20-30%. Likely the best way to improve this somewhat low response rate for patients is to increase the patient's own immune response. Thermal therapy is a way to do this. All forms of thermal therapy, from fever-range systemic thermal therapy, to high-temperature HIFU and even cryotherapy improve the immune response pre-clinically. It is time to test the immune therapies with thermal therapy in vivo to test for optimal timing of the combinations that will best enhance tumour response and then to begin to test the immune therapies with thermal therapy in the clinic as soon as possible.
Collapse
Affiliation(s)
- Joan M C Bull
- a Division of Oncology, Department of Internal Medicine , The University of Texas Medical School at Houston , Houston , TX , USA
| |
Collapse
|
37
|
Neal LR, Bailey SR, Wyatt MM, Bowers JS, Majchrzak K, Nelson MH, Haupt C, Paulos CM, Varela JC. The Basics of Artificial Antigen Presenting Cells in T Cell-Based Cancer Immunotherapies. JOURNAL OF IMMUNOLOGY RESEARCH AND THERAPY 2017; 2:68-79. [PMID: 28825053 PMCID: PMC5560309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Adoptive T cell transfer (ACT) can mediate objective responses in patients with advanced malignancies. There have been major advances in this field, including the optimization of the ex vivo generation of tumor-reactive lymphocytes to ample numbers for effective ACT therapy via the use of natural and artificial antigen presenting cells (APCs). Herein we review the basic properties of APCs and how they have been manufactured through the years to augment vaccine and T cell-based cancer therapies. We then discuss how these novel APCs impact the function and memory properties of T cells. Finally, we propose new ways to synthesize aAPCs to augment the therapeutic effectiveness of antitumor T cells for ACT therapy.
Collapse
Affiliation(s)
- Lillian R. Neal
- Department of Microbiology & Immunology, Medical University of South Carolina, Charleston, SC 29425
- Department of Dermatological Surgery and Dermatology, Medical University of South Carolina, Charleston, SC 29425
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29425
- Department of Hematology and Oncology, Medical University of South Carolina, Charleston, 29425
| | - Stefanie R. Bailey
- Department of Microbiology & Immunology, Medical University of South Carolina, Charleston, SC 29425
- Department of Dermatological Surgery and Dermatology, Medical University of South Carolina, Charleston, SC 29425
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29425
| | - Megan M. Wyatt
- Department of Microbiology & Immunology, Medical University of South Carolina, Charleston, SC 29425
- Department of Dermatological Surgery and Dermatology, Medical University of South Carolina, Charleston, SC 29425
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29425
| | - Jacob S. Bowers
- Department of Microbiology & Immunology, Medical University of South Carolina, Charleston, SC 29425
- Department of Dermatological Surgery and Dermatology, Medical University of South Carolina, Charleston, SC 29425
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29425
| | - Kinga Majchrzak
- Department of Microbiology & Immunology, Medical University of South Carolina, Charleston, SC 29425
- Department of Dermatological Surgery and Dermatology, Medical University of South Carolina, Charleston, SC 29425
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29425
| | - Michelle H. Nelson
- Department of Microbiology & Immunology, Medical University of South Carolina, Charleston, SC 29425
- Department of Dermatological Surgery and Dermatology, Medical University of South Carolina, Charleston, SC 29425
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29425
| | - Carl Haupt
- Department of Microbiology & Immunology, Medical University of South Carolina, Charleston, SC 29425
- Department of Hematology and Oncology, Medical University of South Carolina, Charleston, 29425
| | - Chrystal M. Paulos
- Department of Microbiology & Immunology, Medical University of South Carolina, Charleston, SC 29425
- Department of Dermatological Surgery and Dermatology, Medical University of South Carolina, Charleston, SC 29425
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29425
| | - Juan C. Varela
- Department of Microbiology & Immunology, Medical University of South Carolina, Charleston, SC 29425
- Department of Hematology and Oncology, Medical University of South Carolina, Charleston, 29425
| |
Collapse
|
38
|
Champer M, Wong AM, Champer J, Brito IL, Messer PW, Hou JY, Wright JD. The role of the vaginal microbiome in gynaecological cancer. BJOG 2017; 125:309-315. [DOI: 10.1111/1471-0528.14631] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/05/2017] [Indexed: 12/13/2022]
Affiliation(s)
- M Champer
- Division of Gynecologic Oncology; Department of Obstetrics and Gynecology; Columbia University College of Physicians and Surgeons; New York NY USA
| | - AM Wong
- Department of Computer Science and Center for Computational Molecular Biology; Brown University; Providence RI USA
| | - J Champer
- Department of Biological Statistics and Computational Biology; Cornell University; Ithaca NY USA
- Department of Molecular Biology and Genetics; Cornell University; Ithaca NY USA
| | - IL Brito
- Department of Biomedical Engineering; Cornell University; Ithaca NY USA
| | - PW Messer
- Department of Biological Statistics and Computational Biology; Cornell University; Ithaca NY USA
| | - JY Hou
- Division of Gynecologic Oncology; Department of Obstetrics and Gynecology; Columbia University College of Physicians and Surgeons; New York NY USA
| | - JD Wright
- Division of Gynecologic Oncology; Department of Obstetrics and Gynecology; Columbia University College of Physicians and Surgeons; New York NY USA
| |
Collapse
|
39
|
Spaw M, Anant S, Thomas SM. Stromal contributions to the carcinogenic process. Mol Carcinog 2017; 56:1199-1213. [PMID: 27787930 PMCID: PMC5354948 DOI: 10.1002/mc.22583] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Revised: 10/14/2016] [Accepted: 10/24/2016] [Indexed: 12/20/2022]
Abstract
Tumor-associated stromal cells are dynamic characters that endorse the carcinogenic process in a multitude of ways. The tumor microenvironment plays a crucial role throughout the tumor progression, which includes initiation, growth, invasion, and metastasis. The tumor microenvironment consists of cellular and non-cellular components. Tumor-associated stromal cell types include the microbiome, immune cells including macrophages, dendritic and T-cells, cells associated with blood and lymphatic vessels including pericytes and endothelial cells, fibroblasts, neuronal cells, and adipocytes. The non-cellular components of the microenvironment include matrix proteins and secreted factors. The development of therapies that target the mechanisms by which stromal cells contribute to successful tumorigenesis is major goal of upcoming cancer research. The purpose of this review is to present a comprehensive discussion of the role of each of the tumor-associated stromal cell types in the carcinogenic process with a special focus on target development and therapeutic intervention. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Mark Spaw
- Department of Otolaryngology, University of Kansas Medical Center, Kansas City, Kansas
| | - Shrikant Anant
- Department of Surgery, University of Kansas Medical Center, Kansas City, Kansas
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, Kansas
| | - Sufi Mary Thomas
- Department of Otolaryngology, University of Kansas Medical Center, Kansas City, Kansas
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, Kansas
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, Kansas
| |
Collapse
|
40
|
Gadkaree SK, Fu J, Sen R, Korrer MJ, Allen C, Kim YJ. Induction of tumor regression by intratumoral STING agonists combined with anti-programmed death-L1 blocking antibody in a preclinical squamous cell carcinoma model. Head Neck 2017; 39:1086-1094. [PMID: 28323387 DOI: 10.1002/hed.24704] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2016] [Revised: 10/31/2016] [Accepted: 12/09/2016] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Cyclic dinucleotides (CDNs) are bacterial intracellular messengers that have demonstrated antitumor activity in melanoma and breast tumors, although their role in immunotherapy of head and neck squamous cell cancers (HNSCCs) has not been well investigated. METHODS We measured primary tumor growth rates, mechanism of antitumor activity, and efficacy of programmed death-L1 blockade combinatorial therapy in SCCFVII tumor-bearing C3H/HeOUJ mice undergoing intratumoral injections with RR-cyclic-di-guanine (synthetic CDG), CDG (natural cyclic-di-guanine), R848 (TLR 7/8 agonist), or phosphate buffered saline (PBS, control). RESULTS Intratumoral CDN treatment groups showed decreased tumor size and enhanced splenocyte Th1 response when compared to the PBS treatment control group (p < .05). The RR-CDG tumor microenvironment showed upregulated interferon (IFN)-γ+CD8+ and programmed death-L1. Combining programmed death-L1 blocking antibody with RR-CDG induced regression of established tumors. CONCLUSION This study demonstrates the antitumor effects of CDNs in a HNSCC cell line. These preclinical data strongly support the future clinical development of intratumoral CDN in patients with HNSCC. © 2017 Wiley Periodicals, Inc. Head Neck 39: 1086-1094, 2017.
Collapse
Affiliation(s)
- Shekhar K Gadkaree
- Department of Otolaryngology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - Juan Fu
- Department of Otolaryngology - Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Rupashree Sen
- Department of Otolaryngology - Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Michael J Korrer
- Department of Otolaryngology - Head and Neck Surgery, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Clint Allen
- Department of Otolaryngology - Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Young J Kim
- Department of Otolaryngology - Head and Neck Surgery, Vanderbilt University School of Medicine, Nashville, Tennessee
| |
Collapse
|
41
|
Chen KL, Jung P, Kulkoyluoglu-Cotul E, Liguori C, Lumibao J, Mazewski C, Ranard K, Rowles JL, Wang Y, Xue L, Madak-Erdogan Z. Impact of Diet and Nutrition on Cancer Hallmarks. ACTA ACUST UNITED AC 2017; 7. [PMID: 30581989 DOI: 10.15406/jcpcr.2017.07.00240] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Diet and nutrition are undeniably two factors that have a major impact on the prevention, progression, and treatment of various cancers. In this review, we will discuss how bioactives from diet and nutritional status affect each of the hallmarks of cancer. We will present recent research and discuss using diet and nutrition as a means to prevent and treat cancer.
Collapse
Affiliation(s)
- Karen L Chen
- Division of Nutritional Sciences, University of Illinois, USA
| | - Paul Jung
- Department of Food Science and Human Nutrition, University of Illinois, USA
| | | | - Carli Liguori
- Department of Food Science and Human Nutrition, University of Illinois, USA
| | - Jan Lumibao
- Division of Nutritional Sciences, University of Illinois, USA
| | - Candice Mazewski
- Department of Food Science and Human Nutrition, University of Illinois, USA
| | | | - Joe L Rowles
- Division of Nutritional Sciences, University of Illinois, USA
| | - Yanling Wang
- Department of Food Science and Human Nutrition, University of Illinois, USA
| | - Louisa Xue
- Division of Nutritional Sciences, University of Illinois, USA
| | - Zeynep Madak-Erdogan
- Division of Nutritional Sciences, University of Illinois, USA.,Department of Food Science and Human Nutrition, University of Illinois, USA
| |
Collapse
|
42
|
Ng SSM, Nagy BA, Jensen SM, Hu X, Alicea C, Fox BA, Felber BK, Bergamaschi C, Pavlakis GN. Heterodimeric IL15 Treatment Enhances Tumor Infiltration, Persistence, and Effector Functions of Adoptively Transferred Tumor-specific T Cells in the Absence of Lymphodepletion. Clin Cancer Res 2016; 23:2817-2830. [PMID: 27986749 DOI: 10.1158/1078-0432.ccr-16-1808] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Revised: 11/18/2016] [Accepted: 12/01/2016] [Indexed: 12/25/2022]
Abstract
Purpose: Adoptive cell transfer (ACT) is a promising immunotherapeutic approach for cancer. Host lymphodepletion is associated with favorable ACT therapy outcomes, but it may cause detrimental effects in humans. We tested the hypothesis that IL15 administration enhances ACT in the absence of lymphodepletion. We previously showed that bioactive IL15 in vivo comprises a stable complex of the IL15 chain with the IL15 receptor alpha chain (IL15Rα), termed heterodimeric IL15 (hetIL15).Experimental Design: We evaluated the effects of the combination regimen ACT + hetIL15 in the absence of lymphodepletion by transferring melanoma-specific Pmel-1 T cells into B16 melanoma-bearing mice.Results: hetIL15 treatment delayed tumor growth by promoting infiltration and persistence of both adoptively transferred Pmel-1 cells and endogenous CD8+ T cells into the tumor. In contrast, persistence of Pmel-1 cells was severely reduced following irradiation in comparison with mice treated with hetIL15. Importantly, we found that hetIL15 treatment led to the preferential enrichment of Pmel-1 cells in B16 tumor sites in an antigen-dependent manner. Upon hetIL15 administration, tumor-infiltrating Pmel-1 cells showed a "nonexhausted" effector phenotype, characterized by increased IFNγ secretion, proliferation, and cytotoxic potential and low level of PD-1. hetIL15 treatment also resulted in an improved ratio of Pmel-1 to Treg in the tumor.Conclusions: hetIL15 administration improves the outcome of ACT in lymphoreplete hosts, a finding with significant implications for improving cell-based cancer immunotherapy strategies. Clin Cancer Res; 23(11); 2817-30. ©2016 AACR.
Collapse
Affiliation(s)
- Sinnie Sin Man Ng
- Human Retrovirus Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, Maryland
| | - Bethany A Nagy
- Human Retrovirus Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, Maryland.,Human Retrovirus Pathogenesis Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, Maryland
| | - Shawn M Jensen
- Robert W. Franz Cancer Research Center, Earle A. Chiles Research Institute, Providence Cancer Center, Providence Portland Medical Center, Portland, Oregon
| | - Xintao Hu
- Human Retrovirus Pathogenesis Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, Maryland
| | - Candido Alicea
- Human Retrovirus Pathogenesis Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, Maryland
| | - Bernard A Fox
- Robert W. Franz Cancer Research Center, Earle A. Chiles Research Institute, Providence Cancer Center, Providence Portland Medical Center, Portland, Oregon
| | - Barbara K Felber
- Human Retrovirus Pathogenesis Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, Maryland
| | - Cristina Bergamaschi
- Human Retrovirus Pathogenesis Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, Maryland.
| | - George N Pavlakis
- Human Retrovirus Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, Maryland.
| |
Collapse
|
43
|
Pérez-Regidor L, Zarioh M, Ortega L, Martín-Santamaría S. Virtual Screening Approaches towards the Discovery of Toll-Like Receptor Modulators. Int J Mol Sci 2016; 17:ijms17091508. [PMID: 27618029 PMCID: PMC5037785 DOI: 10.3390/ijms17091508] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Revised: 07/01/2016] [Accepted: 08/22/2016] [Indexed: 02/07/2023] Open
Abstract
This review aims to summarize the latest efforts performed in the search for novel chemical entities such as Toll-like receptor (TLR) modulators by means of virtual screening techniques. This is an emergent research field with only very recent (and successful) contributions. Identification of drug-like molecules with potential therapeutic applications for the treatment of a variety of TLR-regulated diseases has attracted considerable interest due to the clinical potential. Additionally, the virtual screening databases and computational tools employed have been overviewed in a descriptive way, widening the scope for researchers interested in the field.
Collapse
Affiliation(s)
- Lucía Pérez-Regidor
- Department of Chemical & Physical Biology, Centro de Investigaciones Biológicas, CIB-CSIC, C/Ramiro de Maeztu, 9, 28040 Madrid, Spain.
| | - Malik Zarioh
- Department of Chemical & Physical Biology, Centro de Investigaciones Biológicas, CIB-CSIC, C/Ramiro de Maeztu, 9, 28040 Madrid, Spain.
| | - Laura Ortega
- Department of Chemical & Physical Biology, Centro de Investigaciones Biológicas, CIB-CSIC, C/Ramiro de Maeztu, 9, 28040 Madrid, Spain.
| | - Sonsoles Martín-Santamaría
- Department of Chemical & Physical Biology, Centro de Investigaciones Biológicas, CIB-CSIC, C/Ramiro de Maeztu, 9, 28040 Madrid, Spain.
| |
Collapse
|
44
|
The Microbiota Determines Susceptibility to Experimental Autoimmune Uveoretinitis. J Immunol Res 2016; 2016:5065703. [PMID: 27294159 PMCID: PMC4886056 DOI: 10.1155/2016/5065703] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Revised: 03/08/2016] [Accepted: 04/11/2016] [Indexed: 02/07/2023] Open
Abstract
The microbiota is a crucial modulator of the immune system. Here, we evaluated how its absence or reduction modifies the inflammatory response in the murine model of experimental autoimmune uveoretinitis (EAU). We induced EAU in germ-free (GF) or conventionally housed (CV) mice and in CV mice treated with a combination of broad-spectrum antibiotics either from the day of EAU induction or from one week prior to induction of disease. The severity of the inflammation was assessed by fundus biomicroscopy or by histology, including immunohistology. The immunophenotyping of T cells in local and distant lymph nodes was performed by flow cytometry. We found that GF mice and mice where the microbiota was reduced one week before EAU induction were protected from severe autoimmune inflammation. GF mice had lower numbers of infiltrating macrophages and significantly less T cell infiltration in the retina than CV mice with EAU. GF mice also had reduced numbers of IFN-γ and IL-17-producing T cells and increased numbers of regulatory T cells in the eye-draining lymph nodes. These data suggest that the presence of microbiota during autoantigen recognition regulates the inflammatory response by influencing the adaptive immune response.
Collapse
|
45
|
Nelson MH, Bowers JS, Bailey SR, Diven MA, Fugle CW, Kaiser ADM, Wrzesinski C, Liu B, Restifo NP, Paulos CM. Toll-like receptor agonist therapy can profoundly augment the antitumor activity of adoptively transferred CD8(+) T cells without host preconditioning. J Immunother Cancer 2016; 4:6. [PMID: 26885368 PMCID: PMC4754841 DOI: 10.1186/s40425-016-0110-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Accepted: 01/19/2016] [Indexed: 12/16/2022] Open
Abstract
Background Lymphodepletion enhances adoptive T cell transfer (ACT) therapy by activating the innate immune system via microbes released from the radiation-injured gut. Microbial components, such as LPS, are key mediators of total body irradiation (TBI) enhancement, but our ability to strategically use these toll-like receptor (TLR) agonists to bolster the potency of T cell-based therapies for cancer remains elusive. Herein, we used TLR4 agonist LPS as a tool to address how and when to use TLR agonists to effectively improve cancer immunotherapy. Methods To determine the mechanisms of how innate immune activation via lymphodepletion potentiated antitumor T cell immunity, we utilized the pmel-1 melanoma mouse model. B16F10-bearing mice were preconditioned with 5Gy TBI and given a tripartite ACT therapy (consisting of transferred pmel-1 CD8+ T cells, vaccination with fowlpox encoding gp100, and IL-2) along with TLR4 agonist LPS. The timing of LPS administration and the requirement of individual components of the tripartite therapy were evaluated based on tumor growth and the phenotype of recovered splenocytes by flow cytometry. We also evaluated the role of non-toxic and clinically used TLR4 and TLR9 agonists—monophosphoryl lipid A (MPL) and CpG Oligodeoxynucleotide (CpG ODN), respectively— for ACT therapy. Results Here we report that while exogenous administration of LPS was able to enhance adoptively transferred CD8+ T cells’ tumor destruction, LPS treatment alone did not replace individual components of the tripartite ACT regimen, or obviate TBI. Moreover, we found that sequentially administering LPS during or one day prior to ACT therapy compromised tumor regression. In contrast, administering LPS after ACT potentiated the antitumor effectiveness of the regimen, thereby supporting the expansion of transferred tumor-specific CD8+ T cells over host CD4+ T cells. We also found that non-toxic TLR agonists MPL and CpG potentiated the antitumor activity of infused CD8+ T cells. Finally, TBI was no longer needed to regress tumors in mice who were depleted of host CD4+ T cells, given a tripartite ACT regimen and then treated with low dose LPS. Conclusions Collectively, our results identify how and when to administer TLR agonists to augment T cell-based immunotherapy in the absence or presence of host preconditioning for treatment of advanced malignancies. Our findings have clinical implications for the design of next generation immune-based therapies for patients with cancer. Electronic supplementary material The online version of this article (doi:10.1186/s40425-016-0110-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Michelle H Nelson
- Microbiology and Immunology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425 USA
| | - Jacob S Bowers
- Microbiology and Immunology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425 USA
| | - Stefanie R Bailey
- Microbiology and Immunology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425 USA
| | - Marshall A Diven
- Microbiology and Immunology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425 USA
| | - Caroline W Fugle
- Microbiology and Immunology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425 USA
| | - Andrew D M Kaiser
- Center for Cancer Research, National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD 20892 USA
| | - Claudia Wrzesinski
- Center for Cancer Research, National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD 20892 USA
| | - Bei Liu
- Microbiology and Immunology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425 USA
| | - Nicholas P Restifo
- Center for Cancer Research, National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD 20892 USA
| | - Chrystal M Paulos
- Microbiology and Immunology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425 USA
| |
Collapse
|
46
|
Exploiting IL-17-producing CD4+ and CD8+ T cells to improve cancer immunotherapy in the clinic. Cancer Immunol Immunother 2016; 65:247-59. [PMID: 26825102 DOI: 10.1007/s00262-016-1797-6] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Accepted: 01/12/2016] [Indexed: 02/08/2023]
Abstract
Cancer immunotherapy is one the most effective approaches for treating patients with tumors, as it bolsters the generation and persistence of memory T cells. In preclinical work, it has been reported that adoptively transferred CD4+ and CD8+ lymphocytes that secrete IL-17A (i.e., Th17 and Tc17 cells) regress tumors to a greater extent than IFN-γ(+)Th1 or Tc1 cells in vivo. Herein, we review the mechanisms underlying how infused Th17 and Tc17 cells regress established malignancies in clinically relevant mouse models of cancer. We also discuss how unique signaling cues--such as co-stimulatory molecules (ICOS and 41BB), cytokines (IL-12 and IL-23) or pharmaceutical reagents (Akt inhibitors, etc.)--can be exploited to bolster the therapeutic potential of IL-17(+) lymphocytes with an emphasis on using this knowledge to improve next-generation clinical trials for patients with cancer.
Collapse
|
47
|
Abstract
Cancer is one of the most dreaded diseases in humans and most common cause of death in twenty-first century. New cancer therapies are urgently required because of the existing pharmacological side effects of the conventional chemotherapy, radiation, or surgery. Newer modalities such as cancer vaccines and biological therapies are proving very helpful in the treatment of cancer along with the conventional therapies. The success of these novel cancer therapies is attributed to their lesser toxicity and the specific killing of the cancer cells. Bacterial therapy for cancer has been recognized a century ago. Live, attenuated, or genetically modified obligate or facultative anaerobic bacterial species exhibit the inherent property of colonizing the tumors and are capable of multiplying selectively inside the tumors, thereby inhibiting cancerous growths. The bacteria and their spores are used in the target specific therapies, delivering the prodrugs and the various proteins to the tumors. Albeit bacterial treatment of cancer is providing new perspective in the treatment of disease, the use of microorganisms to target tumors has certain confinements. The biosafety, genetic instability and the confounded interaction of the bacteria with treatment drugs, requires the more noteworthy consideration regarding the use of this novel treatment in the cancer treatment.
Collapse
Affiliation(s)
- Pooja Sarotra
- Department of Pharmacology, Post Graduate Institute of Medical Education and Research, Chandigarh, 160012, India
| | - Bikash Medhi
- Department of Pharmacology, Post Graduate Institute of Medical Education and Research, Chandigarh, 160012, India.
| |
Collapse
|