51
|
Gopalakrishnan V, Helmink BA, Spencer CN, Reuben A, Wargo JA. The Influence of the Gut Microbiome on Cancer, Immunity, and Cancer Immunotherapy. Cancer Cell 2018; 33:570-580. [PMID: 29634945 PMCID: PMC6529202 DOI: 10.1016/j.ccell.2018.03.015] [Citation(s) in RCA: 826] [Impact Index Per Article: 137.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2017] [Revised: 03/01/2018] [Accepted: 03/13/2018] [Indexed: 02/07/2023]
Abstract
The microbiome is receiving significant attention given its influence on a host of human diseases including cancer. Its role in response to cancer treatment is becoming increasingly apparent, with evidence suggesting that modulating the gut microbiome may affect responses to numerous forms of cancer therapy. A working knowledge of the microbiome is vital as we move forward in this age of precision medicine, and an understanding of the microbiome's influence on immune responses and cancer is key. It is also important to understand factors influencing the gut microbiome and strategies to manipulate the microbiome to augment therapeutic responses.
Collapse
Affiliation(s)
- Vancheswaran Gopalakrishnan
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, 1400 Pressler Street, Unit Number 1484, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Beth A Helmink
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, 1400 Pressler Street, Unit Number 1484, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Christine N Spencer
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Unit 1954, 1881 East Road, Houston, Texas 77054, USA
| | - Alexandre Reuben
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, 1400 Pressler Street, Unit Number 1484, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Jennifer A Wargo
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, 1400 Pressler Street, Unit Number 1484, 1515 Holcombe Boulevard, Houston, TX 77030, USA; Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Unit 1954, 1881 East Road, Houston, Texas 77054, USA.
| |
Collapse
|
52
|
Peric Z, Botti S, Stringer J, Krawczyk J, van der Werf S, van Biezen A, Aljurf M, Murray J, Liptrott S, Greenfield DM, Duarte RF, Ruutu T, Basak GW. Variability of nutritional practices in peritransplant period after allogeneic hematopoietic stem cell transplantation: a survey by the Complications and Quality of Life Working Party of the EBMT. Bone Marrow Transplant 2018. [PMID: 29515252 DOI: 10.1038/s41409-018-0137-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Recommendations on screening and nutritional support for patients undergoing hematopoietic stem cell transplantation (HSCT) have been presented by international nutritional societies, but nutritional practices remain poorly standardized. Following the general policy of the European Society for Blood and Marrow Transplantation (EBMT) to standardize transplantation procedures, the Complications and Quality of Life Working Party and Nursing Research Group carried out a survey among all EBMT centers about their current nutritional practices. The aim of this study was to better understand current practices, differences from available guidelines, and possible barriers for recommended nutritional therapy. Responses from 90 centers (19%) from 23 countries were received. We observed a marked variability in nutritional care between EBMT centers and a substantial lack of standardized operating procedures in screening patients for malnutrition and management of gastrointestinal GVHD. Furthermore, our study confirmed neutropenic diet as standard of care in most centers as well a preference for parenteral nutritional support over enteral. On the basis of these findings, future EBMT efforts will focus on better implementation of international nutritional guidelines into clinical practice.
Collapse
Affiliation(s)
- Zinaida Peric
- Department of Internal Medicine, School of Medicine, University of Zagreb, Zagreb, Croatia.
| | - Stefano Botti
- Hematology Unit, Oncology Department, Azienda USL-IRCCS Reggio Emilia, Reggio Emilia, Italy
| | - Jacqui Stringer
- Hematology and Transplant Unit, Christie Hospital NHS Foundation Trust, Manchester, UK
| | - Joanna Krawczyk
- Department of Hematology, Oncology and Internal Medicine, Medical University of Warsaw, Warsaw, Poland
| | | | | | - Mahmoud Aljurf
- King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - John Murray
- Hematology and Transplant Unit, Christie Hospital NHS Foundation Trust, Manchester, UK
| | | | | | - Rafael F Duarte
- Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain
| | - Tapani Ruutu
- Clinical Research Institute, Helsinki University Hospital, Helsinki, Finland
| | - Grzegorz W Basak
- Department of Hematology, Oncology and Internal Medicine, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
53
|
Andermann TM, Peled JU, Ho C, Reddy P, Riches M, Storb R, Teshima T, van den Brink MRM, Alousi A, Balderman S, Chiusolo P, Clark WB, Holler E, Howard A, Kean LS, Koh AY, McCarthy PL, McCarty JM, Mohty M, Nakamura R, Rezvani K, Segal BH, Shaw BE, Shpall EJ, Sung AD, Weber D, Whangbo J, Wingard JR, Wood WA, Perales MA, Jenq RR, Bhatt AS. The Microbiome and Hematopoietic Cell Transplantation: Past, Present, and Future. Biol Blood Marrow Transplant 2018; 24:1322-1340. [PMID: 29471034 DOI: 10.1016/j.bbmt.2018.02.009] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 02/08/2018] [Indexed: 01/07/2023]
Affiliation(s)
- Tessa M Andermann
- Division of Infectious Diseases, Department of Medicine, Stanford University, Stanford, California
| | - Jonathan U Peled
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Christine Ho
- Blood and Marrow Transplantation, Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Pavan Reddy
- Department of Medicine, University of Michigan Cancer Center, Ann Arbor, Michigan
| | - Marcie Riches
- Division of Hematology/Oncology, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Rainer Storb
- Fred Hutchinson Cancer Research Center, Seattle, Washington; Department of Medicine, University of Washington School of Medicine, Seattle, Washington
| | - Takanori Teshima
- Department of Hematology, Hokkaido University Faculty of Medicine, Sapporo, Japan
| | - Marcel R M van den Brink
- Immunology Program, Sloan Kettering Institute, New York, New York; Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Amin Alousi
- Multidiscipline GVHD Clinic and Research Program, Department of Stem Cell Transplant and Cellular Therapies, University of Texas, MD Anderson Cancer Center, Houston, Texas
| | - Sophia Balderman
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Patrizia Chiusolo
- Hematology Department, Fondazione Policlinico Universitario A. Gemelli, Università Cattolica Sacro Cuore, Rome, Italy
| | - William B Clark
- Bone Marrow Transplant Program, Division of Hematology/Oncology and Palliative Care, Department of Internal Medicine, Virginia Commonwealth University, Richmond, Virginia
| | - Ernst Holler
- Department of Internal Medicine 3, University Medical Center, Regensburg, Germany
| | - Alan Howard
- Center for International Blood and Marrow Transplant Research, Minneapolis, Minnesota
| | - Leslie S Kean
- Fred Hutchinson Cancer Research Center, Seattle, Washington; Department of Pediatrics, University of Washington School of Medicine, Seattle, Washington; Ben Towne Center for Childhood Cancer Research, Seattle Children's Research Institute, Seattle, Washington
| | - Andrew Y Koh
- Divisions of Hematology/Oncology and Infectious Diseases, Departments of Pediatrics and Microbiology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Philip L McCarthy
- Blood and Marrow Transplantation, Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - John M McCarty
- Bone Marrow Transplantation Program, Virginia Commonwealth University Massey Cancer, Richmond, Virginia
| | - Mohamad Mohty
- Clinical Hematology and Cellular Therapy Department, Hôpital Saint-Antoine, AP-HP, Paris, France; Sorbonne Université, Paris, France; INSERM UMRs U938, Paris, France
| | - Ryotaro Nakamura
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, California
| | - Katy Rezvani
- Section of Cellular Therapy, Good Manufacturing Practices Facility, Department of Stem Cell Transplant and Cellular Therapy, University of Texas MD Anderson Cancer Center, Houston, Texas; Department of Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Brahm H Segal
- Department of Medicine, University at Buffalo Jacobs School of Medicine and Biomedical Sciences, Buffalo, New York; Division of Infectious Diseases, Roswell Park Comprehensive Cancer Center, Buffalo, New York; Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Bronwen E Shaw
- Center for International Blood and Bone Marrow Transplant Research, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Elizabeth J Shpall
- Cell Therapy Laboratory and Cord Blood Bank, Department of Stem Cell Transplantation and Cellular Therapy, University of Texas, MD Anderson Cancer Center, Houston, Texas
| | - Anthony D Sung
- Division of Hematologic Malignancies and Cellular Therapy, Duke University School of Medicine, Duke Cancer Institute, Durham, North Carolina
| | - Daniela Weber
- Department of Internal Medicine 3, University Medical Center, Regensburg, Germany
| | - Jennifer Whangbo
- Dana-Farber Cancer Institute, Boston Children's Hospital, Boston, Massachusetts
| | - John R Wingard
- Department of Medicine, University of Florida Health Cancer Center, Gainesville, Florida; Bone Marrow Transplant Program, Division of Hematology/Oncology, University of Florida College of Medicine, Florida
| | - William A Wood
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Miguel-Angel Perales
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Robert R Jenq
- Departments of Genomic Medicine and Stem Cell Transplantation Cellular Therapy, Division of Cancer Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Ami S Bhatt
- Department of Genetics and Division of Hematology, Department of Medicine, Stanford University, Stanford, California.
| | | |
Collapse
|
54
|
Bonvalet M, Daillère R, Roberti MP, Rauber C, Zitvogel L. The Impact of the Intestinal Microbiota in Therapeutic Responses Against Cancer. Oncoimmunology 2018. [DOI: 10.1007/978-3-319-62431-0_27] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
|
55
|
Shallis RM, Terry CM, Lim SH. Changes in intestinal microbiota and their effects on allogeneic stem cell transplantation. Am J Hematol 2018; 93:122-128. [PMID: 28842931 DOI: 10.1002/ajh.24896] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Revised: 07/31/2017] [Accepted: 08/22/2017] [Indexed: 12/30/2022]
Abstract
The human intestinal microbiota is essential for microbial homeostasis, regulation of metabolism, and intestinal immune tolerance. Rapidly evolving understanding of the importance of the microbiota implicates changes in the composition and function of intestinal microbial communities in an assortment of systemic conditions. Complications following allogeneic stem cell transplant now join the ever-expanding list of pathologic states regulated by intestinal microbiota. Dysbiosis, or disruption of the normal ecology of this microbiome, has been directly implicated in the pathogenesis of entities such as Clostridium difficile infections, graft-versus-host disease (GVHD), and most recently disease relapse, all of which are major causes of morbidity and mortality in patients undergoing allogeneic stem cell transplant. In this review, we elucidate the key origins of microbiotic alterations and discuss how dysbiosis influences complications following allogeneic stem cell transplant. Our emerging understanding of the importance of a balanced and diverse intestinal microbiota is prompting investigation into the appropriate treatment of dysbiosis, reliable and early detection of such, and ultimately its prevention in patients to improve the outcome following allogeneic hematopoietic stem cell transplant.
Collapse
Affiliation(s)
- Rory M. Shallis
- Division of Hematology and Oncology; Rhode Island Hospital/Brown University Warren Alpert School of Medicine; Providence Rhode Island
| | - Christopher M. Terry
- Division of Hematology and Oncology; Rhode Island Hospital/Brown University Warren Alpert School of Medicine; Providence Rhode Island
| | - Seah H. Lim
- Division of Hematology and Oncology; Rhode Island Hospital/Brown University Warren Alpert School of Medicine; Providence Rhode Island
| |
Collapse
|
56
|
Sadowska-Klasa A, Piekarska A, Prejzner W, Bieniaszewska M, Hellmann A. Colonization with multidrug-resistant bacteria increases the risk of complications and a fatal outcome after allogeneic hematopoietic cell transplantation. Ann Hematol 2017; 97:509-517. [PMID: 29255911 PMCID: PMC5797223 DOI: 10.1007/s00277-017-3205-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 12/07/2017] [Indexed: 02/07/2023]
Abstract
Composition of the gut microbiota seems to influence early complications of allogeneic hematopoietic cell transplantation (HCT) such as bacterial infections and acute graft-versus-host disease (GVHD). In this study, we assessed the impact of colonization with multidrug-resistant bacteria (MDRB) prior to HCT and the use of antibiotics against anaerobic bacteria on the outcomes of HCT. We retrospectively analyzed the data of 120 patients who underwent HCT for hematologic disorders between 2012 and 2014. Fifty-one (42.5%) patients were colonized with MDRB and 39 (32.5%) had infections caused by MDRB. Prior colonization was significantly correlated with MDRB infections (P < 0.001), especially bacteremia (P = 0.038). A higher incidence of MDRB infections was observed in patients with acute (P = 0.014) or chronic (P = 0.002) GVHD and in patients aged > 40 years (P = 0.002). Colonization had a negative impact on overall survival (OS) after HCT (64 vs. 47% at 24 months; P = 0.034) and infection-associated mortality (P < 0.001). Use of metronidazole was correlated with an increased incidence of acute GVHD (P < 0.001) and lower OS (P = 0.002). Patients colonized with MDRB are more susceptible to life-threatening infections. Colonization with virulent flora is the most probable source of neutropenic infection; therefore, information about prior positive colonization should be crucial for the selection of empiric antibiotic therapy. The use of metronidazole, affecting the biodiversity of the intestinal microbiome, seems to have a significant impact on OS and acute GVHD.
Collapse
Affiliation(s)
- Alicja Sadowska-Klasa
- Department of Hematology and Transplantology, University Clinical Center, Medical University of Gdansk, Dębinki 7, 80-952, Gdańsk, Poland
| | - Agnieszka Piekarska
- Department of Hematology and Transplantology, University Clinical Center, Medical University of Gdansk, Dębinki 7, 80-952, Gdańsk, Poland.
| | - Witold Prejzner
- Department of Hematology and Transplantology, University Clinical Center, Medical University of Gdansk, Dębinki 7, 80-952, Gdańsk, Poland
| | - Maria Bieniaszewska
- Department of Hematology and Transplantology, University Clinical Center, Medical University of Gdansk, Dębinki 7, 80-952, Gdańsk, Poland
| | - Andrzej Hellmann
- Department of Hematology and Transplantology, University Clinical Center, Medical University of Gdansk, Dębinki 7, 80-952, Gdańsk, Poland
| |
Collapse
|
57
|
Abstract
The term "microbiome" refers to microorganisms (microbiota) and their genomes (metagenome) coexisting with their hosts. Some researchers coined the term "second genome" to underscore the importance of the microbiota and its collective metagenome on their host's health and/or disease. It is now undeniable that the commensal fungal microorganisms, alongside the other components of the microbiota, play a central role in association with the human host. In recognition, projects were launched nationally and internationally to unify efforts to characterize the microbiome and elucidate the functional role of the microbiota and the mechanism(s) by which these organisms and their metabolites (metabolome) may affect health and disease states. In this article, we will highlight the role of the fungal community as an indispensable component of the microbiome.
Collapse
|
58
|
Hayase E, Hashimoto D, Nakamura K, Noizat C, Ogasawara R, Takahashi S, Ohigashi H, Yokoi Y, Sugimoto R, Matsuoka S, Ara T, Yokoyama E, Yamakawa T, Ebata K, Kondo T, Hiramine R, Aizawa T, Ogura Y, Hayashi T, Mori H, Kurokawa K, Tomizuka K, Ayabe T, Teshima T. R-Spondin1 expands Paneth cells and prevents dysbiosis induced by graft-versus-host disease. J Exp Med 2017; 214:3507-3518. [PMID: 29066578 PMCID: PMC5716036 DOI: 10.1084/jem.20170418] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2017] [Revised: 07/09/2017] [Accepted: 09/07/2017] [Indexed: 12/18/2022] Open
Abstract
The intestinal microbial ecosystem is actively regulated by Paneth cell-derived antimicrobial peptides such as α-defensins. Various disorders, including graft-versus-host disease (GVHD), disrupt Paneth cell functions, resulting in unfavorably altered intestinal microbiota (dysbiosis), which further accelerates the underlying diseases. Current strategies to restore the gut ecosystem are bacteriotherapy such as fecal microbiota transplantation and probiotics, and no physiological approach has been developed so far. In this study, we demonstrate a novel approach to restore gut microbial ecology by Wnt agonist R-Spondin1 (R-Spo1) or recombinant α-defensin in mice. R-Spo1 stimulates intestinal stem cells to differentiate to Paneth cells and enhances luminal secretion of α-defensins. Administration of R-Spo1 or recombinant α-defensin prevents GVHD-mediated dysbiosis, thus representing a novel and physiological approach at modifying the gut ecosystem to restore intestinal homeostasis and host-microbiota cross talk toward therapeutic benefits.
Collapse
Affiliation(s)
- Eiko Hayase
- Department of Hematology, Faculty of Medicine, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Daigo Hashimoto
- Department of Hematology, Faculty of Medicine, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Kiminori Nakamura
- Department of Cell Biological Science, Graduate School of Life Science, Faculty of Advanced Life Science, Hokkaido University, Sapporo, Japan
| | - Clara Noizat
- Department of Hematology, Faculty of Medicine, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Reiki Ogasawara
- Department of Hematology, Faculty of Medicine, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Shuichiro Takahashi
- Department of Hematology, Faculty of Medicine, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Hiroyuki Ohigashi
- Department of Hematology, Faculty of Medicine, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Yuki Yokoi
- Innate Immunity Laboratory, Graduate School of Life Science, Hokkaido University, Sapporo, Japan
| | - Rina Sugimoto
- Innate Immunity Laboratory, Graduate School of Life Science, Hokkaido University, Sapporo, Japan
| | - Satomi Matsuoka
- Department of Hematology, Faculty of Medicine, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Takahide Ara
- Department of Hematology, Faculty of Medicine, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Emi Yokoyama
- Department of Hematology, Faculty of Medicine, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Tomohiro Yamakawa
- Department of Hematology, Faculty of Medicine, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Ko Ebata
- Department of Hematology, Faculty of Medicine, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Takeshi Kondo
- Department of Hematology, Faculty of Medicine, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Rina Hiramine
- Department of Protein Science Laboratory, Graduate School of Life Science, Hokkaido University, Sapporo, Japan.,Global Station for Soft Matter, Global Institution for Collaborative Research and Education, Hokkaido University, Sapporo, Japan
| | - Tomoyasu Aizawa
- Department of Protein Science Laboratory, Graduate School of Life Science, Hokkaido University, Sapporo, Japan.,Global Station for Soft Matter, Global Institution for Collaborative Research and Education, Hokkaido University, Sapporo, Japan
| | - Yoshitoshi Ogura
- Department of Bacteriology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Tetsuya Hayashi
- Department of Bacteriology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Hiroshi Mori
- Center for Information Biology, National Institute of Genetics, Mishima, Japan
| | - Ken Kurokawa
- Center for Information Biology, National Institute of Genetics, Mishima, Japan.,Department of Life Science and Technology, Tokyo Institute of Technology, Tokyo, Japan
| | - Kazuma Tomizuka
- Innovative Technology Labs, Research Functions Unit, Research & Development Division, Kyowa Hakko Kirin, Tokyo, Japan
| | - Tokiyoshi Ayabe
- Department of Cell Biological Science, Graduate School of Life Science, Faculty of Advanced Life Science, Hokkaido University, Sapporo, Japan
| | - Takanori Teshima
- Department of Hematology, Faculty of Medicine, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| |
Collapse
|
59
|
Frenkel M, Sapire K. Complementary and Integrative Medicine in Hematologic Malignancies: Questions and Challenges. Curr Oncol Rep 2017; 19:79. [PMID: 29032389 DOI: 10.1007/s11912-017-0635-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Hematologic malignancies represent 9.7% of all cancers, making them the fourth most common type of cancer in the United States. The aggressive and complex treatments administered in hematologic malignancies result in a high burden of psychological needs. Complementary and integrative medicine (CIM) is becoming one of the options that patients use to address their distress during and after cancer treatments. It is not clear whether appropriate CIM can relieve distress in patients affected by these malignancies. This review covers the potential benefits of CIM as relates to nutrition, nutritional supplements, exercise, circadian rhythm, methods for reducing distress during bone marrow aspiration, massage therapy, and acupuncture, in treating patients with hematological malignancies. This review may provide a framework to enhance patient-doctor dialogue regarding CIM use in hematologic malignancies.
Collapse
Affiliation(s)
- Moshe Frenkel
- Department of Family Medicine, The University of Texas Medical Branch at Galveston, Galveston, TX, USA. .,Integrative Medicine Program, Institute of Oncology, Meir Medical Center, Kfar Saba, Israel. .,, Hashoftim 1 B, 30900, Zichron Yaacov, Israel.
| | - Kenneth Sapire
- Department of Anesthesia and Perioperative Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
60
|
Associations between acute gastrointestinal GvHD and the baseline gut microbiota of allogeneic hematopoietic stem cell transplant recipients and donors. Bone Marrow Transplant 2017; 52:1643-1650. [PMID: 28967895 DOI: 10.1038/bmt.2017.200] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2017] [Revised: 07/28/2017] [Accepted: 08/04/2017] [Indexed: 12/13/2022]
Abstract
Growing evidence suggests that host-microbiota interactions influence GvHD risk following allogeneic hematopoietic stem cell transplant. However, little is known about the influence of the transplant recipient's pre-conditioning microbiota nor the influence of the transplant donor's microbiota. Our study examines associations between acute gastrointestinal GvHD (agGvHD) and 16S rRNA fecal bacterial profiles in a prospective cohort of N=57 recipients before preparative conditioning, as well as N=22 of their paired HLA-matched sibling donors. On average, recipients had lower fecal bacterial diversity (P=0.0002) and different phylogenetic membership (UniFrac P=0.001) than the healthy transplant donors. Recipients with lower phylogenetic diversity had higher overall mortality rates (hazard ratio=0.37, P=0.008), but no statistically significant difference in agGvHD risk. In contrast, high bacterial donor diversity was associated with decreased agGvHD risk (odds ratio=0.12, P=0.038). Further investigation is warranted as to whether selection of hematopoietic stem cell transplant donors with high gut microbiota diversity and/or other specific compositional attributes may reduce agGvHD incidence, and by what mechanisms.
Collapse
|
61
|
Role of the intestinal mucosa in acute gastrointestinal GVHD. Blood 2017; 128:2395-2402. [PMID: 27856471 DOI: 10.1182/blood-2016-06-716738] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Accepted: 09/14/2016] [Indexed: 12/11/2022] Open
Abstract
Intestinal graft-versus-host disease (GVHD) remains a significant obstacle to the success of allogeneic hematopoietic cell transplantation. The intestinal mucosa comprises the inner lining of the intestinal tract and maintains close proximity with commensal microbes that reside within the intestinal lumen. Recent advances have significantly improved our understanding of the interactions between the intestinal mucosa and the enteric microbiota. Changes in host mucosal tissue and commensals posttransplant have been actively investigated, and provocative insights into mucosal immunity and the enteric microbiota are now being translated into clinical trials of novel approaches for preventing and treating acute GVHD. In this review, we summarize recent findings related to aspects of the intestinal mucosa during acute GVHD.
Collapse
|
62
|
Ferrara JL, Smith CM, Sheets J, Reddy P, Serody JS. Altered homeostatic regulation of innate and adaptive immunity in lower gastrointestinal tract GVHD pathogenesis. J Clin Invest 2017; 127:2441-2451. [PMID: 28581444 DOI: 10.1172/jci90592] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Lower gastrointestinal (GI) tract graft-versus-host disease (GVHD) is the predominant cause of morbidity and mortality from GVHD after allogeneic stem cell transplantation. Recent data indicate that lower GI tract GVHD is a complicated process mediated by donor/host antigenic disparities. This process is exacerbated by significant changes to the microbiome, and innate and adaptive immune responses that are critical to the induction of disease, persistence of inflammation, and a lack of response to therapy. Here, we discuss new insights into the biology of lower GI tract GVHD and focus on intrinsic pathways and regulatory mechanisms crucial to normal intestinal function. We then describe multiple instances in which these homeostatic mechanisms are altered by donor T cells or conditioning therapy, resulting in exacerbation of GVHD. We also discuss data suggesting that some of these mechanisms produce biomarkers that could be informative as to the severity of GVHD and its response to therapy. Finally, novel therapies that might restore homeostasis in the GI tract during GVHD are highlighted.
Collapse
Affiliation(s)
- James Lm Ferrara
- Departments of Medicine, Pediatrics, and Academic Informatics and Technology, Icahn School of Medicine at Mount Sinai and Tisch Cancer Institute, New York, New York, USA
| | - Christopher M Smith
- Departments of Medicine, Pediatrics, and Academic Informatics and Technology, Icahn School of Medicine at Mount Sinai and Tisch Cancer Institute, New York, New York, USA
| | - Julia Sheets
- University of North Carolina Hospital, Chapel Hill, North Carolina, USA
| | - Pavan Reddy
- Department of Medicine and University of Michigan Comprehensive Cancer Center, University of Michigan, Ann Arbor, Michigan, USA
| | - Jonathan S Serody
- Department of Medicine and UNC Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina, USA
| |
Collapse
|
63
|
Yoshioka K, Kakihana K, Doki N, Ohashi K. Gut microbiota and acute graft-versus-host disease. Pharmacol Res 2017; 122:90-95. [PMID: 28576475 DOI: 10.1016/j.phrs.2017.05.028] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Revised: 04/19/2017] [Accepted: 05/29/2017] [Indexed: 12/12/2022]
Abstract
Although allogeneic stem cell transplantation (allo-SCT) is a potentially curative treatment for various hematological diseases, acute graft-versus-host disease (GVHD) is a major cause of morbidity and mortality, and its management is clinically important. Advances in biological techniques have led to great progress in understanding the complex interactions between the host and the gut microbiota. The gut microbiota clearly modulates the immune response and is associated with the pathogenesis of various disorders. Also in allo-SCT, both preclinical and clinical results indicate that the gut microbiota is closely associated with the development of acute GVHD and transplant outcomes. These results led to the idea that improvement in quantitative and/or qualitative abnormalities of microbiota (dysbiosis) may be a new treatment strategy for acute GVHD. Evaluations of therapies targeting the gut microbiota such as probiotics or fecal microbiota transplantation have just begun. Furthermore, intervention in the gut microbiota with a nutritional approach including prebiotics, postbiotics, and antibiotics selection may also be another promising treatment option for acute GVHD.
Collapse
Affiliation(s)
- Kosuke Yoshioka
- Hematology Division, Tokyo Metropolitan Cancer and Infectious Disease Center, Komagome Hospital, Japan
| | - Kazuhiko Kakihana
- Hematology Division, Tokyo Metropolitan Cancer and Infectious Disease Center, Komagome Hospital, Japan.
| | - Noriko Doki
- Hematology Division, Tokyo Metropolitan Cancer and Infectious Disease Center, Komagome Hospital, Japan
| | - Kazuteru Ohashi
- Hematology Division, Tokyo Metropolitan Cancer and Infectious Disease Center, Komagome Hospital, Japan
| |
Collapse
|
64
|
El-Jurdi N, de Lima M, Lazarus H, Ghannoum MA. Microbiome: Its Impact Is Being Revealed! CURRENT CLINICAL MICROBIOLOGY REPORTS 2017. [DOI: 10.1007/s40588-017-0063-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
65
|
Gorshein E, Wei C, Ambrosy S, Budney S, Vivas J, Shenkerman A, Manago J, McGrath MK, Tyno A, Lin Y, Patel V, Gharibo M, Schaar D, Jenq RR, Khiabanian H, Strair R. Lactobacillus rhamnosus GG probiotic enteric regimen does not appreciably alter the gut microbiome or provide protection against GVHD after allogeneic hematopoietic stem cell transplantation. Clin Transplant 2017; 31. [PMID: 28256022 DOI: 10.1111/ctr.12947] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/26/2017] [Indexed: 12/30/2022]
Abstract
Graft-versus-host disease (GVHD) is a major adverse effect associated with allogeneic stem cell transplant. Previous studies in mice indicated that administration of the probiotic Lactobacillus rhamnosus GG can reduce the incidence of GVHD after hematopoietic stem cell transplant. Here we report results from the first randomized probiotic enteric regimen trial in which allogenic hematopoietic stem cell patients were supplemented with Lactobacillus rhamnosus GG. Gut microbiome analysis confirmed a previously reported gut microbiome association with GVHD. However, the clinical trial was terminated when interim analysis did not detect an appreciable probiotic-related change in the gut microbiome or incidence of GVHD. Additional studies are necessary to determine whether probiotics can alter the incidence of GVHD after allogeneic stem cell transplant.
Collapse
Affiliation(s)
- Elan Gorshein
- Department of Medicine, Rutgers Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ, USA
| | - Catherine Wei
- Department of Medicine, Rutgers Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ, USA
| | - Susan Ambrosy
- Department of Pathology and Laboratory Medicine, Rutgers Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ, USA
| | - Shanna Budney
- Department of Pathology and Laboratory Medicine, Rutgers Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ, USA
| | - Juliana Vivas
- Department of Pathology and Laboratory Medicine, Rutgers Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ, USA
| | - Angelika Shenkerman
- Rutgers Cancer Institute of New Jersey, Rutgers University, New Brunswick, NJ, USA
| | - Jacqueline Manago
- Rutgers Cancer Institute of New Jersey, Rutgers University, New Brunswick, NJ, USA
| | - Mary Kate McGrath
- Department of Pathology and Laboratory Medicine, Rutgers Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ, USA
| | - Anne Tyno
- Rutgers Cancer Institute of New Jersey, Rutgers University, New Brunswick, NJ, USA
| | - Yong Lin
- Rutgers Cancer Institute of New Jersey, Rutgers University, New Brunswick, NJ, USA
| | - Vimal Patel
- Rutgers Cancer Institute of New Jersey, Rutgers University, New Brunswick, NJ, USA
| | - Mecide Gharibo
- Rutgers Cancer Institute of New Jersey, Rutgers University, New Brunswick, NJ, USA
| | - Dale Schaar
- Rutgers Cancer Institute of New Jersey, Rutgers University, New Brunswick, NJ, USA
| | - Robert R Jenq
- Division of Cancer Medicine, Departments of Genomic Medicine and Stem Cell Transplantation Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hossein Khiabanian
- Department of Pathology and Laboratory Medicine, Rutgers Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ, USA
| | - Roger Strair
- Rutgers Cancer Institute of New Jersey, Rutgers University, New Brunswick, NJ, USA
| |
Collapse
|
66
|
Staffas A, Burgos da Silva M, van den Brink MRM. The intestinal microbiota in allogeneic hematopoietic cell transplant and graft-versus-host disease. Blood 2017; 129:927-933. [PMID: 27940475 PMCID: PMC5324712 DOI: 10.1182/blood-2016-09-691394] [Citation(s) in RCA: 146] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Accepted: 12/05/2016] [Indexed: 12/14/2022] Open
Abstract
Hematopoietic cell transplantation (HCT) is a critical treatment of patients with high-risk hematopoietic malignancies, hematological deficiencies, and other immune diseases. In allogeneic HCT (allo-HCT), donor-derived T cells recognize host tissues as foreign, causing graft-versus-host disease (GVHD) which is a main contributor to morbidity and mortality. The intestine is one of the organs most severely affected by GVHD and research has recently highlighted the importance of bacteria, particularly the gut microbiota, in HCT outcome and in GVHD development. Loss of intestinal bacterial diversity is common during the course of HCT and is associated with GVHD development and treatment with broad-spectrum antibiotics. Loss of intestinal diversity and outgrowth of opportunistic pathogens belonging to the phylum Proteobacteria and Enterococcus genus have also been linked to increased treatment-related mortality including GVHD, infections, and organ failure after allo-HCT. Experimental studies in allo-HCT animal models have shown some promising results for prebiotic and probiotic strategies as prophylaxis or treatment of GVHD. Continuous research will be important to define the relation of cause and effect for these associations between microbiota features and HCT outcomes. Importantly, studies focused on geographic and cultural differences in intestinal microbiota are necessary to define applicability of new strategies targeting the intestinal microbiota.
Collapse
Affiliation(s)
- Anna Staffas
- Department of Immunology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Marina Burgos da Silva
- Department of Immunology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Marcel R M van den Brink
- Department of Immunology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Medicine and
- Department of Immunology, Weill Medical College of Cornell University, New York, NY; and
- Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| |
Collapse
|
67
|
Pessach I, Tsirigotis P, Nagler A. The gastrointestinal tract: properties and role in allogeneic hematopoietic stem cell transplantation. Expert Rev Hematol 2017; 10:315-326. [PMID: 28136133 DOI: 10.1080/17474086.2017.1288566] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
INTRODUCTION The GI-tract is a major target for both the intensive chemo and/or radiotherapy conditioning as well as for GVHD and therefore is closely associated with transplant outcome. Apart from being a target, the GI-tract is also a mediator and therefore is also a key player of the pathogenetic process following allogeneic transplantation. Areas covered: The intestinal homeostasis is regulated through complicated interactions between the key players of this process which are the intestinal epithelium, the intestinal immune system, and the intestinal microbiota. A brief description of these elements, based on published english-language articles in PubMed, as well as their role during the process of allo-HSCT is discussed in this review. Expert commentary: Data on GI-tract properties suggest a central role for the intestine in regulation of immunity, both in healthy - steady state conditions and in pathological states such as during allo-HSCT. Given the fact that in the allogeneic transplant setting severe complications such as infections and GVHD are limiting this treatment modality, understanding the mechanisms that mediate intestinal homeostasis could lead to new preventive methods and improved outcomes.
Collapse
Affiliation(s)
- Ilias Pessach
- a Second Department of Internal Medicine, Division of Hematology, ATTIKO University Hospital , National and Kapodistrian University of Athens , Athens , Greece
| | - Panagiotis Tsirigotis
- a Second Department of Internal Medicine, Division of Hematology, ATTIKO University Hospital , National and Kapodistrian University of Athens , Athens , Greece
| | - Arnon Nagler
- b Division of Hematology and Bone Marrow Transplantation , Chaim Sheba Medical Center , Tel Hashomer , Israel
| |
Collapse
|
68
|
Reduction of aGVHD using chicken antibodies directed against intestinal pathogens in a murine model. Blood 2016; 129:1052-1055. [PMID: 28011676 DOI: 10.1182/blood-2016-06-722538] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
|
69
|
Raghunathan VM, Sheng I, Lim SH. Intestinal dysbiosis and allogeneic hematopoietic progenitor cell transplantation. J Transl Med 2016; 14:335. [PMID: 27912759 PMCID: PMC5135767 DOI: 10.1186/s12967-016-1094-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Accepted: 11/23/2016] [Indexed: 02/08/2023] Open
Abstract
The intestinal microbiota is a diverse and dynamic ecosystem that is increasingly understood to play a vital role in human health. Hematopoietic stem cell transplant recipients undergo prolonged exposure to antimicrobials, chemotherapeutic agents, and immunosuppressants, resulting in profound shifts in the gut microbiome. A growing body of research has revealed the ways in which these microbiologic shifts shape immune modulation, affecting susceptibility to infections and graft-versus-host disease, the two major post-transplant complications in this population. As transplant medicine becomes increasingly personalized, the potential for microbiome-modulating treatments holds immense potential. Strategies to preserve the intestinal microbiota, including targeted antibiotics, prebiotics and probiotics, and fecal microbiota transplant could mitigate some of the microbiologic shifts in stem cell transplant recipients, and reduce the incidence of peri-transplant morbidity and mortality.
Collapse
Affiliation(s)
- Vikram M Raghunathan
- Division of Hematology and Oncology, Department of Medicine, Rhode Island Hospital, Room 140 APC Building, 593 Eddy Street, Providence, RI, 02903, USA.,Brown University Warren Alpert School of Medicine, Providence, USA
| | - Iris Sheng
- Division of Hematology and Oncology, Department of Medicine, Rhode Island Hospital, Room 140 APC Building, 593 Eddy Street, Providence, RI, 02903, USA.,Brown University Warren Alpert School of Medicine, Providence, USA
| | - Seah H Lim
- Division of Hematology and Oncology, Department of Medicine, Rhode Island Hospital, Room 140 APC Building, 593 Eddy Street, Providence, RI, 02903, USA. .,Brown University Warren Alpert School of Medicine, Providence, USA.
| |
Collapse
|
70
|
Peled JU, Hanash AM, Jenq RR. Role of the intestinal mucosa in acute gastrointestinal GVHD. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2016; 2016:119-127. [PMID: 27913470 PMCID: PMC5575743 DOI: 10.1182/asheducation-2016.1.119] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
Intestinal graft-versus-host disease (GVHD) remains a significant obstacle to the success of allogeneic hematopoietic cell transplantation. The intestinal mucosa comprises the inner lining of the intestinal tract and maintains close proximity with commensal microbes that reside within the intestinal lumen. Recent advances have significantly improved our understanding of the interactions between the intestinal mucosa and the enteric microbiota. Changes in host mucosal tissue and commensals posttransplant have been actively investigated, and provocative insights into mucosal immunity and the enteric microbiota are now being translated into clinical trials of novel approaches for preventing and treating acute GVHD. In this review, we summarize recent findings related to aspects of the intestinal mucosa during acute GVHD.
Collapse
Affiliation(s)
- Jonathan U Peled
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY; and
- Weill Cornell Medical College, New York, NY
| | - Alan M Hanash
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY; and
| | - Robert R Jenq
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY; and
- Weill Cornell Medical College, New York, NY
| |
Collapse
|
71
|
Zeiser R, Socié G, Blazar BR. Pathogenesis of acute graft-versus-host disease: from intestinal microbiota alterations to donor T cell activation. Br J Haematol 2016; 175:191-207. [PMID: 27619472 DOI: 10.1111/bjh.14295] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Revised: 06/30/2016] [Accepted: 06/30/2016] [Indexed: 01/03/2023]
Abstract
Acute graft-versus-host disease (aGVHD) is a major life-threatening complication of allogeneic haematopoietic cell transplantation (allo-HCT). Here we discuss the aGVHD pathophysiology initiated by multiple signals that cause alloreactive T-cell activation. The outcome of such donor T-cell activation is influenced by T-cell receptor-signal strength, anatomical location, co-stimulatory/co-inhibitory signals and differentiation stage (naive, effector/memory) of T-cells. Additionally, cross-priming of T cells to antigens expressed by pathogens can contribute to aGVHD-mediated tissue injury. In addition to the properties of donor T-cell activation, highly specialized tissue resident cell types, such as innate lymphoid cells, antigen-presenting cells, immune regulatory cells and various intestinal cell populations are critically involved in aGVHD pathogenesis. The role of the thymus and secondary lymphoid tissue injury, non-haematopoietic cells, intestinal microflora, cytokines, chemokines, microRNAs, metabolites and kinases in aGVHD pathophysiology will be highlighted. Acute GVHD pathogenic mechanisms will be connected to novel therapeutic approaches under development for, and tested in, the clinic.
Collapse
Affiliation(s)
- Robert Zeiser
- Department of Haematology, Oncology and Stem Cell Transplantation, Freiburg University Medical Centre, Freiburg, Germany.
| | - Gerard Socié
- Haematology Stem cell transplant Unit, Saint Louis Hospital, APHP, Paris, France
| | - Bruce R Blazar
- Department of Pediatrics, Division of Blood and Marrow Transplantation, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
72
|
Gut microbiota and hematopoietic stem cell transplantation: where do we stand? Bone Marrow Transplant 2016; 52:7-14. [PMID: 27348539 DOI: 10.1038/bmt.2016.173] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Revised: 05/10/2016] [Accepted: 05/13/2016] [Indexed: 12/26/2022]
Abstract
Advances in biological techniques have potentiated great progresses in understanding the interaction between human beings and the ∼10 to 100 trillion microbes living in their gastrointestinal tract: gut microbiota (GM). In this review, we describe recent emerging data on the role of GM in hematopoietic stem cell transplantation, with a focus on immunomodulatory properties in the immune system recovery and the impact in the development of the main complications, as GvHD and infections.
Collapse
|
73
|
Clavel T, Lagkouvardos I, Hiergeist A. Microbiome sequencing: challenges and opportunities for molecular medicine. Expert Rev Mol Diagn 2016; 16:795-805. [DOI: 10.1080/14737159.2016.1184574] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Thomas Clavel
- ZIEL Institute for Food and Health, Technical University of Munich, Munich, Germany
| | - Ilias Lagkouvardos
- ZIEL Institute for Food and Health, Technical University of Munich, Munich, Germany
| | - Andreas Hiergeist
- Institute of Clinical Microbiology and Hygiene, University Hospital Regensburg, Regensburg, Germany
| |
Collapse
|
74
|
The Gut Microbiota and Immune System Relationship in Human Graft-versus-Host Disease. Mediterr J Hematol Infect Dis 2016; 8:e2016025. [PMID: 27158438 PMCID: PMC4848019 DOI: 10.4084/mjhid.2016.025] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Accepted: 04/10/2016] [Indexed: 02/07/2023] Open
Abstract
Gut microbiota has gained increasing interest in the pathogenesis of immune-related diseases. In this context, graft-versus-host disease is a condition characterized by an immune response which frequently complicates and limits the outcomes of hematopoietic stem cell transplantations. Past studies, carried mostly in animals, already supported a relationship between gut microbiota and graft-versus-host disease. However, the possible mechanisms underlying this connection remain elusory. Moreover, strategies to prevent graft-versus-host disease are of great interest as well as the potential role of gut microbiota modulation. We reviewed the role of gut microbiota in the development of immune system and its involvement in the graft-versus-host disease, focusing on data available on humans.
Collapse
|
75
|
Peled JU, Jenq RR, Holler E, van den Brink MRM. Role of gut flora after bone marrow transplantation. Nat Microbiol 2016; 1:16036. [PMID: 27572448 PMCID: PMC5027134 DOI: 10.1038/nmicrobiol.2016.36] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- Jonathan U. Peled
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Robert R. Jenq
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Ernst Holler
- Department of Hematology and Oncology, Internal Medicine III, University Medical Center, Regensburg, Germany
| | - Marcel R. M. van den Brink
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
76
|
Potential protective effect of Helicobacter pylori on the development of gastrointestinal GvHD. Bone Marrow Transplant 2016; 51:819-24. [PMID: 26950379 DOI: 10.1038/bmt.2016.31] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Revised: 01/21/2016] [Accepted: 01/24/2016] [Indexed: 11/08/2022]
Abstract
Previous reports ascribe a modulating capacity of the immune response to Helicobacter pylori (HP). Our hypothesis was to demonstrate in a prospective study that HP infection could have a protective effect against development of gastrointestinal GvHD in patients receiving allogeneic hematopoietic cell transplantation (HCT). Presence of HP before transplant was determined using C(13) urea breath test. Seventy-nine patients receiving an allogeneic HCT were included and 93.7% of them received PBSC; in 51.9%, the donor was unrelated. Acute gastrointestinal GvHD was diagnosed in 51.9% (n=41). In the multivariable analysis, HP infection was associated with a lower frequency of gastrointestinal GvHD (odds ratio (OR)=0.19 (95% confidence interval (CI): 0.05-0.67); in contrast, an unrelated donor was associated with a higher frequency of gastrointestinal GvHD (odds ratio=5.4 (95% confidence interval: 1.6-18.2). One year overall survival (OS) was 74%. In the multivariate Cox proportional-hazards regression analysis, stages 0-II gastrointestinal GvHD (hazards ratio (HR)=0.19), reduced intensity conditioning (HR=0.04) and tacrolimus-sirolimus GvHD prophylaxis (HR=0.06) were all associated with a better OS. In summary, HP infection could have a role in decreasing gastrointestinal GvHD in patients receiving allogeneic HCT from peripheral blood including related and unrelated donors.
Collapse
|
77
|
Rigoni R, Fontana E, Guglielmetti S, Fosso B, D'Erchia AM, Maina V, Taverniti V, Castiello MC, Mantero S, Pacchiana G, Musio S, Pedotti R, Selmi C, Mora JR, Pesole G, Vezzoni P, Poliani PL, Grassi F, Villa A, Cassani B. Intestinal microbiota sustains inflammation and autoimmunity induced by hypomorphic RAG defects. J Exp Med 2016; 213:355-75. [PMID: 26926994 PMCID: PMC4813669 DOI: 10.1084/jem.20151116] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Accepted: 01/25/2016] [Indexed: 12/21/2022] Open
Abstract
Rigoni et al. report that hypomorphic Rag2R229Q mutation is associated with altered microbiota composition and defects in the gut–blood barrier and suggest that intestinal microbes may play a critical role in the distinctive immune dysregulation of Omenn syndrome. Omenn syndrome (OS) is caused by hypomorphic Rag mutations and characterized by a profound immunodeficiency associated with autoimmune-like manifestations. Both in humans and mice, OS is mediated by oligoclonal activated T and B cells. The role of microbial signals in disease pathogenesis is debated. Here, we show that Rag2R229Q knock-in mice developed an inflammatory bowel disease affecting both the small bowel and colon. Lymphocytes were sufficient for disease induction, as intestinal CD4 T cells with a Th1/Th17 phenotype reproduced the pathological picture when transplanted into immunocompromised hosts. Moreover, oral tolerance was impaired in Rag2R229Q mice, and transfer of wild-type (WT) regulatory T cells ameliorated bowel inflammation. Mucosal immunoglobulin A (IgA) deficiency in the gut resulted in enhanced absorption of microbial products and altered composition of commensal communities. The Rag2R229Q microbiota further contributed to the immunopathology because its transplant into WT recipients promoted Th1/Th17 immune response. Consistently, long-term dosing of broad-spectrum antibiotics (ABXs) in Rag2R229Q mice ameliorated intestinal and systemic autoimmunity by diminishing the frequency of mucosal and circulating gut-tropic CCR9+ Th1 and Th17 T cells. Remarkably, serum hyper-IgE, a hallmark of the disease, was also normalized by ABX treatment. These results indicate that intestinal microbes may play a critical role in the distinctive immune dysregulation of OS.
Collapse
Affiliation(s)
- Rosita Rigoni
- Milan Unit, Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche, 20133 Milan, Italy Humanitas Clinical and Research Center, Rozzano, 20089 Milan, Italy
| | - Elena Fontana
- Department of Molecular and Translational Medicine, Pathology Unit, University of Brescia School of Medicine, 25123 Brescia, Italy
| | - Simone Guglielmetti
- Department of Food, Environmental, and Nutritional Sciences (DeFENS), University of Milan, 20122 Milan, Italy
| | - Bruno Fosso
- Institute of Biomembranes and Bioenergetics, National Research Council, 70126 Bari, Italy
| | - Anna Maria D'Erchia
- Department of Biosciences, Biotechnology, and Pharmacological Sciences, University of Bari, 70121 Bari, Italy Institute of Biomembranes and Bioenergetics, National Research Council, 70126 Bari, Italy
| | - Virginia Maina
- Telethon Institute for Gene Therapy, Division of Regenerative Medicine, Stem Cells and Gene Therapy, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Valentina Taverniti
- Department of Food, Environmental, and Nutritional Sciences (DeFENS), University of Milan, 20122 Milan, Italy
| | - Maria Carmina Castiello
- Telethon Institute for Gene Therapy, Division of Regenerative Medicine, Stem Cells and Gene Therapy, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Stefano Mantero
- Telethon Institute for Gene Therapy, Division of Regenerative Medicine, Stem Cells and Gene Therapy, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Giovanni Pacchiana
- Milan Unit, Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche, 20133 Milan, Italy Humanitas Clinical and Research Center, Rozzano, 20089 Milan, Italy
| | - Silvia Musio
- Foundation IRCCS Neurological Institute, C. Besta, Neuroimmunology and Neuromuscular Disorders Unit, 20132 Milan, Italy
| | - Rosetta Pedotti
- Foundation IRCCS Neurological Institute, C. Besta, Neuroimmunology and Neuromuscular Disorders Unit, 20132 Milan, Italy
| | - Carlo Selmi
- Humanitas Clinical and Research Center, Rozzano, 20089 Milan, Italy BIOMETRA Department, University of Milan, 20122 Milan, Italy
| | - J Rodrigo Mora
- Gastrointestinal Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02115
| | - Graziano Pesole
- Department of Biosciences, Biotechnology, and Pharmacological Sciences, University of Bari, 70121 Bari, Italy Institute of Biomembranes and Bioenergetics, National Research Council, 70126 Bari, Italy
| | - Paolo Vezzoni
- Milan Unit, Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche, 20133 Milan, Italy Humanitas Clinical and Research Center, Rozzano, 20089 Milan, Italy
| | - Pietro Luigi Poliani
- Department of Molecular and Translational Medicine, Pathology Unit, University of Brescia School of Medicine, 25123 Brescia, Italy
| | - Fabio Grassi
- Istituto Nazionale Genetica Molecolare, Department of Medical Biotechnology and Translational Medicine, University of Milan, 20122 Milan, Italy Institute for Research in Biomedicine, 6500 Bellinzona, Switzerland
| | - Anna Villa
- Milan Unit, Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche, 20133 Milan, Italy Telethon Institute for Gene Therapy, Division of Regenerative Medicine, Stem Cells and Gene Therapy, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Barbara Cassani
- Milan Unit, Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche, 20133 Milan, Italy Humanitas Clinical and Research Center, Rozzano, 20089 Milan, Italy
| |
Collapse
|
78
|
Andermann TM, Rezvani A, Bhatt AS. Microbiota Manipulation With Prebiotics and Probiotics in Patients Undergoing Stem Cell Transplantation. Curr Hematol Malig Rep 2016; 11:19-28. [PMID: 26780719 PMCID: PMC4996265 DOI: 10.1007/s11899-016-0302-9] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Hematopoietic stem cell transplantation (HSCT) is a potentially life-saving therapy that often comes at the cost of complications such as graft-versus-host disease and post-transplant infections. With improved technology to understand the ecosystem of microorganisms (viruses, bacteria, fungi, and microeukaryotes) that make up the gut microbiota, there is increasing evidence of the microbiota's contribution to the development of post-transplant complications. Antibiotics have traditionally been the mainstay of microbiota-altering therapies available to physicians. Recently, interest is increasing in the use of prebiotics and probiotics to support the development and sustainability of a healthier microbiota. In this review, we will describe the evidence for the use of prebiotics and probiotics in combating microbiota dysbiosis and explore the ways in which they may be used in future research to potentially improve clinical outcomes and decrease rates of graft-versus-host disease (GVHD) and post-transplant infection.
Collapse
Affiliation(s)
- Tessa M Andermann
- Department of Medicine, Division of Infectious Diseases, Stanford University, Stanford, CA, USA
| | - Andrew Rezvani
- Department of Medicine, Division of Blood and Marrow Transplantation, Stanford University, Stanford, CA, USA
| | - Ami S Bhatt
- Department of Medicine, Division of Blood and Marrow Transplantation, Stanford University, Stanford, CA, USA.
- Department of Medicine, Division of Hematology, Stanford University, 269 Campus Drive, Stanford, CA, 94305, USA.
- Department of Genetics, Stanford University, Stanford, CA, USA.
| |
Collapse
|
79
|
Understanding Luminal Microorganisms and Their Potential Effectiveness in Treating Intestinal Inflammation. Inflamm Bowel Dis 2016; 22:194-201. [PMID: 26457381 PMCID: PMC4679592 DOI: 10.1097/mib.0000000000000599] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The human intestine contains 10¹⁴ bacteria, which outnumber the mammalian cells 10-fold. Certain other commensal or infectious agents, like helminthic parasites, become members of this microbial ecosystem, especially in populations living under less hygienic conditions. Intestinal microbes, also called the microbiome or microbiota, shape the host immune reactivity to self and nonself throughout life. Changes in microbiome composition may impair the maturation of immune regulatory pathways and predispose the host to develop various forms of inflammatory disorders, like Crohn's disease or ulcerative colitis. The microbiome is also critical to successful transplantation of organs or grafts. After allogeneic hematopoietic stem cell transplantation, when the new donor cells, such as T lymphocytes learn to discriminate "the new self from nonself" in the transplant recipient, they need healthy microbiota-derived signals to preserve the immune homeostasis. Restoring microbiota through intestinal delivery of bacterial strains, helminths, fecal microbiota transplantation, or stool substitutes have the potential to improve and correct aberrant immune reactivity in various disorders.
Collapse
|
80
|
McCabe L, Britton RA, Parameswaran N. Prebiotic and Probiotic Regulation of Bone Health: Role of the Intestine and its Microbiome. Curr Osteoporos Rep 2015; 13:363-71. [PMID: 26419466 PMCID: PMC4623939 DOI: 10.1007/s11914-015-0292-x] [Citation(s) in RCA: 148] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Recent advances in our understanding of how the intestinal microbiome contributes to health and disease have generated great interest in developing strategies for modulating the abundance of microbes and/or their activity to improve overall human health and prevent pathologies such as osteoporosis. Bone is an organ that the gut has long been known to regulate through absorption of calcium, the key bone mineral. However, it is clear that modulation of the gut and its microbiome can affect bone density and strength in a variety of animal models (zebrafish, rodents, chicken) and humans. This is demonstrated in studies ablating the microbiome through antibiotic treatment or using germ-free mouse conditions as well as in studies modulating the microbiome activity and composition through prebiotic and/or probiotic treatment. This review will discuss recent developments in this new and exciting area.
Collapse
Affiliation(s)
- Laura McCabe
- Department of Physiology, Biomedical Imaging Research Center, Michigan State University, Biomedical Physical Science Building, 567 Wilson Road, East Lansing, MI, 48824, USA.
- Department of Radiology, Biomedical Imaging Research Center, Michigan State University, Biomedical Physical Science Building, 846 Service Road, East Lansing, MI, 48824, USA.
| | - Robert A Britton
- Baylor College of Medicine, Department of Molecular Virology and Microbiology, Alkek Center for Metagenomics and Microbiome Research, One Baylor Plaza, Houston, TX, 77030, USA.
| | - Narayanan Parameswaran
- Department of Physiology, Biomedical Imaging Research Center, Michigan State University, Biomedical Physical Science Building, 567 Wilson Road, East Lansing, MI, 48824, USA.
| |
Collapse
|
81
|
Ladas EJ, Bhatia M, Chen L, Sandler E, Petrovic A, Berman DM, Hamblin F, Gates M, Hawks R, Sung L, Nieder M. The safety and feasibility of probiotics in children and adolescents undergoing hematopoietic cell transplantation. Bone Marrow Transplant 2015; 51:262-6. [PMID: 26569091 DOI: 10.1038/bmt.2015.275] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2015] [Revised: 09/18/2015] [Accepted: 10/07/2015] [Indexed: 12/19/2022]
Abstract
Hematopoietic cell transplantation (HCT) has become a standard treatment for many adult and pediatric conditions. Emerging evidence suggests that perturbations in the microbiota diversity increase recipients' susceptibilities to gut-mediated conditions such as diarrhea, infection and acute GvHD. Probiotics preserve the microbiota and may minimize the risk of developing a gut-mediated condition; however, their safety has not been evaluated in the setting of HCT. We evaluated the safety and feasibility of the probiotic, Lactobacillus plantarum (LBP), in children and adolescents undergoing allogeneic HCT. Participants received once-daily supplementation with LBP beginning on day -8 or -7 and continued until day +14. Outcomes were compliance with daily administration and incidence of LBP bacteremia. Administration of LBP was feasible with 97% (30/31, 95% confidence interval (CI) 83-100%) of children receiving at least 50% of the probiotic dose (median 97%; range 50-100%). We did not observe any case of LBP bacteremia (0% (0/30) with 95% CI 0-12%). There were not any unexpected adverse events related to LBP. Our study provides preliminary evidence that administration of LBP is safe and feasible in children and adolescents undergoing HCT. Future steps include the conduct of an approved randomized, controlled trial through Children's Oncology Group.
Collapse
Affiliation(s)
- E J Ladas
- Division of Pediatric Hematology/Oncology/Stem Cell Transplant, Columbia University Medical Center, New York, NY, USA.,Institute of Human Nutrition, College of Physicians and Surgeons, Columbia University Medical Center, New York, NY, USA
| | - M Bhatia
- Division of Pediatric Hematology/Oncology/Stem Cell Transplant, Columbia University Medical Center, New York, NY, USA
| | - L Chen
- Children's Oncology Group, Acadia, CA, USA.,Department of Preventive Medicine, University of Southern California, Los Angeles, CA, USA
| | - E Sandler
- Division of Hematology/Oncology, Nemours Children's Health System, Jacksonville, FL, USA
| | - A Petrovic
- Blood and Bone Marrow Transplantation, All Children's Hospital John Hopkins Medicine, St Petersburg, FL, USA
| | - D M Berman
- Division of Pediatric Infectious Diseases, All Children's Hospital, John Hopkins Medicine, St Petersburg, FL, USA
| | - F Hamblin
- All Children's Hospital, John Hopkins Medicine, St Petersburg, FL, USA
| | - M Gates
- All Children's Hospital, John Hopkins Medicine, St Petersburg, FL, USA
| | - R Hawks
- Division of Pediatric Hematology/Oncology/Stem Cell Transplant, Columbia University Medical Center, New York, NY, USA
| | - L Sung
- Division of Haematology/Oncology, The Hospital for Sick Kids, Toronto, Ontario, Canada
| | - M Nieder
- Department of Blood and Marrow Transplantation, Moffitt Cancer Center, Tampa, FL, USA
| |
Collapse
|
82
|
Wang W, Xu S, Ren Z, Jiang J, Zheng S. Gut microbiota and allogeneic transplantation. J Transl Med 2015; 13:275. [PMID: 26298517 PMCID: PMC4546814 DOI: 10.1186/s12967-015-0640-8] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Accepted: 08/14/2015] [Indexed: 02/06/2023] Open
Abstract
The latest high-throughput sequencing technologies show that there are more than 1000 types of microbiota in the human gut. These microbes are not only important to maintain human health, but also closely related to the occurrence and development of various diseases. With the development of transplantation technologies, allogeneic transplantation has become an effective therapy for a variety of end-stage diseases. However, complications after transplantation still restrict its further development. Post-transplantation complications are closely associated with a host's immune system. There is also an interaction between a person's gut microbiota and immune system. Recently, animal and human studies have shown that gut microbial populations and diversity are altered after allogeneic transplantations, such as liver transplantation (LT), small bowel transplantation (SBT), kidney transplantation (KT) and hematopoietic stem cell transplantation (HTCT). Moreover, when complications, such as infection, rejection and graft versus host disease (GVHD) occur, gut microbial populations and diversity present a significant dysbiosis. Several animal and clinical studies have demonstrated that taking probiotics and prebiotics can effectively regulate gut microbiota and reduce the incidence of complications after transplantation. However, the role of intestinal decontamination in allogeneic transplantation is controversial. This paper reviews gut microbial status after transplantation and its relationship with complications. The role of intervention methods, including antibiotics, probiotics and prebiotics, in complications after transplantation are also discussed. Further research in this new field needs to determine the definite relationship between gut microbial dysbiosis and complications after transplantation. Additionally, further research examining gut microbial intervention methods to ameliorate complications after transplantation is warranted. A better understanding of the relationship between gut microbiota and complications after allogeneic transplantation may make gut microbiota as a therapeutic target in the future.
Collapse
Affiliation(s)
- Weilin Wang
- Department of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, 310003, Zhejiang, China. .,Key Laboratory of Combined Multi-organ Transplantation, Ministry of Public Health, First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, 310003, Zhejiang, China. .,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, 310003, Zhejiang, China.
| | - Shaoyan Xu
- Department of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, 310003, Zhejiang, China. .,Key Laboratory of Combined Multi-organ Transplantation, Ministry of Public Health, First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, 310003, Zhejiang, China. .,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, 310003, Zhejiang, China.
| | - Zhigang Ren
- Department of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, 310003, Zhejiang, China. .,Key Laboratory of Combined Multi-organ Transplantation, Ministry of Public Health, First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, 310003, Zhejiang, China. .,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, 310003, Zhejiang, China.
| | - Jianwen Jiang
- Department of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, 310003, Zhejiang, China. .,Key Laboratory of Combined Multi-organ Transplantation, Ministry of Public Health, First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, 310003, Zhejiang, China. .,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, 310003, Zhejiang, China.
| | - Shusen Zheng
- Department of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, 310003, Zhejiang, China. .,Key Laboratory of Combined Multi-organ Transplantation, Ministry of Public Health, First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, 310003, Zhejiang, China. .,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, 310003, Zhejiang, China.
| |
Collapse
|
83
|
Abstract
This opinion statement discusses indications, efficacy and safety of probiotics in immunosuppressed patients. The best evidence available is for the prophylaxis of infections in patients after liver transplantation and for patients with liver cirrhosis. For other organ transplantations and for bone marrow transplantation the efficacy of probiotic interventions has not been proven yet, but in these patient groups safety is a concern. Also in critically ill patients, the data on efficacy are inconclusive and safety is a concern. In HIV patients and patients after major surgery, probiotic bacteria seem to be safe since there are no associations with increased risks of side effects.
Collapse
Affiliation(s)
- V Stadlbauer
- 1 Medical University of Graz, Department of Gastroenterology and Hematology, Auenbruggerplatz, 8010 Graz, Austria
| |
Collapse
|
84
|
Poutsiaka DD. Editorial commentary: the gut microbiota strikes again. Clin Infect Dis 2015; 61:358-60. [PMID: 25948062 DOI: 10.1093/cid/civ292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Accepted: 04/05/2015] [Indexed: 11/14/2022] Open
Affiliation(s)
- Debra D Poutsiaka
- Division of Geographic Medicine and Infectious Disease, Tufts Medical Center, Boston, Massachusetts
| |
Collapse
|
85
|
Jenq RR, Taur Y, Devlin SM, Ponce DM, Goldberg JD, Ahr KF, Littmann ER, Ling L, Gobourne AC, Miller LC, Docampo MD, Peled JU, Arpaia N, Cross JR, Peets TK, Lumish MA, Shono Y, Dudakov JA, Poeck H, Hanash AM, Barker JN, Perales MA, Giralt SA, Pamer EG, van den Brink MRM. Intestinal Blautia Is Associated with Reduced Death from Graft-versus-Host Disease. Biol Blood Marrow Transplant 2015; 21:1373-83. [PMID: 25977230 PMCID: PMC4516127 DOI: 10.1016/j.bbmt.2015.04.016] [Citation(s) in RCA: 535] [Impact Index Per Article: 59.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Accepted: 04/13/2015] [Indexed: 02/07/2023]
Abstract
The relationship between intestinal microbiota composition and acute graft-versus-host disease (GVHD) after allogeneic blood/marrow transplantation (allo-BMT) is not well understood. Intestinal bacteria have long been thought to contribute to GVHD pathophysiology, but recent animal studies in nontransplant settings have found that anti-inflammatory effects are mediated by certain subpopulations of intestinal commensals. Hypothesizing that a more nuanced relationship may exist between the intestinal bacteria and GVHD, we evaluated the fecal bacterial composition of 64 patients 12 days after BMT. We found that increased bacterial diversity was associated with reduced GVHD-related mortality. Furthermore, harboring increased amounts of bacteria belonging to the genus Blautia was associated with reduced GVHD lethality in this cohort and was confirmed in another independent cohort of 51 patients from the same institution. Blautia abundance was also associated with improved overall survival. We evaluated the abundance of Blautia with respect to clinical factors and found that loss of Blautia was associated with treatment with antibiotics that inhibit anaerobic bacteria and receiving total parenteral nutrition for longer durations. We conclude that increased abundance of commensal bacteria belonging to the Blautia genus is associated with reduced lethal GVHD and improved overall survival.
Collapse
Affiliation(s)
- Robert R Jenq
- Adult BMT, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Weill Cornell Medical College, New York, New York.
| | - Ying Taur
- Weill Cornell Medical College, New York, New York; Infectious Diseases Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Sean M Devlin
- Department of Epidemiology & Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Doris M Ponce
- Adult BMT, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Weill Cornell Medical College, New York, New York
| | - Jenna D Goldberg
- Adult BMT, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Weill Cornell Medical College, New York, New York
| | - Katya F Ahr
- Department of Immunology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Eric R Littmann
- Infectious Diseases Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Lilan Ling
- Department of Immunology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Asia C Gobourne
- Department of Immunology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Liza C Miller
- Department of Immunology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Melissa D Docampo
- Department of Immunology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jonathan U Peled
- Adult BMT, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Weill Cornell Medical College, New York, New York
| | - Nicholas Arpaia
- Department of Immunology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Justin R Cross
- Cell Metabolism Core, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Tatanisha K Peets
- Department of Nutrition, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Melissa A Lumish
- Department of Immunology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Yusuke Shono
- Department of Immunology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jarrod A Dudakov
- Department of Immunology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Hendrik Poeck
- Department of Immunology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Alan M Hanash
- Adult BMT, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Weill Cornell Medical College, New York, New York
| | - Juliet N Barker
- Adult BMT, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Weill Cornell Medical College, New York, New York
| | - Miguel-Angel Perales
- Adult BMT, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Weill Cornell Medical College, New York, New York
| | - Sergio A Giralt
- Adult BMT, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Weill Cornell Medical College, New York, New York
| | - Eric G Pamer
- Weill Cornell Medical College, New York, New York; Infectious Diseases Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Marcel R M van den Brink
- Adult BMT, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Weill Cornell Medical College, New York, New York
| |
Collapse
|
86
|
Chen Y, Zhao Y, Cheng Q, Wu D, Liu H. The Role of Intestinal Microbiota in Acute Graft-versus-Host Disease. J Immunol Res 2015; 2015:145859. [PMID: 26090477 PMCID: PMC4452092 DOI: 10.1155/2015/145859] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2014] [Revised: 02/16/2015] [Accepted: 02/28/2015] [Indexed: 01/16/2023] Open
Abstract
The mammalian intestinal microbiota is a complex ecosystem that plays an important role in host immune responses. Recent studies have demonstrated that alterations in intestinal microbiota composition are linked to multiple inflammatory diseases in humans, including acute graft-versus-host disease (aGVHD). aGVHD is one of the major obstacles in allogeneic hematopoietic stem cell transplantation (allo-HSCT), characterized by tissue damage in the gastrointestinal (GI) tract, liver, lung, and skin. Here, we review the current understanding of the role of intestinal microbiota in the control of immune responses during aGVHD. Additionally, the possibility of using probiotic strains for potential treatment or prevention of aGVHD will be discussed.
Collapse
Affiliation(s)
- Yuanyuan Chen
- Laboratory of Cellular and Molecular Tumor Immunology, Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, China
| | - Ye Zhao
- Cyrus Tang Hematology Center, Department of Hematology, Jiangsu Institute of Hematology, Collaborative Innovation Center of Hematology, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Qiao Cheng
- Laboratory of Cellular and Molecular Tumor Immunology, Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, China
| | - Depei Wu
- Cyrus Tang Hematology Center, Department of Hematology, Jiangsu Institute of Hematology, Collaborative Innovation Center of Hematology, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Haiyan Liu
- Laboratory of Cellular and Molecular Tumor Immunology, Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, China
- Cyrus Tang Hematology Center, Department of Hematology, Jiangsu Institute of Hematology, Collaborative Innovation Center of Hematology, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| |
Collapse
|
87
|
Shono Y, Docampo MD, Peled JU, Perobelli SM, Jenq RR. Intestinal microbiota-related effects on graft-versus-host disease. Int J Hematol 2015; 101:428-37. [PMID: 25812838 DOI: 10.1007/s12185-015-1781-5] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Accepted: 03/16/2015] [Indexed: 12/18/2022]
Abstract
Allogeneic hematopoietic stem cell transplantation (allo-HSCT) is an increasingly important treatment for conditions including hematopoietic malignancies and inherited hematopoietic disorders, and is considered to be the most effective form of tumor immunotherapy available to date. However, graft-versus-host disease (GVHD) remains a major source of morbidity and mortality following allo-HSCT, and understanding the mechanisms of GVHD has been highlighted as a key research priority. During development of GVHD, activation of various immune cells, especially donor T cells, leads to damage of target organs including skin, liver, hematopoietic system, and of particular clinical importance, gut. In addition to histocompatibility complex differences between the donor and recipient, pretransplant conditioning with chemotherapy and irradiation also contributes to GVHD by damaging the gut, resulting in systemic exposure to microbial products normally confined to the intestinal lumen. The intestinal microbiota is a modulator of gastrointestinal immune homeostasis. It also promotes the maintenance of epithelial cells. Recent reports provide growing evidence of the impact of intestinal microbiota on GVHD pathophysiology. This review summarizes current knowledge of changes and effects of intestinal microbiota in the setting of allo-HSCT. We will also discuss potential future strategies of intestinal microbiota manipulation that might be advantageous in decreasing allo-HSCT-related morbidity and mortality.
Collapse
Affiliation(s)
- Yusuke Shono
- Department of Immunology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA,
| | | | | | | | | |
Collapse
|
88
|
Zeiser R. Activation of Innate Immunity in Graft-versus-Host Disease: Implications for Novel Targets? Oncol Res Treat 2015; 38:239-43. [PMID: 25966771 DOI: 10.1159/000381296] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Accepted: 03/02/2015] [Indexed: 11/19/2022]
Abstract
Acute graft-versus-host disease (GvHD) is mediated by alloreactive donor-derived T cells with a suitable T cell receptor recognizing recipient major histocompatibility complex or minor histocompatibility antigens. However, the process of T cell activation and tissue injury sensing is also dependent on innate immune cells and non-hematopoietic cells. Different cell types of the innate immune system have the ability to sense danger-associated and pathogen-associated molecular patterns via pattern recognition receptors which can be transmembrane Toll-like receptors or cytoplasmic nucleotide-binding oligomerization domain-like receptors. Infectious stimuli include bacterial, viral, and fungal components, while non-infectious stimuli can be components derived from damaged cells or extracellular matrix. A better understanding of the complex sensing and effector mechanisms of innate immune cells in GvHD may help to improve preventive and therapeutic strategies in GvHD.
Collapse
Affiliation(s)
- Robert Zeiser
- Department of Hematology and Oncology, Freiburg University Medical Center, Albert-Ludwigs-University, Freiburg i.Br., Germany
| |
Collapse
|
89
|
Sporrer D, Gessner A, Hehlgans T, Oefner PJ, Holler E. The Microbiome and Allogeneic Stem Cell Transplantation. CURRENT STEM CELL REPORTS 2015. [DOI: 10.1007/s40778-014-0006-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
90
|
Docampo MD, Auletta JJ, Jenq RR. Emerging Influence of the Intestinal Microbiota during Allogeneic Hematopoietic Cell Transplantation: Control the Gut and the Body Will Follow. Biol Blood Marrow Transplant 2015; 21:1360-6. [PMID: 25708215 DOI: 10.1016/j.bbmt.2015.02.016] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Accepted: 02/16/2015] [Indexed: 02/07/2023]
Abstract
The intestinal microbiota has many critical roles in maintaining gastrointestinal epithelial and gastrointestinal systemic immune homeostasis. This review provides insight into how allogeneic hematopoietic cell transplantation (HCT) and its associated complications and supportive care therapies affect the microbiota. Additionally, the review discusses how preservation and restoration of the microbiota might be advantageous in decreasing HCT-related morbidity and mortality.
Collapse
Affiliation(s)
- Melissa D Docampo
- Department of Immunology, Weill Cornell Graduate School of Medical Sciences, New York, New York.
| | - Jeffery J Auletta
- Host Defense Program, Hematology/Oncology/BMT and Infectious Diseases, Nationwide Children's Hospital and Department of Pediatrics, The Ohio State University College of Medicine, Columbus, Ohio
| | - Robert R Jenq
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan-Kettering Cancer Center and Weill Cornell Medical College, New York, New York
| |
Collapse
|
91
|
Galvani RG, Lemos R, Areal RB, Salvador PA, Zamboni DS, Wanderley JLM, Bonomo A. Disease severity and mortality can be independently regulated in a mouse model of experimental graft versus host disease. PLoS One 2015; 10:e0118079. [PMID: 25643148 PMCID: PMC4313938 DOI: 10.1371/journal.pone.0118079] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Accepted: 01/05/2015] [Indexed: 12/16/2022] Open
Abstract
Graft versus host disease (GVHD) is the major limitation of allogeneic hematopoietic stem cell transplantation (HSCT) presenting high mortality and morbidity rates. However, the exact cause of death is not completely understood and does not correlate with specific clinical and histological parameters of disease. Here we show, by using a semi-allogeneic mouse model of GVHD, that mortality and morbidity can be experimentally separated. We injected bone marrow-derived dendritic cells (BMDC) from NOD2/CARD15-deficient donors into semi-allogeneic irradiated chimaeras and observed that recipients were protected from death. However, no protection was observed regarding clinical or pathological scores up to 20 days after transplantation. Protection from death was associated with decreased bacterial translocation, faster hematologic recovery and epithelial integrity maintenance despite mononuclear infiltration at day 20 post-GVHD induction with no skew towards different T helper phenotypes. The protected mice recovered from aGVHD and progressively reached scores compatible with healthy animals. Altogether, our data indicate that severity and mortality can be separate events providing a model to study transplant-related mortality.
Collapse
Affiliation(s)
- Rômulo G. Galvani
- Divisão de Medicina Experimental, Coordenação de Pesquisa, Instituto Nacional de Câncer, Rio de Janeiro, Brazil
- Departamento de Imunologia, Instituto de Microbiologia Professor Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ramon Lemos
- Divisão de Medicina Experimental, Coordenação de Pesquisa, Instituto Nacional de Câncer, Rio de Janeiro, Brazil
- Departamento de Imunologia, Instituto de Microbiologia Professor Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Rômulo B. Areal
- Divisão de Medicina Experimental, Coordenação de Pesquisa, Instituto Nacional de Câncer, Rio de Janeiro, Brazil
| | - Pollyanna A. Salvador
- Divisão de Medicina Experimental, Coordenação de Pesquisa, Instituto Nacional de Câncer, Rio de Janeiro, Brazil
| | - Dario S. Zamboni
- Departamento de Biologia Celular, Molecular e Bioagentes Patogênicos, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - João Luiz M. Wanderley
- Divisão de Medicina Experimental, Coordenação de Pesquisa, Instituto Nacional de Câncer, Rio de Janeiro, Brazil
- NUPEM, Campus Macaé Professor Aloísio Teixeira, Universidade Federal do Rio de Janeiro, Macaé, Rio de Janeiro, Brazil
- * E-mail:
| | - Adriana Bonomo
- Divisão de Medicina Experimental, Coordenação de Pesquisa, Instituto Nacional de Câncer, Rio de Janeiro, Brazil
- Departamento de Imunologia, Instituto de Microbiologia Professor Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Laboratorio de Pesquisa sobre o Timo, Instituo Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brazil
| |
Collapse
|
92
|
Integrative Therapy Use for Management of Side Effects and Toxicities Experienced by Pediatric Oncology Patients. CHILDREN-BASEL 2014; 1:424-40. [PMID: 27417488 PMCID: PMC4928739 DOI: 10.3390/children1030424] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Revised: 10/11/2014] [Accepted: 10/29/2014] [Indexed: 01/29/2023]
Abstract
Integrative Therapies (IT), otherwise known as Complementary and Alternative Medicine, are widely used among pediatric oncology patients, despite a paucity of available evidence. This review summarizes surveys that describe the prevalence of IT use by pediatric oncology patients, both during therapy and in survivorship, as well as the modalities being used. Additionally, the evidence that exists for specific treatments that appear to be efficacious in controlling specific symptoms is described. Finally, there are recommendations for practitioners on how to best counsel patients about IT use.
Collapse
|
93
|
Heidegger S, van den Brink MRM, Haas T, Poeck H. The role of pattern-recognition receptors in graft-versus-host disease and graft-versus-leukemia after allogeneic stem cell transplantation. Front Immunol 2014; 5:337. [PMID: 25101080 PMCID: PMC4102927 DOI: 10.3389/fimmu.2014.00337] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2014] [Accepted: 07/03/2014] [Indexed: 02/04/2023] Open
Abstract
Allogeneic hematopoietic stem cell transplantation (allo-HSCT) is the only treatment with curative potential for certain aggressive hematopoietic malignancies. Its success is limited by acute graft-versus-host disease (GVHD), a life-threatening complication that occurs when allo-reactive donor T cells attack recipient organs. There is growing evidence that microbes and innate pattern-recognition receptors (PRRs) such as toll-like receptors (TLR) and nod-like receptors (NLR) are critically involved in the pathogenesis of acute GVHD. Currently, a widely accepted model postulates that intensive chemotherapy and/or total-body irradiation during pre-transplant conditioning results in tissue damage and a loss of epithelial barrier function. Subsequent translocation of bacterial components as well as release of endogenous danger molecules stimulate PRRs of host antigen-presenting cells to trigger the production of pro-inflammatory cytokines (cytokine storm) that modulate T cell allo-reactivity against host tissues, but eventually also the beneficial graft-versus-leukemia (GVL) effect. Given the limitations of existing immunosuppressive therapies, a better understanding of the molecular mechanisms that govern GVHD versus GVL is urgently needed. This may ultimately allow to design modulators, which protect from GvHD but preserve donor T-cell attack on hematologic malignancies. Here, we will briefly summarize current knowledge about the role of innate immunity in the pathogenesis of GVHD and GVL following allo-HSCT.
Collapse
Affiliation(s)
- Simon Heidegger
- III. Medizinische Klinik, Klinikum Rechts der Isar, Technische Universität München , Munich , Germany
| | - Marcel R M van den Brink
- Department of Medicine and Immunology, Memorial Sloan-Kettering Cancer Center , New York, NY , USA
| | - Tobias Haas
- III. Medizinische Klinik, Klinikum Rechts der Isar, Technische Universität München , Munich , Germany
| | - Hendrik Poeck
- III. Medizinische Klinik, Klinikum Rechts der Isar, Technische Universität München , Munich , Germany ; Department of Medicine and Immunology, Memorial Sloan-Kettering Cancer Center , New York, NY , USA
| |
Collapse
|
94
|
Ringdén O, Remberger M, Törlén J, Engström M, Fjaertoft G, Mattsson J, Svahn BM. Home care during neutropenia after allogeneic hematopoietic stem cell transplantation in children and adolescents is safe and may be more advantageous than isolation in hospital. Pediatr Transplant 2014; 18:398-404. [PMID: 24802347 DOI: 10.1111/petr.12262] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/26/2014] [Indexed: 12/25/2022]
Abstract
After ASCT, children are isolated in hospital to prevent neutropenic infections. Patients living within two-h drive from the hospital were given the option of treatment at home after ASCT. Daily visits by an experienced nurse and phone calls from a physician from the unit were included in the protocol. We compared 29 children and adolescents treated at home with 58 matched hospital controls. The children spent a median time of 13 days at home (range 2-24 days) and 6 (0-35) days in hospital. The cumulative incidence of acute GVHD grades II-IV was 21% in the home-care children and 39% in the controls (p = 0.1). Chronic GVHD and probability of relapse were similar in the two groups. TRM at five yr was 11% in the home-care patients and 18% in the controls. Overall survival at three yr was 77% and 62%, respectively (p = 0.33). None of the patients died at home. Median costs were 38,748 euros in the home-care patients and 49,282 euros in those treated in the hospital (p = 0.2). We conclude that it is safe for children and adolescents to be treated at home during the pancytopenic phase after ASCT.
Collapse
Affiliation(s)
- Olle Ringdén
- Division of Therapeutic Immunology, Karolinska Institutet, Stockholm, Sweden; Center for Allogeneic Stem Cell Transplantation, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | | | | | | | | | | | | |
Collapse
|
95
|
Fuji S, Kapp M, Einsele H. Possible implication of bacterial infection in acute graft-versus-host disease after allogeneic hematopoietic stem cell transplantation. Front Oncol 2014; 4:89. [PMID: 24795865 PMCID: PMC4006055 DOI: 10.3389/fonc.2014.00089] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2014] [Accepted: 04/10/2014] [Indexed: 12/24/2022] Open
Abstract
Graft-versus-host disease (GVHD) is still one of the major causes of morbidity and mortality in allogeneic hematopoietic stem cell transplantation (HSCT). In the pathogenesis of acute GVHD, it has been established that donor-derived T-cells activated in the recipient play a major role in GVHD in initiation and maintenance within an inflammatory cascade. To reduce the risk of GVHD, intensification of GVHD prophylaxis like T-cell depletion is effective, but it inevitably increases the risk of infectious diseases and abrogates beneficial graft-versus-leukemia effects. Although various cytokines are considered to play an important role in the pathogenesis of GVHD, GVHD initiation is such a complex process that cannot be prevented by means of single inflammatory cytokine inhibition. Thus, efficient methods to control the whole inflammatory milieu both on cellular and humoral view are needed. In this context, infectious diseases can theoretically contribute to an elevation of inflammatory cytokines after allogeneic HSCT and activation of various subtypes of immune effector cells, which might in summary lead to an aggravation of acute GVHD. The appropriate treatments or prophylaxis of bacterial infection during the early phase after allogeneic HSCT might be beneficial to reduce not only infectious-related but also GVHD-related mortality. Here, we aim to review the literature addressing the interactions of bacterial infections and GVHD after allogeneic HSCT.
Collapse
Affiliation(s)
- Shigeo Fuji
- Division of Hematology, Department of Internal Medicine II, University Hospital of Würzburg , Würzburg , Germany ; Division of Hematopoietic Stem Cell Transplantation, National Cancer Center Hospital , Tokyo , Japan
| | - Markus Kapp
- Division of Hematology, Department of Internal Medicine II, University Hospital of Würzburg , Würzburg , Germany
| | - Hermann Einsele
- Division of Hematology, Department of Internal Medicine II, University Hospital of Würzburg , Würzburg , Germany
| |
Collapse
|
96
|
Marchetti G, Cozzi-Lepri A, Tincati C, Calcagno A, Ceccherini-Silberstein F, De Luca A, Antinori A, Castagna A, Puoti M, Monforte AD. Immune activation and microbial translocation in liver disease progression in HIV/hepatitis co-infected patients: results from the Icona Foundation study. BMC Infect Dis 2014; 14:79. [PMID: 24520976 PMCID: PMC3923986 DOI: 10.1186/1471-2334-14-79] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2013] [Accepted: 02/05/2014] [Indexed: 01/11/2023] Open
Abstract
Background We evaluated whether immune activation (IA) and microbial translocation (MT) might play a role in accelerating liver disease progression in HIV-HBV/HCV co-infected patients. Methods ART-naïve HIV/viral hepatitis co-infected patients from Icona with a CD4 cell count >200/μl and with a known date of prior HIV neg/pos tests and ≥1 plasma sample stored were included in the study. Plasma MT (LPS, sCD14) and IA (IL-6,TNFα) were measured using ELISA while activated CD8 + CD38 + HLA-DR + were measured by flow cytometry, with one measurement being performed for all patients and two measurements for a smaller group of subjects. The association between these biomarkers and the time to i) a single ALT >200 IU/l and ii) a Fib-4 >1.45 was also investigated. A standard survival analysis with robust standard errors was used for all evaluations. Follow-up was censored at patients’ last clinical follow-up. Results We studied 127 HIV-infected hepatitis viruses co-infected patients (118 HCV, 9 HBV). Overall median (IQR) CD4, VL, age were 596/μl (208–1303), 3.8 log10cp/mL (3–4.3), 34 years (22–56). While heightened TNF-α was associated with a 13-fold increased risk of Fib-4 > 1.45 (RH 13.05, 95% CI 2.43-70; p = 0.003), markers of MT did not show an association with liver illness. Interestingly, higher sCD14 was associated with a decreased risk of Fib-4 > 1.45, independently of other biomarkers considered (RH 0.20, 95% CI 0.04-0,9; p = 0.04). Conclusions In HIV/hepatitis virus co-infected ART-naive patients, higher TNF-α plasma levels were associated with a 13-fold increase in the risk of progression to a Fib-4 >1.45, suggesting that the pro-inflammatory status in HIV infection might hasten the course of HCV. In view of the fact that sCD14 may hinder the interaction between LPS and the phagocyte membrane CD14, we herewith propose a model which aims to demonstrate that high sCD14 levels might contribute to shelter liver function through the down-regulation of the inflammatory cascade.
Collapse
Affiliation(s)
- Giulia Marchetti
- Department of Health Sciences- Clinic of Infectious Diseases - "San Paolo" Hospital, University of Milan, via A, di Rudinì, 8-20142 Milan, Italy.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
97
|
Holler E, Butzhammer P, Schmid K, Hundsrucker C, Koestler J, Peter K, Zhu W, Sporrer D, Hehlgans T, Kreutz M, Holler B, Wolff D, Edinger M, Andreesen R, Levine JE, Ferrara JL, Gessner A, Spang R, Oefner PJ. Metagenomic analysis of the stool microbiome in patients receiving allogeneic stem cell transplantation: loss of diversity is associated with use of systemic antibiotics and more pronounced in gastrointestinal graft-versus-host disease. Biol Blood Marrow Transplant 2014; 20:640-5. [PMID: 24492144 DOI: 10.1016/j.bbmt.2014.01.030] [Citation(s) in RCA: 406] [Impact Index Per Article: 40.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2013] [Accepted: 01/28/2014] [Indexed: 12/12/2022]
Abstract
Next-generation sequencing of the hypervariable V3 region of the 16s rRNA gene isolated from serial stool specimens collected from 31 patients receiving allogeneic stem cell transplantation (SCT) was performed to elucidate variations in the composition of the intestinal microbiome in the course of allogeneic SCT. Metagenomic analysis was complemented by strain-specific enterococcal PCR and indirect assessment of bacterial load by liquid chromatography-tandem mass spectrometry of urinary indoxyl sulfate. At the time of admission, patients showed a predominance of commensal bacteria. After transplantation, a relative shift toward enterococci was observed, which was more pronounced under antibiotic prophylaxis and treatment of neutropenic infections. The shift was particularly prominent in patients that developed subsequently or suffered from active gastrointestinal (GI) graft-versus-host disease (GVHD). The mean proportion of enterococci in post-transplant stool specimens was 21% in patients who did not develop GI GVHD as compared with 46% in those that subsequently developed GI GVHD and 74% at the time of active GVHD. Enterococcal PCR confirmed predominance of Enterococcus faecium or both E. faecium and Enterococcus faecalis in these specimens. As a consequence of the loss of bacterial diversity, mean urinary indoxyl sulfate levels dropped from 42.5 ± 11 μmol/L to 11.8 ± 2.8 μmol/L in all post-transplant samples and to 3.5 ± 3 μmol/L in samples from patients with active GVHD. Our study reveals major microbiome shifts in the course of allogeneic SCT that occur in the period of antibiotic treatment but are more prominent in association with GI GVHD. Our data indicate early microbiome shifts and a loss of diversity of the intestinal microbiome that may affect intestinal inflammation in the setting of allogeneic SCT.
Collapse
Affiliation(s)
- Ernst Holler
- Department of Hematology/Oncology, University Medical Center, Regensburg, Germany.
| | - Peter Butzhammer
- Institute of Functional Genomics, University of Regensburg, Regensburg, Germany
| | - Karin Schmid
- Department of Hematology/Oncology, University Medical Center, Regensburg, Germany
| | | | - Josef Koestler
- Institute of Microbiology, University Medical Center, Regensburg, Germany
| | - Katrin Peter
- Department of Hematology/Oncology, University Medical Center, Regensburg, Germany
| | - Wentao Zhu
- Institute of Functional Genomics, University of Regensburg, Regensburg, Germany
| | - Daniela Sporrer
- Department of Hematology/Oncology, University Medical Center, Regensburg, Germany
| | - Thomas Hehlgans
- Institute of Immunology, University of Regensburg, Regensburg, Germany
| | - Marina Kreutz
- Department of Hematology/Oncology, University Medical Center, Regensburg, Germany
| | - Barbara Holler
- Department of Hematology/Oncology, University Medical Center, Regensburg, Germany
| | - Daniel Wolff
- Department of Hematology/Oncology, University Medical Center, Regensburg, Germany
| | - Matthias Edinger
- Department of Hematology/Oncology, University Medical Center, Regensburg, Germany
| | - Reinhard Andreesen
- Department of Hematology/Oncology, University Medical Center, Regensburg, Germany
| | - John E Levine
- University of Michigan Medical Center, Blood and Marrow Transplantation Program, Ann Arbor, Michigan
| | - James L Ferrara
- University of Michigan Medical Center, Blood and Marrow Transplantation Program, Ann Arbor, Michigan
| | - Andre Gessner
- Institute of Microbiology, University Medical Center, Regensburg, Germany
| | - Rainer Spang
- Institute of Functional Genomics, University of Regensburg, Regensburg, Germany
| | - Peter J Oefner
- Institute of Functional Genomics, University of Regensburg, Regensburg, Germany
| |
Collapse
|
98
|
Mercadante ACT, Perobelli SM, Alves APG, Gonçalves-Silva T, Mello W, Gomes-Santos AC, Miyoshi A, Azevedo V, Faria AMC, Bonomo A. Oral combined therapy with probiotics and alloantigen induces B cell-dependent long-lasting specific tolerance. THE JOURNAL OF IMMUNOLOGY 2014; 192:1928-37. [PMID: 24453248 DOI: 10.4049/jimmunol.1301034] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Allogeneic hematopietic stem cell transplantation (aHSCT) is widely used for the treatment of hematologic malignancies. Although aHSCT provides a good response against the malignant cells (graft-versus-leukemia [GVL]), it also leads to the development of graft-versus-host disease (GVHD), a severe disease with high mortality and morbidity rates. Therapy for GVHD is commonly based on nonspecific immunosupression of the transplanted recipient, resulting in the concomitant inhibition of the GVL effect. In this study, we propose an alternative approach to specifically suppress GVHD while sparing the GVL, based on oral treatment of transplant donors with recipient Ags, associated with the intake of probiotic Lactococcus lactis as tolerogenic adjuvant (combined therapy). We show that treatment of C57BL/6 donor mice with combined therapy before the transplant protects the recipients F1 (C57BL/6 × BAL/c) mice from clinical and pathological manifestations of disease, resulting in 100% survival rate. Importantly, the animals keep the immunological competence maintaining the GVL response as well as the response to third-party Ags. The protection is specific, long lasting and dependent on donor IL-10-sufficient B cells activity, which induces regulatory T cells in the host. These data suggest that combined therapy is a promising strategy for prevention of GVHD with preservation of GVL, opening new possibilities to treat human patients subjected to transplantation.
Collapse
Affiliation(s)
- Ana C T Mercadante
- Department of Experimental Medicine, Brazilian National Cancer Institute, Rio de Janeiro 20231-050, Brazil
| | | | | | | | | | | | | | | | | | | |
Collapse
|
99
|
Rogler G. Recognition of commensal microbes: if innate responses are NOD in balance. Expert Rev Clin Immunol 2014; 6:205-10. [DOI: 10.1586/eci.10.1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
100
|
Martin L, Granier A, Lemoine R, Dauba A, Vermeersch S, Aubert-Jacquin C, Baron C, Lebranchu Y, Hoarau C, Velge-Roussel F. Bifidobacteria BbC50 Fermentation Products Induce Human Cd4 + Regulatory T Cells with Antigen-Specific Activation and Bystander Suppression. EUR J INFLAMM 2014. [DOI: 10.1177/1721727x1401200116] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Probiotic bacteria have been shown to have health benefits in various situations (inflammation, allergy, infection). We previously showed that a bacteria-free fermentation product of Bifidobacterium breve C50 (BbC50sn) induced high IL-10 secretion by human dendritic cells. As IL-10 is a regulatory cytokine, the aim of the present study was to examine whether DCs cultured in the presence of BbC50sn could induce regulatory T cells in an allogeneic context. Purified CD4+CD25− human T cells were co-cultured with allogeneic BbC50sn-treated dendritic cells for 4 weeks. The T cell population (BbC50sn-T) was analysed both at phenotypical and functional [ability to inhibit a mixed lymphocyte reaction (MLR)] levels. We showed that T lymphocytes acquired phenotype characteristics of regulatory T cells after 4 weeks of co-culture with BbC50sn-DCs, and inhibited in vitro T lymphocyte proliferation and IFN-γ production in an MLR. Transwell experiments demonstrated that this suppressive activity was not T cell contact-dependent but probably mediated by a soluble factor. Although BbC50sn-T cells secreted significant amounts of IL-10 and TGF-β, their suppressive effect is most likely not mediated through these cytokines. This is, to our knowledge, the first demonstration of in vitro regulatory T cell induction by a bacteria-free fermentation product in an allogeneic context.
Collapse
Affiliation(s)
- L. Martin
- EA 4245 «Cellules Dendritiques, Immunomodulation et Greffes», UFR de Médecine, Université François-Rabelais de Tours, Tours, France
| | - A. Granier
- EA 4245 «Cellules Dendritiques, Immunomodulation et Greffes», UFR de Médecine, Université François-Rabelais de Tours, Tours, France
| | - R. Lemoine
- EA 4245 «Cellules Dendritiques, Immunomodulation et Greffes», UFR de Médecine, Université François-Rabelais de Tours, Tours, France
| | - A. Dauba
- EA 4245 «Cellules Dendritiques, Immunomodulation et Greffes», UFR de Médecine, Université François-Rabelais de Tours, Tours, France
| | - S. Vermeersch
- EA 4245 «Cellules Dendritiques, Immunomodulation et Greffes», UFR de Médecine, Université François-Rabelais de Tours, Tours, France
| | | | - C. Baron
- EA 4245 «Cellules Dendritiques, Immunomodulation et Greffes», UFR de Médecine, Université François-Rabelais de Tours, Tours, France
- Service de Néphrologie et d'Immunologie Clinique, CHRU de Tours, Tours, France
| | - Y. Lebranchu
- EA 4245 «Cellules Dendritiques, Immunomodulation et Greffes», UFR de Médecine, Université François-Rabelais de Tours, Tours, France
- Service de Néphrologie et d'Immunologie Clinique, CHRU de Tours, Tours, France
| | - C. Hoarau
- EA 4245 «Cellules Dendritiques, Immunomodulation et Greffes», UFR de Médecine, Université François-Rabelais de Tours, Tours, France
| | - F. Velge-Roussel
- EA 4245 «Cellules Dendritiques, Immunomodulation et Greffes», UFR de Médecine, Université François-Rabelais de Tours, Tours, France
- UFR des Sciences Pharmaceutiques, Tours, France
| |
Collapse
|