1
|
Xie J, Smith M. The intestinal microbiota and cellular therapy: implications for impact and mechanisms. Blood 2024; 144:1557-1569. [PMID: 39141827 DOI: 10.1182/blood.2024024219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 06/25/2024] [Accepted: 07/16/2024] [Indexed: 08/16/2024] Open
Abstract
ABSTRACT The microbiota, comprising bacteria, fungi, and viruses residing within our bodies, functions as a key modulator in host health and states, including immune responses. Studies have linked microbiota and microbiota-derived metabolites to immune cell functions. In this review, we probe the complex relationship between the human microbiota and clinical outcomes of cellular therapies that leverage immune cells to fight various cancers. With a particular emphasis on hematopoietic cell transplantation and chimeric antigen receptor T-cell therapy, we explore the potential mechanisms underpinning this interaction. We also highlight the interventional applications of the microbiota in cellular therapy while outlining future research directions in the field.
Collapse
Affiliation(s)
- Jiayi Xie
- Division of Blood and Marrow Transplantation and Cellular Therapy, Department of Medicine, Stanford University School of Medicine, Stanford, CA
| | - Melody Smith
- Division of Blood and Marrow Transplantation and Cellular Therapy, Department of Medicine, Stanford University School of Medicine, Stanford, CA
| |
Collapse
|
2
|
Pötgens SA, Havelange V, Lecop S, Li F, Neyrinck AM, Bindels F, Neveux N, Demoulin JB, Moors I, Kerre T, Maertens J, Walter J, Schoemans H, Delzenne NM, Bindels LB. Gut microbiome alterations at acute myeloid leukemia diagnosis are associated with muscle weakness and anorexia. Haematologica 2024; 109:3194-3208. [PMID: 38546675 PMCID: PMC11443375 DOI: 10.3324/haematol.2023.284138] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 03/19/2024] [Indexed: 10/02/2024] Open
Abstract
The gut microbiota makes critical contributions to host homeostasis, and its role in the treatment of acute myeloid leukemia (AML) has attracted attention. We investigated whether the gut microbiome is affected by AML, and whether such changes are associated with hallmarks of cachexia. Biological samples and clinical data were collected from 30 antibiotic- free AML patients at diagnosis and matched volunteers (1:1) in a multicenter, cross-sectional, prospective study. The composition and functional potential of the fecal microbiota were analyzed using shotgun metagenomics. Fecal, blood, and urinary metabolomics analyses were performed. AML patients displayed muscle weakness, anorexia, signs of altered gut function, and glycemic disorders. The composition of the fecal microbiota differed between patients with AML and control subjects, with an increase in oral bacteria. Alterations in bacterial functions and fecal metabolome support an altered redox status in the gut microbiota, which may contribute to the altered redox status observed in patients with AML. Eubacterium eligens, reduced 3-fold in AML patients, was strongly correlated with muscle strength and citrulline, a marker of enterocyte mass and function. Blautia and Parabacteroides, increased in patients with AML, were correlated with anorexia. Several bacterial taxa and metabolites (e.g., Blautia, Prevotella, phenylacetate, and hippurate) previously associated with glycemic disorders were altered. Our work revealed important perturbations in the gut microbiome of AML patients at diagnosis, which are associated with muscle strength, altered redox status, and anorexia. These findings pave the way for future mechanistic work to explore the function and therapeutic potential of the bacteria identified in this study.
Collapse
Affiliation(s)
- Sarah A Pötgens
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels
| | - Violaine Havelange
- Department of Hematology, Cliniques Universitaires Saint-Luc, UCLouvain, Université catholique de Louvain, Brussels, Belgium; Experimental Medicine Unit, De Duve Institute, UCLouvain, Université catholique de Louvain, Brussels
| | - Sophie Lecop
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels
| | - Fuyong Li
- Department of Infectious Diseases and Public Health, Jockey Club College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Kowloon, Hong Kong SAR, China; Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta
| | - Audrey M Neyrinck
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels
| | | | - Nathalie Neveux
- Clinical Chemistry Department, Cochin Hospital, Paris Centre University Hospitals, Paris
| | - Jean-Baptiste Demoulin
- Experimental Medicine Unit, De Duve Institute, UCLouvain, Université catholique de Louvain, Brussels
| | - Ine Moors
- Department of Hematology, Ghent University Hospital, Ghent University, Ghent
| | - Tessa Kerre
- Department of Hematology, Ghent University Hospital, Ghent University, Ghent
| | - Johan Maertens
- Department of Hematology, University Hospitals Leuven, Leuven, Belgium; Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven
| | - Jens Walter
- Department of Medicine, School of Microbiology, APC Microbiome Ireland, University College Cork, Cork
| | - Hélène Schoemans
- Department of Hematology, University Hospitals Leuven, Leuven, Belgium; Department of Public Health and Primary Care, ACCENT VV, KU Leuven, Leuven
| | - Nathalie M Delzenne
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels
| | - Laure B Bindels
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium; Welbio Department, WEL Research Institute, Wavre.
| |
Collapse
|
3
|
Harris AC, Ganjaei K, Vilela C, Geyer A. Late-Onset Noninfectious Pulmonary Complications after Hematopoietic Stem Cell Transplantation. Transplant Cell Ther 2024; 30:S585-S596. [PMID: 39370238 DOI: 10.1016/j.jtct.2024.05.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 05/25/2024] [Indexed: 10/08/2024]
Affiliation(s)
- Andrew C Harris
- Pediatric Transplantation and Cellular Therapy, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Kimia Ganjaei
- Pulmonary Service. Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Camila Vilela
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Alexander Geyer
- Division of Pulmonary, Critical Care and Sleep Medicine, Lenox Hill Hospital, Northwell Health, New York, New York.
| |
Collapse
|
4
|
Liu J, Lyu M, Zhang YY, Mo XD, Sun YQ, Yan CH, Wang Y, Xu LP, Zhang XH, Liu KY, Huang XJ. [Causes and characteristics of pre-engraftment mortality after allogeneic hematopoietic stem cell transplantation]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2024; 45:542-548. [PMID: 39134484 PMCID: PMC11310806 DOI: 10.3760/cma.j.cn121090-20231030-00240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Indexed: 12/06/2024]
Abstract
Objective: To analyze the causes and demographic characteristics of pre-engraftment mortality in patients who underwent allogeneic hematopoietic stem cell transplantation (allo-HSCT) and investigate the risk factors and measures for preventing pre-engraftment mortality. Methods: A retrospective case analysis, involving a total of 7 427 patients who underwent allo-HSCT at Peking University People's Hospital between January 2016 and July 2023, was conducted. Results: Among the 7 427 patients who underwent allo-HSCT, 56 cases (0.75% ) experienced pre-engraftment mortality. The median time to death for these 56 patients was +7 (-3 to +38) days after stem cell infusion. The median times to death for patients with acute leukemia (AL), severe aplastic anemia (SAA), and myelodysplastic syndrome (MDS) were +11 (-1 to +38), +3 (-1 to +34), and +16 (-1 to +38) days, respectively (P=0.013). The main causes of pre-engraftment mortality were infection (39.3% ), cardiac toxicity (28.6% ), and intracranial hemorrhage (26.8% ). Infection was the most common cause of pre-engraftment mortality in patients with AL and MDS (55.0% and 60.0% ), whereas cardiac toxicity was predominantly observed in patients with SAA (71.4% ), with no cases in patients with AL and only one case in patients with MDS. Among patients who died from intracranial hemorrhage, 53.3% had severe infections. The median times to death for infection, cardiac toxicity, and intracranial hemorrhage was +11 (-1 to +38), +2.5 (-1 to +17), and +8 (-3 to +37) days, respectively (P<0.001) . Conclusions: Infection is the primary cause of pre-engraftment mortality in allo-HSCT, and severe cardiac toxicity leading to pre-engraftment mortality should be closely monitored in patients with SAA.
Collapse
Affiliation(s)
- J Liu
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematological Diseases, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation for the Treatment of Hematology, Beijing 100044, China
| | - M Lyu
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematological Diseases, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation for the Treatment of Hematology, Beijing 100044, China
| | - Y Y Zhang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematological Diseases, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation for the Treatment of Hematology, Beijing 100044, China
| | - X D Mo
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematological Diseases, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation for the Treatment of Hematology, Beijing 100044, China
| | - Y Q Sun
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematological Diseases, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation for the Treatment of Hematology, Beijing 100044, China
| | - C H Yan
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematological Diseases, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation for the Treatment of Hematology, Beijing 100044, China
| | - Y Wang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematological Diseases, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation for the Treatment of Hematology, Beijing 100044, China
| | - L P Xu
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematological Diseases, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation for the Treatment of Hematology, Beijing 100044, China
| | - X H Zhang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematological Diseases, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation for the Treatment of Hematology, Beijing 100044, China
| | - K Y Liu
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematological Diseases, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation for the Treatment of Hematology, Beijing 100044, China
| | - X J Huang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematological Diseases, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation for the Treatment of Hematology, Beijing 100044, China
| |
Collapse
|
5
|
Elgarten CW, Margolis EB, Kelly MS. The Microbiome and Pediatric Transplantation. J Pediatric Infect Dis Soc 2024; 13:S80-S89. [PMID: 38417089 PMCID: PMC10901476 DOI: 10.1093/jpids/piad062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 08/25/2023] [Indexed: 03/01/2024]
Abstract
The microbial communities that inhabit our bodies have been increasingly linked to host physiology and pathophysiology. This microbiome, through its role in colonization resistance, influences the risk of infections after transplantation, including those caused by multidrug-resistant organisms. In addition, through both direct interactions with the host immune system and via the production of metabolites that impact local and systemic immunity, the microbiome plays an important role in the establishment of immune tolerance after transplantation, and conversely, in the development of graft-versus-host disease and graft rejection. This review offers a comprehensive overview of the evidence for the role of the microbiome in hematopoietic cell and solid organ transplant complications, drivers of microbiome shift during transplantation, and the potential of microbiome-based therapies to improve pediatric transplantation outcomes.
Collapse
Affiliation(s)
- Caitlin W Elgarten
- Division of Oncology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Pediatrics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Elisa B Margolis
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
- Department of Pediatrics, University of Tennessee Health Sciences Center, Memphis, Tennessee, USA
| | - Matthew S Kelly
- Departments of Pediatrics and Molecular Genetics & Microbiology, Duke University School of Medicine, Durham, North Carolina, USA
| |
Collapse
|
6
|
Wang S, Yue X, Zhou H, Chen X, Chen H, Hu L, Pan W, Zhao X, Xiao H. The association of intestinal microbiota diversity and outcomes of allogeneic hematopoietic cell transplantation: a systematic review and meta-analysis. Ann Hematol 2023; 102:3555-3566. [PMID: 37770617 DOI: 10.1007/s00277-023-05460-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Accepted: 09/17/2023] [Indexed: 09/30/2023]
Abstract
Growing evidence suggests that highly intestinal microbiota diversity modulates host inflammation and promotes immune tolerance. Several studies have reported that patients undergoing allo-HSCT have experienced microbiota disruption that is characterized by expansion of potentially pathogenic bacteria and loss of microbiota diversity. Thus, the primary aim of this meta-analysis was to determine the association of intestinal microbiota diversity and outcomes after allo-HSCT, and the secondary aim was to analyze the associations of some specific microbiota abundances with the outcomes of allo-HSCT. Electronic databases of Pubmed, Embase, Web of Science, and Cochrane Library were searched from inception to August 2023, and 17 studies were found eligible. The pooled estimate suggested that higher intestinal microbiota diversity was significantly associated with overall survival (OS) benefit (HR = 0.66, 95% CI: 0.55-0.78), as well as decreased risk of transplant-related mortality (HR = 0.56, 95% CI: 0.41-0.76), and lower incidence of grade II-IV aGVHD (HR = 0.41, 95% CI: 0.27-0.63). Furthermore, higher abundance of Clostridiales was associated with a superior OS (HR = 0.40, 95% CI: 0.18-0.87), while higher abundance of Enterococcus (HR = 2.03, 95% CI: 1.55-2.65), γ-proteobacteria (HR = 2.82, 95% CI: 1.53-5.20), and Candida (HR = 3.80, 95% CI: 1.32-10.94) was an adverse prognostic factor for OS. Overall, this meta-analysis highlights the protective role of higher intestinal microbiota diversity on outcomes after allo-HSCT during both pre-transplant and post-transplant periods. Some specific microbiota can be useful in the identification of patients at risk of mortality, offering new tools for individualized pre-emptive or therapeutic strategies to improve allo-HSCT outcomes.
Collapse
Affiliation(s)
- Shufen Wang
- Department of Hematology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, No. 3 Qingchun East Rd, Hangzhou, 310016, Zhejiang Province, People's Republic of China
- Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang Province, People's Republic of China
| | - Xiaoyan Yue
- Department of Hematology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, No. 3 Qingchun East Rd, Hangzhou, 310016, Zhejiang Province, People's Republic of China
- Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang Province, People's Republic of China
| | - Hongyu Zhou
- Department of Hematology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, No. 3 Qingchun East Rd, Hangzhou, 310016, Zhejiang Province, People's Republic of China
- Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang Province, People's Republic of China
| | - Xu Chen
- Department of Hematology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, No. 3 Qingchun East Rd, Hangzhou, 310016, Zhejiang Province, People's Republic of China
- Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang Province, People's Republic of China
| | - Huiqiao Chen
- Department of Hematology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, No. 3 Qingchun East Rd, Hangzhou, 310016, Zhejiang Province, People's Republic of China
- Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang Province, People's Republic of China
| | - Liangning Hu
- Department of Hematology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, No. 3 Qingchun East Rd, Hangzhou, 310016, Zhejiang Province, People's Republic of China
| | - Wenjue Pan
- Department of Hematology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, No. 3 Qingchun East Rd, Hangzhou, 310016, Zhejiang Province, People's Republic of China
| | - Xiujie Zhao
- Department of Hematology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, No. 3 Qingchun East Rd, Hangzhou, 310016, Zhejiang Province, People's Republic of China
| | - Haowen Xiao
- Department of Hematology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, No. 3 Qingchun East Rd, Hangzhou, 310016, Zhejiang Province, People's Republic of China.
- Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang Province, People's Republic of China.
| |
Collapse
|
7
|
Rashidi A, Ebadi M, Rehman TU, Elhusseini H, Kazadi D, Halaweish H, Khan MH, Hoeschen A, Cao Q, Luo X, Kabage AJ, Lopez S, Holtan SG, Weisdorf DJ, Khoruts A, Staley C. Randomized Double-Blind Phase II Trial of Fecal Microbiota Transplantation Versus Placebo in Allogeneic Hematopoietic Cell Transplantation and AML. J Clin Oncol 2023; 41:5306-5319. [PMID: 37235836 PMCID: PMC10691796 DOI: 10.1200/jco.22.02366] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 04/03/2023] [Accepted: 04/27/2023] [Indexed: 05/28/2023] Open
Abstract
PURPOSE Gut microbiota injury in allogeneic hematopoietic cell transplantation (HCT) recipients and patients with AML has been associated with adverse clinical outcomes. Previous studies in these patients have shown improvements in various microbiome indices after fecal microbiota transplantation (FMT). However, whether microbiome improvements translate into improved clinical outcomes remains unclear. We examined this question in a randomized, double-blind, placebo-controlled phase II trial. METHODS Two independent cohorts of allogeneic HCT recipients and patients with AML receiving induction chemotherapy were randomly assigned in a 2:1 ratio to receive standardized oral encapsulated FMT versus placebo upon neutrophil recovery. After each course of antibacterial antibiotics, patients received a study treatment. Up to three treatments were administered within 3 months. The primary end point was 4-month all-cause infection rate. Patients were followed for 9 months. RESULTS In the HCT cohort (74 patients), 4-month infection density was 0.74 and 0.91 events per 100 patient-days in FMT and placebo arms, respectively (infection rate ratio, 0.83; 95% CI, 0.48 to 1.42; P = .49). In the AML cohort (26 patients), 4-month infection density was 0.93 in the FMT arm and 1.25 in the placebo arm, with an infection rate ratio of 0.74 (95% CI, 0.32 to 1.71; P = .48). Unique donor bacterial sequences comprised 25%-30% of the fecal microbiota after FMT. FMT improved postantibiotic recovery of microbiota diversity, restored several depleted obligate anaerobic commensals, and reduced the abundance of expanded genera Enterococcus, Streptococcus, Veillonella, and Dialister. CONCLUSION In allogeneic HCT recipients and patients with AML, third-party FMT was safe and ameliorated intestinal dysbiosis, but did not decrease infections. Novel findings from this trial will inform future development of FMT trials.
Collapse
Affiliation(s)
- Armin Rashidi
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN
- Clinical Research Division, Fred Hutchinson Cancer Center; and Division of Oncology, University of Washington, Seattle, WA
| | - Maryam Ebadi
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN
- Department of Radiation Oncology, University of Washington, Seattle, WA
| | - Tauseef Ur Rehman
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN
| | - Heba Elhusseini
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN
| | - David Kazadi
- Department of Medicine, University of Minnesota, Minneapolis, MN
| | | | | | - Andrea Hoeschen
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN
| | - Qing Cao
- Biostatistics Core, Masonic Cancer Center, University of Minnesota, Minneapolis, MN
| | - Xianghua Luo
- Biostatistics Core, Masonic Cancer Center, University of Minnesota, Minneapolis, MN
- Division of Biostatistics, School of Public Health, University of Minnesota, Minneapolis, MN
| | | | - Sharon Lopez
- Center for Immunology, University of Minnesota, Minneapolis, MN
| | - Shernan G. Holtan
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN
| | - Daniel J. Weisdorf
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN
| | - Alexander Khoruts
- Center for Immunology, University of Minnesota, Minneapolis, MN
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, University of Minnesota, Minneapolis, MN
- Biotechnology Institute, University of Minnesota, St Paul, MN
| | | |
Collapse
|
8
|
El Jurdi N, Holtan SG, Hoeschen A, Velguth J, Hillmann B, Betts BC, MacMillan ML, Weisdorf DJ, Khoruts A, Rashidi A, Shields-Cutler R. Pre-transplant and longitudinal changes in faecal microbiome characteristics are associated with subsequent development of chronic graft-versus-host disease. Br J Haematol 2023; 203:288-294. [PMID: 37553783 DOI: 10.1111/bjh.19016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 06/26/2023] [Accepted: 07/24/2023] [Indexed: 08/10/2023]
Abstract
The role of the gastrointestinal microbiome in predisposing to chronic graft-versus-host disease (cGVHD), an immune-mediated haematopoietic cell transplant (HCT) complication, is not well defined. We examined the relationship of the host faecal microbiome with subsequent cGVHD development by analysing baseline stool samples as well as post-HCT changes in microbiome composition and metabolite pathway analyses. We analysed pre-transplant baseline samples from 11 patients who subsequently developed cGVHD compared to 13 controls who did not develop acute GVHD or cGVHD at any time. We found a significant differential abundance of multiple taxa at baseline between cGVHD versus controls, including the Actinobacteria phylum and Clostridium genus. A subgroup analysis of longitudinal samples within each patient revealed a greater loss of alpha diversity from baseline to post-engraftment in patients who subsequently developed cGVHD. Metabolic pathways analysis revealed that two pathways associated with short-chain fatty acid metabolism were enriched in cGVHD patient microbiomes: β-oxidation and acyl-CoA synthesis, and γ-aminobutyrate shunt. In contrast, a tryptophan catabolism pathway was enriched in controls. Our findings show a distinct pattern of baseline microbiome and metabolic capacity that may play a role in modulating alloreactivity in patients developing cGVHD. These findings support the therapeutic potential of microbiome manipulation for cGVHD prevention.
Collapse
Affiliation(s)
- Najla El Jurdi
- Blood and Marrow Transplant Program, Departments of Medicine and Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| | - Shernan G Holtan
- Blood and Marrow Transplant Program, Departments of Medicine and Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| | - Andrea Hoeschen
- Blood and Marrow Transplant Program, Departments of Medicine and Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| | - Jessica Velguth
- College of Biological Sciences, BioTechnology Institute, University of Minnesota, Saint Paul, Minnesota, USA
| | - Benjamin Hillmann
- Department of Computer Science and Engineering, University of Minnesota, Minneapolis, Minnesota, USA
| | - Brian C Betts
- Blood and Marrow Transplant Program, Departments of Medicine and Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| | - Margaret L MacMillan
- Blood and Marrow Transplant Program, Departments of Medicine and Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Daniel J Weisdorf
- Blood and Marrow Transplant Program, Departments of Medicine and Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| | - Alexander Khoruts
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, University of Minnesota, Minneapolis, Minnesota, USA
- Center for Immunology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Armin Rashidi
- Blood and Marrow Transplant Program, Departments of Medicine and Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| | | |
Collapse
|
9
|
Margolis EB, Maron G, Sun Y, Dallas RH, Allison KJ, Ferrolino J, Ross HS, Davis AE, Jia Q, Turner P, Mackay V, Morin CE, Triplett BM, Klein EJ, Englund JA, Tang L, Hayden RT. Microbiota Predict Infections and Acute Graft-Versus-Host Disease After Pediatric Allogeneic Hematopoietic Stem Cell Transplantation. J Infect Dis 2023; 228:627-636. [PMID: 37249910 PMCID: PMC10469318 DOI: 10.1093/infdis/jiad190] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 04/19/2023] [Accepted: 05/27/2023] [Indexed: 05/31/2023] Open
Abstract
BACKGROUND Despite preventive measures, infections continue to pose significant risks to pediatric allogeneic hematopoietic cell transplantation (allo-HCT) recipients. The gut microbiota has been linked to clinical outcomes following adult allo-HCT. This study evaluated whether similar disruptions or differing microbiota patterns were associated with infection risk in pediatric allo-HCT. METHODS In a prospective observational study, fecal samples were obtained from 74 children before conditioning and upon neutrophil recovery. Microbiome signatures identified through sequencing were examined for their associations with infections or acute graft-versus-host disease (aGVHD) in the first-year post-HCT using Cox proportional hazards analysis. RESULTS Microbiome disruption in adults, did not predict infection risk in pediatric allo-HCT. Unique microbiota signatures were associated with different infections or aGVHD. A ratio of strict and facultative anaerobes (eg, Lachnoclostridium, Parabacteroides) prior to conditioning predicted bacteremia risk (Cox hazard ratio [HR], 3.89). A distinct ratio of oral (eg, Rothia, Veillonella) to intestinal anaerobes (eg, Anaerobutyricum, Romboutsia) at neutrophil recovery predicted likelihood of bacterial infections (Cox HR, 1.81) and viral enterocolitis (Cox HR, 1.96). CONCLUSIONS Interactions between medical interventions, pediatric hosts, and microbial communities contribute to microbiota signatures that predict infections. Further multicenter study is necessary to validate the generalizability of these ratios as biomarkers.
Collapse
Affiliation(s)
- Elisa B Margolis
- Department of Infectious Diseases, St Jude Children’s Research Hospital
- Department of Pediatrics, University of Tennessee Health Sciences Center
| | - Gabriela Maron
- Department of Infectious Diseases, St Jude Children’s Research Hospital
- Department of Pediatrics, University of Tennessee Health Sciences Center
| | - Yilun Sun
- Department of Biostatistics, St Jude Children’s Research Hospital
| | - Ronald H Dallas
- Department of Infectious Diseases, St Jude Children’s Research Hospital
| | - Kim J Allison
- Department of Infectious Diseases, St Jude Children’s Research Hospital
| | - Jose Ferrolino
- Department of Infectious Diseases, St Jude Children’s Research Hospital
| | - Hailey S Ross
- Department of Infectious Diseases, St Jude Children’s Research Hospital
| | - Amy E Davis
- Department of Infectious Diseases, St Jude Children’s Research Hospital
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Sciences Center, Memphis
| | - Qidong Jia
- Department of Infectious Diseases, St Jude Children’s Research Hospital
| | - Paige Turner
- Department of Infectious Diseases, St Jude Children’s Research Hospital
| | - Victoria Mackay
- Department of Infectious Diseases, St Jude Children’s Research Hospital
| | - Cara E Morin
- Division of Radiology and Medical Imaging, Cincinnati Children's Hospital, Ohio
| | - Brandon M Triplett
- Department of Bone Marrow Transplantation and Cellular Therapy, St Jude Children’s Research Hospital, Memphis, Tennessee
| | | | | | - Li Tang
- Department of Biostatistics, St Jude Children’s Research Hospital
| | | |
Collapse
|
10
|
Chen D, Zeng Q, Liu L, Zhou Z, Qi W, Yu S, Zhao L. Global Research Trends on the Link Between the Microbiome and COPD: A Bibliometric Analysis. Int J Chron Obstruct Pulmon Dis 2023; 18:765-783. [PMID: 37180751 PMCID: PMC10167978 DOI: 10.2147/copd.s405310] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 04/30/2023] [Indexed: 05/16/2023] Open
Abstract
Background The pathogenesis of chronic obstructive pulmonary disease (COPD) has been studied in relation to the microbiome, providing space for more targeted interventions and new treatments. Numerous papers on the COPD microbiome have been reported in the last 10 years, yet few publications have used bibliometric methods to evaluate this area. Methods We searched the Web of Science Core Collection for all original research articles in the field of COPD microbiome from January 2011 to August 2022 and used CiteSpace for visual analysis. Results A total of 505 relevant publications were obtained, and the number of global publications in this field is steadily increasing every year, with China and the USA occupying the first two spots in international publications. Imperial College London and the University of Leicester produced the most publications. Brightling C from the UK was the most prolific writer, while Huang Y and Sze M from the USA were first and second among the authors cited. The American Journal of Respiratory and Critical Care Medicine had the highest frequency of citations. The top 10 institutions, cited authors and journals are mostly from the UK and the US. In the ranking of citations, the first article was a paper published by Sze M on changes in the lung tissue's microbiota in COPD patients. The keywords "exacerbation", "gut microbiota", "lung microbiome", "airway microbiome", "bacterial colonization", and "inflammation" were identified as cutting-edge research projects for 2011-2022. Conclusion Based on the visualization results, in the future, we can use the gut-lung axis as the starting point to explore the immunoinflammatory mechanism of COPD, and study how to predict the effects of different treatments of COPD by identifying the microbiota, and how to achieve the optimal enrichment of beneficial bacteria and the optimal consumption of harmful bacteria to improve COPD.
Collapse
Affiliation(s)
- Daohong Chen
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, People’s Republic of China
| | - Qian Zeng
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, People’s Republic of China
| | - Lu Liu
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, People’s Republic of China
| | - Ziyang Zhou
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, People’s Republic of China
| | - Wenchuan Qi
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, People’s Republic of China
| | - Shuguang Yu
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, People’s Republic of China
| | - Ling Zhao
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, People’s Republic of China
| |
Collapse
|
11
|
Yin G, Guo Y, Ding Q, Ma S, Chen F, Wang Q, Chen H, Wang H. Klebsiella quasipneumoniae in intestine damages bile acid metabolism in hematopoietic stem cell transplantation patients with bloodstream infection. J Transl Med 2023; 21:230. [PMID: 36991414 PMCID: PMC10061697 DOI: 10.1186/s12967-023-04068-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 03/17/2023] [Indexed: 03/31/2023] Open
Abstract
BACKGROUND Bloodstream infection (BSI) is a serious hematopoietic stem cell transplantation (HSCT) complication. The intestinal microbiome regulates host metabolism and maintains intestinal homeostasis. Thus, the impact of microbiome on HSCT patients with BSI is essential. METHODS Stool and serum specimens of HSCT patients were prospectively collected from the pretransplant conditioning period till 4 months after transplantation. Specimens of 16 patients without BSI and 21 patients before BSI onset were screened for omics study using 16S rRNA gene sequencing and untargeted metabolomics. The predictive infection model was constructed using LASSO and the logistic regression algorithm. The correlation and influence of microbiome and metabolism were examined in mouse and Caco-2 cell monolayer models. RESULTS The microbial diversity and abundance of Lactobacillaceae were remarkably reduced, but the abundance of Enterobacteriaceae (especially Klebsiella quasipneumoniae) was significantly increased in the BSI group before onset, compared with the non-BSI group. The family score of microbiome features (Enterobacteriaceae and Butyricicoccaceae) could highly predict BSI (AUC = 0.879). The serum metabolomic analysis showed that 16 differential metabolites were mainly enriched in the primary bile acid biosynthesis pathway, and the level of chenodeoxycholic acid (CDCA) was positively correlated with the abundance of K. quasipneumoniae (R = 0.406, P = 0.006). The results of mouse experiments confirmed that three serum primary bile acids levels (cholic acid, isoCDCA and ursocholic acid), the mRNA expression levels of bile acid farnesol X receptor gene and apical sodium-dependent bile acid transporter gene in K. quasipneumoniae colonized mice were significantly higher than those in non-colonized mice. The intestinal villus height, crypt depth, and the mRNA expression level of tight junction protein claudin-1 gene in K. quasipneumoniae intestinal colonized mice were significantly lower than those in non-colonized mice. In vitro, K. quasipneumoniae increased the clearance of FITC-dextran by Caco-2 cell monolayer. CONCLUSIONS This study demonstrated that the intestinal opportunistic pathogen, K. quasipneumoniae, was increased in HSCT patients before BSI onset, causing increased serum primary bile acids. The colonization of K. quasipneumoniae in mice intestines could lead to mucosal integrity damage. The intestinal microbiome features of HSCT patients were highly predictive of BSI and could be further used as potential biomarkers.
Collapse
Affiliation(s)
- Guankun Yin
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, 100044, China
- Institute of Medical Technology, Peking University Health Science Center, Beijing, 100191, China
| | - Yifan Guo
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, 100044, China
| | - Qi Ding
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, 100044, China
| | - Shuai Ma
- Institute of Medical Technology, Peking University Health Science Center, Beijing, 100191, China
| | - Fengning Chen
- Institute of Medical Technology, Peking University Health Science Center, Beijing, 100191, China
| | - Qi Wang
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, 100044, China
| | - Hongbin Chen
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, 100044, China
| | - Hui Wang
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, 100044, China.
- Institute of Medical Technology, Peking University Health Science Center, Beijing, 100191, China.
| |
Collapse
|
12
|
Upadhyay Banskota S, Skupa SA, El-Gamal D, D’Angelo CR. Defining the Role of the Gut Microbiome in the Pathogenesis and Treatment of Lymphoid Malignancies. Int J Mol Sci 2023; 24:2309. [PMID: 36768631 PMCID: PMC9916782 DOI: 10.3390/ijms24032309] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 01/15/2023] [Accepted: 01/17/2023] [Indexed: 01/26/2023] Open
Abstract
The gut microbiome is increasingly being recognized as an important immunologic environment, with direct links to the host immune system. The scale of the gut microbiome's genomic repertoire extends the capacity of its host's genome by providing additional metabolic output, and the close communication between gut microbiota and mucosal immune cells provides a continued opportunity for immune education. The relationship between the gut microbiome and the host immune system has important implications for oncologic disease, including lymphoma, a malignancy derived from within the immune system itself. In this review, we explore past and recent discoveries describing the role that bacterial populations play in lymphomagenesis, diagnosis, and therapy. We highlight key relationships within the gut microbiome-immune-oncology axis that present exciting opportunities for directed interventions intended to shape the microbiome for therapeutic effect. We conclude with a limited summary of active clinical trials targeting the microbiome in hematologic malignancies, along with future directions on gut microbiome investigations within lymphoid malignancies.
Collapse
Affiliation(s)
- Shristi Upadhyay Banskota
- Division of Hematology and Oncology, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Sydney A. Skupa
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Dalia El-Gamal
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Christopher R. D’Angelo
- Division of Hematology and Oncology, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
13
|
Shi Z, Li H, Song W, Zhou Z, Li Z, Zhang M. Emerging roles of the gut microbiota in cancer immunotherapy. Front Immunol 2023; 14:1139821. [PMID: 36911704 PMCID: PMC9992551 DOI: 10.3389/fimmu.2023.1139821] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Accepted: 02/13/2023] [Indexed: 02/24/2023] Open
Abstract
Gut microbiota represents a hidden treasure vault encompassing trillions of microorganisms that inhabit the intestinal epithelial barrier of the host. In the past decade, numerous in-vitro, animal and clinical studies have revealed the profound roles of gut microbiota in maintaining the homeostasis of various physiological functions, especially immune modulation, and remarkable differences in the configuration of microbial communities between cancers and healthy individuals. In addition, although considerable efforts have been devoted to cancer treatments, there remain many patients succumb to their disease with the incremental cancer burden worldwide. Nevertheless, compared with the stability of human genome, the plasticity of gut microbiota renders it a promising opportunity for individualized treatment. Meanwhile, burgeoning findings indicate that gut microbiota is involved in close interactions with the outcomes of diverse cancer immunotherapy protocols, including immune checkpoint blockade therapy, allogeneic hematopoietic stem cell transplantation, and chimeric antigen receptor T cell therapy. Here, we reviewed the evidence for the capacity of gut microflora to modulate cancer immunotherapies, and highlighted the opportunities of microbiota-based prognostic prediction, as well as microbiotherapy by targeting the microflora to potentiate anticancer efficacy while attenuating toxicity, which will be pivotal to the development of personalized cancer treatment strategies.
Collapse
Affiliation(s)
- Zhuangzhuang Shi
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Lymphoma Diagnosis and Treatment Centre of Henan Province, Zhengzhou, China.,Academy of Medical Sciences of Zhengzhou University, Zhengzhou, Henan, China
| | - Hongwen Li
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Lymphoma Diagnosis and Treatment Centre of Henan Province, Zhengzhou, China
| | - Wenting Song
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Lymphoma Diagnosis and Treatment Centre of Henan Province, Zhengzhou, China.,Academy of Medical Sciences of Zhengzhou University, Zhengzhou, Henan, China
| | - Zhiyuan Zhou
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Lymphoma Diagnosis and Treatment Centre of Henan Province, Zhengzhou, China
| | - Zhaoming Li
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Lymphoma Diagnosis and Treatment Centre of Henan Province, Zhengzhou, China.,State Key Laboratory of Esophageal Cancer Prevention and Treatment and Henan Key Laboratory for Esophageal Cancer Research, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Mingzhi Zhang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Lymphoma Diagnosis and Treatment Centre of Henan Province, Zhengzhou, China.,State Key Laboratory of Esophageal Cancer Prevention and Treatment and Henan Key Laboratory for Esophageal Cancer Research, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
14
|
Chi M, Jiang T, He X, Peng H, Li Y, Zhang J, Wang L, Nian Q, Ma K, Liu C. Role of Gut Microbiota and Oxidative Stress in the Progression of Transplant-Related Complications following Hematopoietic Stem Cell Transplantation. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2023; 2023:3532756. [PMID: 37113743 PMCID: PMC10129428 DOI: 10.1155/2023/3532756] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 10/12/2022] [Accepted: 11/24/2022] [Indexed: 04/29/2023]
Abstract
Hematopoietic stem cell transplantation (HSCT), also known as bone marrow transplantation, has curative potential for various hematologic malignancies but is associated with risks such as graft-versus-host disease (GvHD), severe bloodstream infection, viral pneumonia, idiopathic pneumonia syndrome (IPS), lung fibrosis, and sinusoidal obstruction syndrome (SOS), which severely deteriorate clinical outcomes and limit the wide application of HSCT. Recent research has provided important insights into the effects of gut microbiota and oxidative stress (OS) on HSCT complications. Therefore, based on recent studies, we describe intestinal dysbiosis and OS in patients with HSCT and review recent molecular findings underlying the causal relationships of gut microbiota, OS, and transplant-related complications, focusing particularly on the involvement of gut microbiota-mediated OS in postengraftment complications. Also, we discuss the use of antioxidative and anti-inflammatory probiotics to manipulate gut microbiota and OS, which have been associated with promising effects in improving HSCT outcomes.
Collapse
Affiliation(s)
- Mingxuan Chi
- Department of Nephrology, Sichuan Provincial People's Hospital, Sichuan Renal Disease Clinical Research Center, University of Electronic Science and Technology of China, Chengdu, China
- Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu 610072, China
| | - Tao Jiang
- Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu 610072, China
- Department of Hematology, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan Province 610072, China
| | - Xing He
- School of Clinical Medicine, Chengdu Medical College, China
| | - Haoyu Peng
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Yunlong Li
- Department of Urology, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Jiong Zhang
- Department of Nephrology, Sichuan Provincial People's Hospital, Sichuan Renal Disease Clinical Research Center, University of Electronic Science and Technology of China, Chengdu, China
- Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu 610072, China
| | - Li Wang
- Department of Nephrology, Sichuan Provincial People's Hospital, Sichuan Renal Disease Clinical Research Center, University of Electronic Science and Technology of China, Chengdu, China
- Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu 610072, China
| | - Qing Nian
- Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu 610072, China
- Department of Blood Transfusion, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Kuai Ma
- Department of Nephrology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Chi Liu
- Department of Nephrology, Sichuan Provincial People's Hospital, Sichuan Renal Disease Clinical Research Center, University of Electronic Science and Technology of China, Chengdu, China
- Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu 610072, China
| |
Collapse
|
15
|
Tyszka M, Maciejewska-Markiewicz D, Biliński J, Lubas A, Stachowska E, Basak GW. Increased Intestinal Permeability and Stool Zonulin, Calprotectin and Beta-Defensin-2 Concentrations in Allogenic Hematopoietic Cell Transplantation Recipients. Int J Mol Sci 2022; 23:ijms232415962. [PMID: 36555600 PMCID: PMC9781277 DOI: 10.3390/ijms232415962] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 12/08/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022] Open
Abstract
Significant progress has been made in understanding the connection between intestinal barrier function and allogenic hematopoietic cell transplantation (allo-HCT) recipients' outcomes. The purpose of this study was to further evaluate gut barrier permeability and other potential intestinal barrier disruption markers in the allo-HCT setting. Fifty-one patients were enrolled in the study. Intestinal permeability was assessed with the sugar absorption test and faecal concentrations of the zonulin, calprotectin and beta-defensin-2 levels in the peri-transplantation period. Most patients undergoing allo-HCT in our department had a disrupted intestinal barrier at the baseline, which was associated with older age and higher Hematopoietic Cell Transplantation-specific Comorbidity Index (HCT-CI). Regardless of this, we observed a further increase in gut barrier permeability after allo-HCT in most patients. However, there was no association between permeability assay and other markers (zonulin, calprotectin and beta-defensin-2). Patients with acute GVHD had significantly higher median calprotectin concentrations after allo-HCT compared with the patients without this complication. Our findings indicate that gut barrier damage develops prior to allo-HCT with progression after the procedure and precedes further complications, but did not prove other markers to be useful surrogates of intestinal permeability.
Collapse
Affiliation(s)
- Martyna Tyszka
- Department of Hematology, Transplantation and Internal Medicine, Medical University of Warsaw, 02-091 Warsaw, Poland
- Correspondence: (M.T.); (D.M.-M.)
| | - Dominika Maciejewska-Markiewicz
- Department of Human Nutrition and Metabolomics, Pomeranian Medical University, 71-460 Szczecin, Poland
- Correspondence: (M.T.); (D.M.-M.)
| | - Jarosław Biliński
- Department of Hematology, Transplantation and Internal Medicine, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Arkadiusz Lubas
- Department of Internal Medicine, Nephrology and Dialysis, Military Institute of Medicine, 04-141 Warsaw, Poland
| | - Ewa Stachowska
- Department of Human Nutrition and Metabolomics, Pomeranian Medical University, 71-460 Szczecin, Poland
| | - Grzegorz W. Basak
- Department of Hematology, Transplantation and Internal Medicine, Medical University of Warsaw, 02-091 Warsaw, Poland
| |
Collapse
|
16
|
Muratore E, Leardini D, Baccelli F, Venturelli F, Prete A, Masetti R. Nutritional modulation of the gut microbiome in allogeneic hematopoietic stem cell transplantation recipients. Front Nutr 2022; 9:993668. [PMID: 36337625 PMCID: PMC9632163 DOI: 10.3389/fnut.2022.993668] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 10/04/2022] [Indexed: 11/23/2022] Open
Abstract
Allogeneic hematopoietic stem cell transplantation (allo-HSCT) represents a potentially curative strategy for many oncological and non-oncological diseases, but it is associated with marked morbidity and mortality. The disruption of gut microbiota (GM) eubiosis has been linked to major allo-HSCT complications, including infections and acute graft vs. host disease (aGvHD), and correlates with mortality. This increasing knowledge on the role of the GM in the allo-HSCT procedure has led to fascinating ideas for modulating the intestinal ecosystem in order to improve clinical outcomes. Nutritional strategies, either by changing the route of nutritional supplementation or by administering specific molecules, are increasingly being considered as cost- and risk-effective methods of modulating the GM. Nutritional support has also emerged in the past several years as a key feature in supportive care for allo-HSCT recipients, and deterioration of nutritional status is associated with decreased overall survival and higher complication rates during treatment. Herein we provide a complete overview focused on nutritional modulation of the GM in allo-HSCT recipients. We address how pre transplant diet could affect GM composition and its ability to withstand the upsetting events occurring during transplantation. We also provide a complete overview on the influence of the route of nutritional administration on the intestinal ecosystem, with a particular focus on the comparison between enteral and parenteral nutrition (PN). Moreover, as mounting evidence are showing how specific components of post-transplant diet, such as lactose, could drastically shape the GM, we will also summarize the role of prebiotic supplementation in the modulation of the intestinal flora and in allo-HSCT outcomes.
Collapse
Affiliation(s)
- Edoardo Muratore
- Pediatric Oncology and Hematology “Lalla Seràgnoli,” IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Davide Leardini
- Pediatric Oncology and Hematology “Lalla Seràgnoli,” IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Francesco Baccelli
- Pediatric Oncology and Hematology “Lalla Seràgnoli,” IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
- *Correspondence: Francesco Baccelli,
| | - Francesco Venturelli
- Pediatric Oncology and Hematology “Lalla Seràgnoli,” IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
| | - Arcangelo Prete
- Pediatric Oncology and Hematology “Lalla Seràgnoli,” IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Riccardo Masetti
- Pediatric Oncology and Hematology “Lalla Seràgnoli,” IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| |
Collapse
|
17
|
He Y, Li J, Yu W, Zheng Y, Yang D, Xu Y, Zhao L, Ma X, Gong P, Gao Z. Characteristics of lower respiratory tract microbiota in the patients with post-hematopoietic stem cell transplantation pneumonia. Front Cell Infect Microbiol 2022; 12:943317. [PMID: 36176576 PMCID: PMC9513191 DOI: 10.3389/fcimb.2022.943317] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 08/23/2022] [Indexed: 11/25/2022] Open
Abstract
Background Pneumonia is a leading cause of non-relapse mortality after hematopoietic stem cell transplantation (HSCT), and the lower respiratory tract (LRT) microbiome has been proven to be associated with various respiratory diseases. However, little is known about the characteristics of the LRT microbiome in patients with post-HSCT compared to healthy controls (HC) and community-acquired pneumonia (CAP). Methods Bronchoalveolar lavage samples from 55 patients with post-HSCT pneumonia, 44 patients with CAP, and 30 healthy volunteers were used to detect microbiota using 16S rRNA gene sequencing. Results The diversity of the LRT microbiome significantly decreased in patients with post-HSCT pneumonia, and the overall community was different from the CAP and HC groups. At the phylum level, post-HSCT pneumonia samples had a high abundance of Actinobacteria and a relatively low abundance of Bacteroidetes. The same is true for non-survivors compared with survivors in patients with post-HSCT pneumonia. At the genus level, the abundances of Pseudomonas, Acinetobacter, Burkholderia, and Mycobacterium were prominent in the pneumonia group after HSCT. On the other hand, gut-associated bacteria, Enterococcus were more abundant in the non-survivors. Some pathways concerning amino acid and lipid metabolism were predicted to be altered in patients with post-HSCT pneumonia. Conclusions Our results reveal that the LRT microbiome in patients with post-HSCT pneumonia differs from CAP patients and healthy controls, which could be associated with the outcome. The LRT microbiota could be a target for intervention during post-HSCT pneumonia.
Collapse
Affiliation(s)
- Yukun He
- Department of Respiratory and Critical Care Medicine, Peking University People's Hospital, Beijing, China
| | - Jia Li
- Department of Respiratory and Critical Care Medicine, Peking University People's Hospital, Beijing, China
| | - Wenyi Yu
- Department of Respiratory and Critical Care Medicine, Peking University People's Hospital, Beijing, China
| | - Yali Zheng
- Department of Respiratory and Critical Care Medicine, Peking University People's Hospital, Beijing, China
- Department of Respiratory, Critical Care, and Sleep Medicine, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Donghong Yang
- Department of Respiratory and Critical Care Medicine, Peking University People's Hospital, Beijing, China
| | - Yu Xu
- Department of Respiratory and Critical Care Medicine, Peking University People's Hospital, Beijing, China
| | - Lili Zhao
- Department of Respiratory and Critical Care Medicine, Peking University People's Hospital, Beijing, China
| | - Xinqian Ma
- Department of Respiratory and Critical Care Medicine, Peking University People's Hospital, Beijing, China
| | - Pihua Gong
- Department of Respiratory and Critical Care Medicine, Peking University People's Hospital, Beijing, China
- *Correspondence: Pihua Gong, ; Zhancheng Gao,
| | - Zhancheng Gao
- Department of Respiratory and Critical Care Medicine, Peking University People's Hospital, Beijing, China
- *Correspondence: Pihua Gong, ; Zhancheng Gao,
| |
Collapse
|
18
|
Sen T, Thummer RP. The Impact of Human Microbiotas in Hematopoietic Stem Cell and Organ Transplantation. Front Immunol 2022; 13:932228. [PMID: 35874759 PMCID: PMC9300833 DOI: 10.3389/fimmu.2022.932228] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 06/06/2022] [Indexed: 11/18/2022] Open
Abstract
The human microbiota heavily influences most vital aspects of human physiology including organ transplantation outcomes and transplant rejection risk. A variety of organ transplantation scenarios such as lung and heart transplantation as well as hematopoietic stem cell transplantation is heavily influenced by the human microbiotas. The human microbiota refers to a rich, diverse, and complex ecosystem of bacteria, fungi, archaea, helminths, protozoans, parasites, and viruses. Research accumulating over the past decade has established the existence of complex cross-species, cross-kingdom interactions between the residents of the various human microbiotas and the human body. Since the gut microbiota is the densest, most popular, and most studied human microbiota, the impact of other human microbiotas such as the oral, lung, urinary, and genital microbiotas is often overshadowed. However, these microbiotas also provide critical and unique insights pertaining to transplantation success, rejection risk, and overall host health, across multiple different transplantation scenarios. Organ transplantation as well as the pre-, peri-, and post-transplant pharmacological regimens patients undergo is known to adversely impact the microbiotas, thereby increasing the risk of adverse patient outcomes. Over the past decade, holistic approaches to post-transplant patient care such as the administration of clinical and dietary interventions aiming at restoring deranged microbiota community structures have been gaining momentum. Examples of these include prebiotic and probiotic administration, fecal microbial transplantation, and bacteriophage-mediated multidrug-resistant bacterial decolonization. This review will discuss these perspectives and explore the role of different human microbiotas in the context of various transplantation scenarios.
Collapse
Affiliation(s)
| | - Rajkumar P. Thummer
- Laboratory for Stem Cell Engineering and Regenerative Medicine, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, India
| |
Collapse
|
19
|
Wang D, Wang W, Ding Y, Tang M, Zhang L, Chen J, You H. Metagenomic Next-Generation Sequencing Successfully Detects Pulmonary Infectious Pathogens in Children With Hematologic Malignancy. Front Cell Infect Microbiol 2022; 12:899028. [PMID: 35837477 PMCID: PMC9273861 DOI: 10.3389/fcimb.2022.899028] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 05/31/2022] [Indexed: 12/24/2022] Open
Abstract
Background Pulmonary infection is a leading cause of mortality in pediatric patients with hematologic malignancy (HM). In clinical settings, pulmonary pathogens are frequently undetectable, and empiric therapies may be costly, ineffective and lead to poor outcomes in this vulnerable population. Metagenomic next-generation sequencing (mNGS) enhances pathogen detection, but data on its application in pediatric patients with HM and pulmonary infections are scarce. Methods We retrospectively reviewed 55 pediatric patients with HM and pulmonary infection who were performed mNGS on bronchoalveolar lavage fluid from January 2020 to October 2021. The performances of mNGS methods and conventional microbiological methods in pathogenic diagnosis and subsequently antibiotic adjustment were investigated. Results A definite or probable microbial etiology of pulmonary infection was established for 50 of the 55 patients (90.9%) when mNGS was combined with conventional microbiological tests. The positive rate was 87.3% (48 of 55 patients) for mNGS versus 34.5% (19 of 55 patients) with conventional microbiological methods (P < 0.001). Bacteria, viruses and fungi were detected in 17/55 (30.9%), 25/55 (45.5%) and 19/55 (34.5%) cases using mNGS, respectively. Furthermore, 17 patients (30.9%) were identified as pulmonary mixed infections. Among the 50 pathogen-positive cases, 38% (19/50) were not completely pathogen-covered by empirical antibiotics and all of them were accordingly made an antibiotic adjustment. In the present study, the 30-day mortality rate was 7.3%. Conclusion mNGS is a valuable diagnostic tool to determine the etiology and appropriate treatment in pediatric patients with HM and pulmonary infection. In these vulnerable children with HM, pulmonary infections are life-threatening, so we recommend that mNGS should be considered as a front-line diagnostic test.
Collapse
|
20
|
Zhang ZJ, Lehmann CJ, Cole CG, Pamer EG. Translating Microbiome Research From and To the Clinic. Annu Rev Microbiol 2022; 76:435-460. [DOI: 10.1146/annurev-micro-041020-022206] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Extensive research has elucidated the influence of the gut microbiota on human health and disease susceptibility and resistance. We review recent clinical and laboratory-based experimental studies associating the gut microbiota with certain human diseases. We also highlight ongoing translational advances that manipulate the gut microbiota to treat human diseases and discuss opportunities and challenges in translating microbiome research from and to the bedside. Expected final online publication date for the Annual Review of Microbiology, Volume 76 is September 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Zhenrun J. Zhang
- Duchossois Family Institute, University of Chicago, Chicago, Illinois, USA
| | | | - Cody G. Cole
- Duchossois Family Institute, University of Chicago, Chicago, Illinois, USA
- Department of Microbiology, University of Chicago, Chicago, Illinois, USA
| | - Eric G. Pamer
- Duchossois Family Institute, University of Chicago, Chicago, Illinois, USA
- Department of Microbiology, University of Chicago, Chicago, Illinois, USA
- Department of Medicine and Pathology, University of Chicago, Chicago, Illinois, USA
| |
Collapse
|
21
|
Pianko MJ, Golob JL. Host-microbe interactions and outcomes in multiple myeloma and hematopoietic stem cell transplantation. Cancer Metastasis Rev 2022; 41:367-382. [PMID: 35488106 PMCID: PMC9378527 DOI: 10.1007/s10555-022-10033-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 04/16/2022] [Indexed: 12/12/2022]
Abstract
Microbiota are essential to normal immune development and there is growing recognition of its importance to human health and disease and deepening understanding of the complexity of host-microbe interactions in the human gut and other tissues. Commensal microbes not only can influence host immunity locally through impacts of bioactive microbial metabolites and direct interactions with epithelial cells and innate immune receptors but also can exert systemic immunomodulatory effects via impacts on host immune cells capable of trafficking beyond the gut. Emerging data suggest microbiota influence the development of multiple myeloma (MM), a malignancy of the immune system derived from immunoglobulin-producing bone marrow plasma cells, through the promotion of inflammation. Superior treatment outcomes for MM correlate with a higher abundance of commensal microbiota capable of influencing inflammatory responses through the production of butyrate. In patients with hematologic malignancies, higher levels of diversity of the gut microbiota correlate with superior outcomes after hematopoietic stem cell transplantation. Correlative data support the impact of commensal microbiota on survival, risk of infection, disease relapse, and graft-versus-host disease (GVHD) after transplant. In this review, we will discuss the current understanding of the role of host-microbe interactions and the inflammatory tumor microenvironment of multiple myeloma, discuss data describing the key role of microbiota in hematopoietic stem cell transplantation for treatment of hematologic malignancies, and highlight several possible concepts for interventions directed at the gut microbiota to influence treatment outcomes.
Collapse
Affiliation(s)
- Matthew J Pianko
- Department of Internal Medicine, Division of Hematology/Oncology, University of Michigan, Ann Arbor, MI, USA.
| | - Jonathan L Golob
- Department of Internal Medicine, Division of Infectious Diseases, University of Michigan, Ann Arbor, MI, USA
- Department of Microbiology & Immunology, Division of Infectious Diseases, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
22
|
Miltiadous O, Waters NR, Andrlová H, Dai A, Nguyen CL, Burgos da Silva M, Lindner S, Slingerland J, Giardina P, Clurman A, Armijo GK, Gomes ALC, Lakkaraja M, Maslak P, Scordo M, Shouval R, Staffas A, O'Reilly R, Taur Y, Prockop S, Boelens JJ, Giralt S, Perales MA, Devlin SM, Peled JU, Markey KA, van den Brink MRM. Early intestinal microbial features are associated with CD4 T-cell recovery after allogeneic hematopoietic transplant. Blood 2022; 139:2758-2769. [PMID: 35061893 PMCID: PMC9074404 DOI: 10.1182/blood.2021014255] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 12/23/2021] [Indexed: 11/20/2022] Open
Abstract
Low intestinal microbial diversity is associated with poor outcomes after allogeneic hematopoietic cell transplantation (HCT). Using 16S rRNA sequencing of 2067 stool samples and flow cytometry data from 2370 peripheral blood samples drawn from 894 patients who underwent allogeneic HCT, we have linked features of the early post-HCT microbiome with subsequent immune cell recovery. We examined lymphocyte recovery and microbiota features in recipients of both unmodified and CD34-selected allografts. We observed that fecal microbial diversity was an independent predictor of CD4 T-cell count 3 months after HCT in recipients of a CD34-selected allograft, who are dependent on de novo lymphopoiesis for their immune recovery. In multivariate models using clinical factors and microbiota features, we consistently observed that increased fecal relative abundance of genus Staphylococcus during the early posttransplant period was associated with worse CD4 T-cell recovery. Our observations suggest that the intestinal bacteria, or the factors they produce, can affect early lymphopoiesis and the homeostasis of allograft-derived T cells after transplantation.
Collapse
Affiliation(s)
- Oriana Miltiadous
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Nicholas R Waters
- Department of Immunology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center (MSKCC), New York, NY
| | - Hana Andrlová
- Department of Immunology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center (MSKCC), New York, NY
| | - Anqi Dai
- Department of Immunology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center (MSKCC), New York, NY
| | - Chi L Nguyen
- Department of Immunology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center (MSKCC), New York, NY
| | - Marina Burgos da Silva
- Department of Immunology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center (MSKCC), New York, NY
| | - Sarah Lindner
- Department of Immunology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center (MSKCC), New York, NY
| | - John Slingerland
- Department of Immunology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center (MSKCC), New York, NY
| | - Paul Giardina
- Department of Immunology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center (MSKCC), New York, NY
| | - Annelie Clurman
- Department of Immunology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center (MSKCC), New York, NY
| | - Gabriel K Armijo
- Department of Immunology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center (MSKCC), New York, NY
| | - Antonio L C Gomes
- Department of Immunology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center (MSKCC), New York, NY
| | - Madhavi Lakkaraja
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Pediatrics, Weill Cornell Medicine, New York, NY
| | - Peter Maslak
- Immunology Laboratory Service, Department of Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Leukemia Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Medicine, Weill Cornell Medical College, New York, NY
| | - Michael Scordo
- Department of Medicine, Weill Cornell Medical College, New York, NY
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Roni Shouval
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Anna Staffas
- Sahlgrenska Center for Cancer Research, Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Sweden
- Clinical Genetics and Genomics, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Richard O'Reilly
- Stem Cell Transplant and Cellular Therapy Service, Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Ying Taur
- Department of Medicine, Weill Cornell Medical College, New York, NY
- Infectious Disease Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Susan Prockop
- Department of Pediatrics, Weill Cornell Medicine, New York, NY
- Stem Cell Transplant and Cellular Therapy Service, Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Jaap Jan Boelens
- Department of Pediatrics, Weill Cornell Medicine, New York, NY
- Stem Cell Transplant and Cellular Therapy Service, Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Sergio Giralt
- Department of Medicine, Weill Cornell Medical College, New York, NY
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Miguel-Angel Perales
- Department of Medicine, Weill Cornell Medical College, New York, NY
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Sean M Devlin
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Jonathan U Peled
- Department of Medicine, Weill Cornell Medical College, New York, NY
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Kate A Markey
- Department of Medicine, Weill Cornell Medical College, New York, NY
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Clinical Research Division, Fred Hutchinson Cancer Research Center (FHCRC), Seattle, WA; and
- Division of Medical Oncology, University of Washington, Seattle, WA
| | - Marcel R M van den Brink
- Department of Immunology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center (MSKCC), New York, NY
- Department of Medicine, Weill Cornell Medical College, New York, NY
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| |
Collapse
|
23
|
Brevi A, Cogrossi LL, Lorenzoni M, Mattorre B, Bellone M. The Insider: Impact of the Gut Microbiota on Cancer Immunity and Response to Therapies in Multiple Myeloma. Front Immunol 2022; 13:845422. [PMID: 35371048 PMCID: PMC8968065 DOI: 10.3389/fimmu.2022.845422] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 02/25/2022] [Indexed: 11/13/2022] Open
Abstract
The human microbiota is a unique set of microorganisms colonizing the human body and evolving within it from the very beginning. Acting as an insider, the microbiota provides nutrients, and mutualistically interacts with the host’s immune system, thus contributing to the generation of barriers against pathogens. While a strong link has been documented between intestinal dysbiosis (i.e., disruption to the microbiota homeostasis) and diseases, the mechanisms by which commensal bacteria impact a wide spectrum of mucosal and extramucosal human disorders have only partially been deciphered. This is particularly puzzling for multiple myeloma (MM), a treatable but incurable neoplasia of plasma cells that accumulate in the bone marrow and lead to end-organ damage. Here we revise the most recent literature on data from both the bench and the bedside that show how the gut microbiota modulates cancer immunity, potentially impacting the progression of asymptomatic monoclonal gammopathy of undetermined significance (MGUS) and smoldering MM (SMM) to full blown MM. We also explore the effect of the gut microbiome on hematopoietic stem cell transplantation, chemotherapy, immunomodulating therapy and cancer immunotherapy in MM patients. Additionally, we identify the most cogent area of investigation that have the highest chance to delineate microbiota-related and pathobiology-based parameters for patient risk stratification. Lastly, we highlight microbiota-modulating strategies (i.e., diet, prebiotics, probiotics, fecal microbiota transplantation and postbiotics) that may reduce treatment-related toxicity in patients affected by MM as well as the rates of undertreatment of SMM patients.
Collapse
Affiliation(s)
- Arianna Brevi
- Cellular Immunology Unit, Department of Immunology, Transplantation and Infectious Diseases, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ospedale San Raffaele, Milan, Italy
| | - Laura Lucia Cogrossi
- Cellular Immunology Unit, Department of Immunology, Transplantation and Infectious Diseases, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ospedale San Raffaele, Milan, Italy
- Università Vita-Salute San Raffaele, Milan, Italy
| | - Marco Lorenzoni
- Cellular Immunology Unit, Department of Immunology, Transplantation and Infectious Diseases, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ospedale San Raffaele, Milan, Italy
| | - Benedetta Mattorre
- Cellular Immunology Unit, Department of Immunology, Transplantation and Infectious Diseases, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ospedale San Raffaele, Milan, Italy
| | - Matteo Bellone
- Cellular Immunology Unit, Department of Immunology, Transplantation and Infectious Diseases, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ospedale San Raffaele, Milan, Italy
- *Correspondence: Matteo Bellone,
| |
Collapse
|
24
|
Caldararo M, Algazaq J, Schmidt E, Joanow A, Mariano A, Predmore J, Politikos I, Cho C, Stein EM, Wen X, Travis WD, Raible K, Cintron M, Babady NE, Papanicolaou G, Lee YJ. Atypical pathogens presenting with pulmonary consolidations detected by cell-free DNA next-generation sequencing in patients with hematologic malignancies. INFECTIOUS DISEASES IN CLINICAL PRACTICE 2022; 30:e1101. [PMID: 35586753 PMCID: PMC9109825 DOI: 10.1097/ipc.0000000000001101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Identification of pathogens with pulmonary presentation in patients with hematologic malignancies may be challenging due to diagnostic difficulty related to the underlying malignancy and limitations of conventional microbiologic methods. Herein, we present a case series of three patients with pulmonary consolidations due to Legionella bozemanae necrotizing pneumonia, Pneumocystis jirovecii pneumonia, and disseminated Scedosporium infection, who were diagnosed by microbial cell-free DNA next-generation sequencing. We observed that this new sequencing modality was in agreement with gold-standard diagnostics, posing a potential solution to the problem of limited capability in diagnosing infections in hematological malignancy patients.
Collapse
Affiliation(s)
- Mario Caldararo
- Infectious Diseases Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Jumanah Algazaq
- Infectious Diseases Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Elizabeth Schmidt
- Infectious Diseases Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Alexa Joanow
- Infectious Diseases Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Amanda Mariano
- Infectious Diseases Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Jacqueline Predmore
- Infectious Diseases Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Ioannis Politikos
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Weill Medical College, Cornell University, New York, NY
| | - Christina Cho
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Weill Medical College, Cornell University, New York, NY
| | - Eytan M. Stein
- Leukemia Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Weill Medical College, Cornell University, New York, NY
| | - Xiaoyoun Wen
- Weill Medical College, Cornell University, New York, NY
| | - William D. Travis
- Pathology Department, Memorial Sloan Kettering Cancer Center, New York, NY
- Weill Medical College, Cornell University, New York, NY
| | - Kevin Raible
- Clinical Microbiology Service, Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Melvili Cintron
- Clinical Microbiology Service, Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - N. Esther Babady
- Infectious Diseases Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Clinical Microbiology Service, Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Genovefa Papanicolaou
- Infectious Diseases Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Weill Medical College, Cornell University, New York, NY
| | - Yeon Joo Lee
- Infectious Diseases Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Weill Medical College, Cornell University, New York, NY
| |
Collapse
|
25
|
Tu H, Xiao E, Liu O. Taking Microbiota into Consideration in Mesenchymal Stem Cell Research. J Dent Res 2022; 101:880-886. [PMID: 35196924 DOI: 10.1177/00220345221077986] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are a promising therapy in regenerative medicine, but the clinical efficacy has yet to be identified, because the functions of MSCs are modulated by many factors, including the age and health condition of donors, origin of the tissue, and several other unknown factors. Recently, it has been revealed that, besides host factors, the microbiota that inhabits the human body is a modulator of MSCs as well. Here, we highlight the role of microbiota in the alteration of MSCs functions, with a specific focus on the self-renewal ability, multiple differentiation potential, and the immunomodulation capacity of MSCs. We also review the clinical trials and model research on the synergic and antagonistic effects of microbiota in stem cell therapy. In addition, we discuss the underlying mechanisms of the interplay between microbiota and MSCs, which are elucidated using omics approaches followed by verification experiments. As oral and maxillofacial tissues are important sources of MSCs, as well as a major access to diverse microbes, further studies are needed to elucidate these interactions in the oral field to make greater advancements in regenerative medicine.
Collapse
Affiliation(s)
- H Tu
- Hunan Key Laboratory of Oral Health Research and Human 3D Printing Engineering Research Central of Oral Care and Hunan Clinical Research Center of Oral Major Diseases and Oral Health and Xiangya Stomatological Hospital and Xiangya School of Stomatology, Central South University, Changsha City, Hunan Province, P.R. China
| | - E Xiao
- Beijing Maybio Pharmaceutical Biotechnology Development Co., Ltd., Changsha City, Hunan Province, P.R. China
| | - O Liu
- Hunan Key Laboratory of Oral Health Research and Human 3D Printing Engineering Research Central of Oral Care and Hunan Clinical Research Center of Oral Major Diseases and Oral Health and Xiangya Stomatological Hospital and Xiangya School of Stomatology, Central South University, Changsha City, Hunan Province, P.R. China
| |
Collapse
|
26
|
Saksena N, Keon M. Gut microbiome: a balancing act between degeneration and regeneration. Neural Regen Res 2022; 17:1967-1968. [PMID: 35142678 PMCID: PMC8848625 DOI: 10.4103/1673-5374.335151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Affiliation(s)
- Nitin Saksena
- OMICS Centre of Excellence, G42 Healthcare, Masdar City, Abu Dhabi. UAE. And Victoria University, Melbourne, Australia
| | - Matt Keon
- GeneiUS, Darlinghurst, Sydney, Australia
| |
Collapse
|
27
|
Elgarten CW, Tanes C, Lee JJ, Danziger-Isakov LA, Grimley MS, Green M, Michaels MG, Barnum JL, Ardura MI, Auletta JJ, Blumenstock J, Seif AE, Bittinger KL, Fisher BT. Early stool microbiome and metabolome signatures in pediatric patients undergoing allogeneic hematopoietic cell transplantation. Pediatr Blood Cancer 2022; 69:e29384. [PMID: 34709713 PMCID: PMC8629955 DOI: 10.1002/pbc.29384] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 08/25/2021] [Accepted: 09/16/2021] [Indexed: 01/03/2023]
Abstract
BACKGROUND The contribution of the gastrointestinal tract microbiome to outcomes after allogeneic hematopoietic cell transplantation (HCT) is increasingly recognized. Investigations of larger pediatric cohorts aimed at defining the microbiome state and associated metabolic patterns pretransplant are needed. METHODS We sought to describe the pretransplant stool microbiome in pediatric allogenic HCT patients at four centers. We performed shotgun metagenomic sequencing and untargeted metabolic profiling on pretransplant stool samples. Samples were compared with normal age-matched controls and by clinical characteristics. We then explored associations between stool microbiome measurements and metabolite concentrations. RESULTS We profiled stool samples from 88 pediatric allogeneic HCT patients, a median of 4 days before transplant. Pretransplant stool samples differed from healthy controls based on indices of alpha diversity and in the proportional abundance of specific taxa and bacterial genes. Relative to stool from healthy patients, samples from HCT patients had decreased proportion of Bacteroides, Ruminococcaeae, and genes involved in butyrate production, but were enriched for gammaproteobacterial species. No systematic differences in stool microbiome or metabolomic profiles by age, transplant indication, or hospital were noted. Stool metabolites demonstrated strong correlations with microbiome composition. DISCUSSION Stool samples from pediatric allogeneic HCT patients demonstrate substantial dysbiosis early in the transplant course. As microbiome disruptions associate with adverse transplant outcomes, pediatric-specific analyses examining longitudinal microbiome and metabolome changes are imperative to identify causal associations and to inform rational design of interventions.
Collapse
Affiliation(s)
- Caitlin W. Elgarten
- Division of Oncology, Department of Pediatrics, Children’s Hospital of Philadelphia,Center for Pediatric Clinical Effectiveness Research, Children’s Hospital of Philadelphia
| | - Ceylan Tanes
- PennCHOP Microbiome Program, Children’s Hospital of Philadelphia Research Institute
| | - Jung-jin Lee
- PennCHOP Microbiome Program, Children’s Hospital of Philadelphia Research Institute
| | - Lara A. Danziger-Isakov
- Division of Infectious Diseases, Department of Pediatrics, Cincinnati Children’s Hospital, Medical Center and University of Cincinnati
| | - Michael S. Grimley
- Division of Bone Marrow Transplantation and Immune Deficiency, Department of Pediatrics, Cincinnati Children’s Hospital
| | - Michael Green
- Division of Infectious Diseases, UPMC Children’s Hospital of Pittsburgh
| | | | - Jessie L. Barnum
- Division of Blood and Marrow Transplantation, UPMC Children’s Hospital of Pittsburgh
| | | | - Jeffery J. Auletta
- Division of Infectious Diseases, Nationwide Children’s Hospital,Division of Hematology/Oncology/BMT, Nationwide Children’s Hospital,National Marrow Donor Program/Be The Match
| | - Jesse Blumenstock
- Center for Pediatric Clinical Effectiveness Research, Children’s Hospital of Philadelphia
| | - Alix E. Seif
- Division of Oncology, Department of Pediatrics, Children’s Hospital of Philadelphia,Center for Pediatric Clinical Effectiveness Research, Children’s Hospital of Philadelphia
| | - Kyle L. Bittinger
- PennCHOP Microbiome Program, Children’s Hospital of Philadelphia Research Institute,Division of Gastroenterology, Department of Pediatrics, Children’s Hospital of Philadelphia
| | - Brian T. Fisher
- Center for Pediatric Clinical Effectiveness Research, Children’s Hospital of Philadelphia,Division of Infectious Diseases, Department of Pediatrics, Children’s Hospital of Philadelphia
| |
Collapse
|
28
|
Alexander T, Snowden JA, Burman J, Chang HD, Del Papa N, Farge D, Lindsay JO, Malard F, Muraro PA, Nitti R, Salas A, Sharrack B, Mohty M, Greco R. Intestinal Microbiome in Hematopoietic Stem Cell Transplantation For Autoimmune Diseases: Considerations and Perspectives on Behalf of Autoimmune Diseases Working Party (ADWP) of the EBMT. Front Oncol 2021; 11:722436. [PMID: 34745944 PMCID: PMC8569851 DOI: 10.3389/fonc.2021.722436] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 10/07/2021] [Indexed: 12/12/2022] Open
Abstract
Over the past decades, hematopoietic stem cell transplantation (HSCT) has been evolving as specific treatment for patients with severe and refractory autoimmune diseases (ADs), where mechanistic studies have provided evidence for a profound immune renewal facilitating the observed beneficial responses. The intestinal microbiome plays an important role in host physiology including shaping the immune repertoire. The relationships between intestinal microbiota composition and outcomes after HSCT for hematologic diseases have been identified, particularly for predicting the mortality from infectious and non-infectious causes. Furthermore, therapeutic manipulations of the gut microbiota, such as fecal microbiota transplant (FMT), have emerged as promising therapeutic approaches for restoring the functional and anatomical integrity of the intestinal microbiota post-transplantation. Although changes in the intestinal microbiome have been linked to various ADs, studies investigating the effect of intestinal dysbiosis on HSCT outcomes for ADs are scarce and require further attention. Herein, we describe some of the landmark microbiome studies in HSCT recipients and patients with chronic ADs, and discuss the challenges and opportunities of microbiome research for diagnostic and therapeutic purposes in the context of HSCT for ADs.
Collapse
Affiliation(s)
- Tobias Alexander
- Department of Rheumatology and Clinical Immunology - Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and the Berlin Institute of Health (BIH), Berlin, Germany.,Deutsches Rheuma-Forschungszentrum (DRFZ Berlin) - a Leibniz Institute, Berlin, Germany
| | - John A Snowden
- Department of Haematology, Sheffield Teaching Hospitals Foundation NHS Trust, Sheffield, United Kingdom.,Department of Oncology and Metabolism, University of Sheffield, Sheffield, United Kingdom
| | - Joachim Burman
- Department of Neuroscience, Uppsala University, Uppsala, Sweden
| | - Hyun-Dong Chang
- Deutsches Rheuma-Forschungszentrum (DRFZ Berlin) - a Leibniz Institute, Berlin, Germany.,Institute of Biotechnology, Technische Universität Berlin, Berlin, Germany
| | | | - Dominique Farge
- Unité de Médecine Interne: (UF 04) CRMR MATHEC, Maladies Auto-Immunes et Thérapie Cellulaire, Paris, France.,Universite de Paris, IRSL, Recherche Clinique Appliquee `à l'´hématologie, Paris, France.,Department of Medicine, McGill University, Montreal, QC, Canada
| | - James O Lindsay
- Centre for Immunobiology, Blizard Institute, Barts and the London School of Medicine, Queen Mary University of London, London, United Kingdom
| | - Florent Malard
- Service d'hématologie Clinique et de Thérapie Cellulaire, Hôpital Saint Antoine, APHP, Sorbonne Université, INSERM UMRs 938, Paris, France
| | - Paolo A Muraro
- Department of Brain Sciences, Imperial College London, London, United Kingdom
| | - Rosamaria Nitti
- Unit of Hematology and Bone Marrow Transplantation, IRCCS San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy
| | | | - Basil Sharrack
- Department of Neuroscience, Sheffield Teaching Hospitals NHS, Foundation Trust, Sheffield, United Kingdom.,NIHR Neurosciences Biomedical Research Centre, University of Sheffield, Sheffield, United Kingdom
| | - Mohamad Mohty
- Service d'hématologie Clinique et de Thérapie Cellulaire, Hôpital Saint Antoine, APHP, Sorbonne Université, INSERM UMRs 938, Paris, France
| | - Raffaella Greco
- Unit of Hematology and Bone Marrow Transplantation, IRCCS San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy
| |
Collapse
|
29
|
Pan P, Atkinson SN, Taylor B, Zhu H, Zhou D, Flejsierowicz P, Wang LS, Morse M, Liu C, Gunsolus IL, Chen X. Retinoic Acid Signaling Modulates Recipient Gut Barrier Integrity and Microbiota After Allogeneic Hematopoietic Stem Cell Transplantation in Mice. Front Immunol 2021; 12:749002. [PMID: 34759928 PMCID: PMC8573259 DOI: 10.3389/fimmu.2021.749002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 09/30/2021] [Indexed: 11/13/2022] Open
Abstract
Graft-versus-host disease (GVHD) remains a major complication after allogeneic hematopoietic stem cell transplantation (HSCT). An impaired intestinal epithelial barrier is an important component of GVHD pathogenesis. However, contributing host factors that modulate mucosal barrier integrity during GVHD are poorly defined. We hypothesized that vitamin A and retinoic acid (RA) exert positive impacts on maintaining intestinal barrier function after HSCT, thus preventing or dampening GVHD severity. Unexpectedly, we found that exogenous RA increased intestinal permeability of recipient mice after allogeneic HSCT. Serum bacterial endotoxin levels were significantly higher in GVHD mice fed a vitamin A-high (VAH) diet compared to those fed a vitamin A-normal (VAN) diet, indicating a more compromised intestinal barrier function. Furthermore, VAH mice showed more severe lung GVHD with increased donor T cell infiltration in this tissue and died significantly faster than VAN recipients. 16S rRNA sequencing of fecal samples revealed significant differences in the diversity and composition of gut microbiota between VAN and VAH transplant recipients. Collectively, we show that retinoic acid signaling may negatively impact intestinal barrier function during GVHD. Mild vitamin A supplementation is associated with increased lung GVHD and more profound gut dysbiosis. Micronutrients such as vitamin A could modulate complications of allogeneic HSCT, which may be mediated by shaping gut microbiota.
Collapse
Affiliation(s)
- Pan Pan
- Division of Hematology & Oncology, Medical College of Wisconsin, Milwaukee, WI, United States
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Samantha N. Atkinson
- Center for Microbiome Research, Medical College of Wisconsin, Milwaukee, WI, United States
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Brian Taylor
- Division of Hematology & Oncology, Medical College of Wisconsin, Milwaukee, WI, United States
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Haojie Zhu
- Division of Hematology & Oncology, Medical College of Wisconsin, Milwaukee, WI, United States
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Dian Zhou
- Division of Hematology & Oncology, Medical College of Wisconsin, Milwaukee, WI, United States
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Philip Flejsierowicz
- Division of Hematology & Oncology, Medical College of Wisconsin, Milwaukee, WI, United States
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Li-Shu Wang
- Division of Hematology & Oncology, Medical College of Wisconsin, Milwaukee, WI, United States
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Matthew Morse
- Division of Hematology & Oncology, Medical College of Wisconsin, Milwaukee, WI, United States
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Chen Liu
- Department of Pathology, Yale University School of Medicine, New Haven, CT, United States
| | - Ian L. Gunsolus
- Department of Pathology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Xiao Chen
- Division of Hematology & Oncology, Medical College of Wisconsin, Milwaukee, WI, United States
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
| |
Collapse
|
30
|
Zhou X, Moore BB. Experimental Models of Infectious Pulmonary Complications Following Hematopoietic Cell Transplantation. Front Immunol 2021; 12:718603. [PMID: 34484223 PMCID: PMC8415416 DOI: 10.3389/fimmu.2021.718603] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 07/26/2021] [Indexed: 12/23/2022] Open
Abstract
Pulmonary infections remain a major cause of morbidity and mortality in hematopoietic cell transplantation (HCT) recipients. The prevalence and type of infection changes over time and is influenced by the course of immune reconstitution post-transplant. The interaction between pathogens and host immune responses is complex in HCT settings, since the conditioning regimens create periods of neutropenia and immunosuppressive drugs are often needed to prevent graft rejection and limit graft-versus-host disease (GVHD). Experimental murine models of transplantation are valuable tools for dissecting the procedure-related alterations to innate and adaptive immunity. Here we review mouse models of post-HCT infectious pulmonary complications, primarily focused on three groups of pathogens that frequently infect HCT recipients: bacteria (often P. aeruginosa), fungus (primarily Aspergillus fumigatus), and viruses (primarily herpesviruses). These mouse models have advanced our knowledge regarding how the conditioning and HCT process negatively impacts innate immunity and have provided new potential strategies of managing the infections. Studies using mouse models have also validated clinical observations suggesting that prior or occult infections are a potential etiology of noninfectious pulmonary complications post-HCT as well.
Collapse
Affiliation(s)
- Xiaofeng Zhou
- Dept. of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, United States.,Division of Pulmonary and Critical Care Medicine, Dept. of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Bethany B Moore
- Dept. of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, United States.,Division of Pulmonary and Critical Care Medicine, Dept. of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, United States
| |
Collapse
|
31
|
High Throughput Analysis Reveals Changes in Gut Microbiota and Specific Fecal Metabolomic Signature in Hematopoietic Stem Cell Transplant Patients. Microorganisms 2021; 9:microorganisms9091845. [PMID: 34576740 PMCID: PMC8469814 DOI: 10.3390/microorganisms9091845] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 08/02/2021] [Accepted: 08/23/2021] [Indexed: 12/29/2022] Open
Abstract
There is mounting evidence for the emerging role of gut microbiota (GM) and its metabolites in profoundly impacting allogenic hematopoietic stem cell transplantation (allo-HSCT) and its subsequent complications, mainly infections and graft versus host-disease (GvHD). The present study was performed in order to investigate changes in GM composition and fecal metabolic signature between transplant patients (n = 15) and healthy controls (n = 18). The intestinal microbiota was characterized by NGS and gas chromatography-mass spectrometry was employed to perform untargeted analysis of fecal metabolites. We found lower relative abundances of Actinobacteria, Firmicutes, and Bacteroidetes and a higher abundance of Proteobacteria phylum after allo-HSCT. Particularly, the GvHD microbiota was characterized by a lower relative abundance of the short-chain fatty acid-producing bacteria, namely, the Feacalibacterium, Akkermansia, and Veillonella genera and the Lachnospiraceae family, and an enrichment in multidrug-resistant bacteria belonging to Escherichia, Shigella, and Bacteroides. Moreover, network analysis showed that GvHD was linked to a higher number of positive interactions of Blautia and a significant mutual-exclusion rate of Citrobacter. The fecal metabolome was dominated by lipids in the transplant group when compared with the healthy individuals (p < 0.05). Overall, 76 metabolites were significantly altered within transplant recipients, of which 24 were selected as potential biomarkers. Furthermore, the most notable altered metabolic pathways included the TCA cycle; butanoate, propanoate, and pyruvate metabolisms; steroid biosynthesis; and glycolysis/gluconeogenesis. Specific biomarkers and altered metabolic pathways were correlated to GvHD onset. Our results showed significant shifts in gut microbiota structure and fecal metabolites characterizing allo-HSCT.
Collapse
|
32
|
Pulmonary Complications of Pediatric Hematopoietic Cell Transplantation. A National Institutes of Health Workshop Summary. Ann Am Thorac Soc 2021; 18:381-394. [PMID: 33058742 DOI: 10.1513/annalsats.202001-006ot] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Approximately 2,500 pediatric hematopoietic cell transplants (HCTs), most of which are allogeneic, are performed annually in the United States for life-threatening malignant and nonmalignant conditions. Although HCT is undertaken with curative intent, post-HCT complications limit successful outcomes, with pulmonary dysfunction representing the leading cause of nonrelapse mortality. To better understand, predict, prevent, and/or treat pulmonary complications after HCT, a multidisciplinary group of 33 experts met in a 2-day National Institutes of Health Workshop to identify knowledge gaps and research strategies most likely to improve outcomes. This summary of Workshop deliberations outlines the consensus focus areas for future research.
Collapse
|
33
|
Van Lier YF, Van den Brink MRM, Hazenberg MD, Markey KA. The post-hematopoietic cell transplantation microbiome: relationships with transplant outcome and potential therapeutic targets. Haematologica 2021; 106:2042-2053. [PMID: 33882637 PMCID: PMC8327718 DOI: 10.3324/haematol.2020.270835] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Indexed: 01/16/2023] Open
Abstract
Microbiota injury occurs in many patients undergoing allogeneic hematopoietic cell transplantation, likely as a consequence of conditioning regimens involving chemo- and radiotherapy, the widespread use of both prophylactic and therapeutic antibiotics, and profound dietary changes during the peri-transplant period. Peri-transplant dysbiosis is characterized by a decrease in bacterial diversity, loss of commensal bacteria and single-taxon domination (e.g., with Enterococcal strains). Clinically, deviation of the post-transplant microbiota from a normal, high-diversity, healthy state has been associated with increased risk of bacteremia, development of graft-versus-host disease and decreases in overall survival. A number of recent clinical trials have attempted to target the microbiota in allogeneic hematopoietic cell transplantation patients via dietary interventions, selection of therapeutic antibiotics, administration of pre- or pro-biotics, or by performing fecal microbiota transplantation. These strategies have yielded promising results but the mechanisms by which these interventions influence transplant-related complications remain largely unknown. In this review we summarize the current approaches to targeting the microbiota, discuss potential underlying mechanisms and highlight the key outstanding areas that require further investigation in order to advance microbiota- targeting therapies.
Collapse
Affiliation(s)
- Yannouck F Van Lier
- Department of Hematology, Amsterdam UMC, Amsterdam, the Netherlands; Department of Experimental Immunology, Amsterdam Institute for Infection and Immunity (AII), Cancer Center Amsterdam, Amsterdam UMC, Amsterdam
| | - Marcel R M Van den Brink
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Weill Cornell Medical College, New York, NY
| | - Mette D Hazenberg
- Department of Hematology, Amsterdam UMC, Amsterdam, the Netherlands; Department of Experimental Immunology, Amsterdam Institute for Infection and Immunity (AII), Cancer Center Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands; Department of Hematopoiesis, Sanquin Research, Amsterdam
| | - Kate A Markey
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Weill Cornell Medical College, New York, NY.
| |
Collapse
|
34
|
Kumari R, Palaniyandi S, Hildebrandt GC. The microbiome-the revealing of a long time unbeknownst factor for outcome in murine models of graft-versus-host disease. Bone Marrow Transplant 2021; 56:1777-1783. [PMID: 34052837 DOI: 10.1038/s41409-021-01325-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 04/13/2021] [Accepted: 04/21/2021] [Indexed: 02/04/2023]
Affiliation(s)
- Reena Kumari
- Division of Hematology & Blood and Marrow Transplantation, Markey Cancer Center, University of Kentucky, Lexington, KY, USA
| | - Senthilnathan Palaniyandi
- Division of Hematology & Blood and Marrow Transplantation, Markey Cancer Center, University of Kentucky, Lexington, KY, USA
| | - Gerhard Carl Hildebrandt
- Division of Hematology & Blood and Marrow Transplantation, Markey Cancer Center, University of Kentucky, Lexington, KY, USA.
| |
Collapse
|
35
|
Wei LQ, Cheong IH, Yang GH, Li XG, Kozlakidis Z, Ding L, Liu NN, Wang H. The Application of High-Throughput Technologies for the Study of Microbiome and Cancer. Front Genet 2021; 12:699793. [PMID: 34394190 PMCID: PMC8355622 DOI: 10.3389/fgene.2021.699793] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Accepted: 06/21/2021] [Indexed: 12/14/2022] Open
Abstract
Human gut microbiome research, especially gut microbiome, has been developing at a considerable pace over the last decades, driven by a rapid technological advancement. The emergence of high-throughput technologies, such as genomics, transcriptomics, and others, has afforded the generation of large volumes of data, and in relation to specific pathologies such as different cancer types. The current review identifies high-throughput technologies as they have been implemented in the study of microbiome and cancer. Four main thematic areas have emerged: the characterization of microbial diversity and composition, microbial functional analyses, biomarker prediction, and, lastly, potential therapeutic applications. The majority of studies identified focus on the microbiome diversity characterization, which is reaching technological maturity, while the remaining three thematic areas could be described as emerging.
Collapse
Affiliation(s)
- Lu Qi Wei
- State Key Laboratory of Oncogenes and Related Genes, Centre for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Io Hong Cheong
- State Key Laboratory of Oncogenes and Related Genes, Centre for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guang Huan Yang
- State Key Laboratory of Oncogenes and Related Genes, Centre for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiao Guang Li
- State Key Laboratory of Oncogenes and Related Genes, Centre for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zisis Kozlakidis
- International Agency for Research on Cancer, World Health Organization, Lyon, France
| | - Lei Ding
- State Key Laboratory of Oncogenes and Related Genes, Centre for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ning Ning Liu
- State Key Laboratory of Oncogenes and Related Genes, Centre for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hui Wang
- State Key Laboratory of Oncogenes and Related Genes, Centre for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
36
|
Pharyngeal Microbial Signatures Are Predictive of the Risk of Fungal Pneumonia in Hematologic Patients. Infect Immun 2021; 89:e0010521. [PMID: 33782152 DOI: 10.1128/iai.00105-21] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The ability to predict invasive fungal infections (IFI) in patients with hematological malignancies is fundamental for successful therapy. Although gut dysbiosis is known to occur in hematological patients, whether airway dysbiosis also contributes to the risk of IFI has not been investigated. Nasal and oropharyngeal swabs were collected for functional microbiota characterization in 173 patients with hematological malignancies recruited in a multicenter, prospective, observational study and stratified according to the risk of developing IFI. A lower microbial richness and evenness were found in the pharyngeal microbiota of high-risk patients that were associated with a distinct taxonomic and metabolic profile. A murine model of IFI provided biologic plausibility for the finding that loss of protective anaerobes, such as Clostridiales and Bacteroidetes, along with an apparent restricted availability of tryptophan, is causally linked to the risk of IFI in hematologic patients and indicates avenues for antimicrobial stewardship and metabolic reequilibrium in IFI.
Collapse
|
37
|
An infectious diseases perspective on the microbiome and allogeneic stem cell transplant. Curr Opin Infect Dis 2021; 33:426-432. [PMID: 33148984 DOI: 10.1097/qco.0000000000000683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
PURPOSE OF REVIEW The gut microbiome presents a novel source of diagnostic and therapeutic potential to modify post allogeneic stem cell transplant complications. There is an explosion of interest in microbiome research, mostly in the form of single-centre prospective time-series cohorts utilizing a variety of sampling frequencies and metagenomic technologies to sequence the microbiome. The purpose of this review is to summarize important recent publications and contextualize them within what has already been described in this rapidly growing field. RECENT FINDING Results from observational human cohort and animal transplant models add to the growing body of evidence that the microbiome modulates the immunopathogenesis of posttransplant complications. This is particularly the case for recipients of grafts replete with T cells where the evidence that acute graft-versus-host disease is mediated by anaerobic commensal-associated short-chain fatty acids, which interact with mucosa-associated (CD4FOXP3) T-regulatory cells. SUMMARY Future human research into the role of the microbiome in allogeneic stem transplant should incorporate rigorous and considered experimental design in addition to next-generation sequencing technology to better portray microbiome functional potential and active gene expression. In combination with host immune phenotyping, which would facilitate a robust understanding of the host--microbiome interaction that is required before meaningful translation into clinical diagnostics and therapeutics can be expected.
Collapse
|
38
|
Ma T, Chen Y, Li LJ, Zhang LS. Opportunities and Challenges for Gut Microbiota in Acute Leukemia. Front Oncol 2021; 11:692951. [PMID: 34307157 PMCID: PMC8293295 DOI: 10.3389/fonc.2021.692951] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 06/23/2021] [Indexed: 12/12/2022] Open
Abstract
Acute leukemia (AL) is a highly heterogeneous hematologic malignancy, and although great progress has been made in the treatment of AL with allogeneic hematopoietic stem cell transplantation (Allo-HSCT) and new targeted drugs, problems such as infection and GVHD in AL treatment are still serious. How to reduce the incidence of AL, improve its prognosis and reduce the side effects of treatment is a crucial issue. The gut microbiota plays an important role in regulating disease progression, pathogen colonization, and immune responses. This article reviews recent advances in the gut microbiota and AL pathogenesis, infection, treatment and its role in allo-HSCT.
Collapse
Affiliation(s)
- Tao Ma
- Department of Hematology, Lanzhou University Second Hospital, Lanzhou, China.,Department of Hematology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Yan Chen
- Department of Hematology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Li-Juan Li
- Department of Hematology, Lanzhou University Second Hospital, Lanzhou, China
| | - Lian-Sheng Zhang
- Department of Hematology, Lanzhou University Second Hospital, Lanzhou, China
| |
Collapse
|
39
|
Hérivaux A, Willis JR, Mercier T, Lagrou K, Gonçalves SM, Gonçales RA, Maertens J, Carvalho A, Gabaldón T, Cunha C. Lung microbiota predict invasive pulmonary aspergillosis and its outcome in immunocompromised patients. Thorax 2021; 77:283-291. [PMID: 34172558 PMCID: PMC8867272 DOI: 10.1136/thoraxjnl-2020-216179] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 05/26/2021] [Indexed: 11/17/2022]
Abstract
Rationale Recent studies have revealed that the lung microbiota of critically ill patients is altered and predicts clinical outcomes. The incidence of invasive fungal infections, namely, invasive pulmonary aspergillosis (IPA), in immunocompromised patients is increasing, but the clinical significance of variations in lung bacterial communities is unknown. Objectives To define the contribution of the lung microbiota to the development and course of IPA. Methods and measurements We performed an observational cohort study to characterise the lung microbiota in 104 immunocompromised patients using bacterial 16S ribosomal RNA gene sequencing on bronchoalveolar lavage samples sampled on clinical suspicion of infection. Associations between lung dysbiosis in IPA and pulmonary immunity were evaluated by quantifying alveolar cytokines and chemokines and immune cells. The contribution of microbial signatures to patient outcome was assessed by estimating overall survival. Main results Patients diagnosed with IPA displayed a decreased alpha diversity, driven by a markedly increased abundance of the Staphylococcus, Escherichia, Paraclostridium and Finegoldia genera and a decreased proportion of the Prevotella and Veillonella genera. The overall composition of the lung microbiome was influenced by the neutrophil counts and associated with differential levels of alveolar cytokines. Importantly, the degree of bacterial diversity at the onset of IPA predicted the survival of infected patients. Conclusions Our results reveal the lung microbiota as an understudied source of clinical variation in patients at risk of IPA and highlight its potential as a diagnostic and therapeutic target in the context of respiratory fungal diseases.
Collapse
Affiliation(s)
- Anaïs Hérivaux
- Life and Health Sciences Research Institute (ICVS), University of Minho, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Guimarães/Braga, Portugal
| | - Jesse R Willis
- Barcelona Supercomputing Centre (BSC-CNS), Barcelona, Spain.,Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Toine Mercier
- Department of Hematology, University Hospitals Leuven, Leuven, Belgium.,Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
| | - Katrien Lagrou
- Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium.,Clinical Department of Laboratory Medicine, University Hospitals Leuven, Leuven, Belgium
| | - Samuel M Gonçalves
- Life and Health Sciences Research Institute (ICVS), University of Minho, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Guimarães/Braga, Portugal
| | - Relber A Gonçales
- Life and Health Sciences Research Institute (ICVS), University of Minho, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Guimarães/Braga, Portugal
| | - Johan Maertens
- Department of Hematology, University Hospitals Leuven, Leuven, Belgium.,Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
| | - Agostinho Carvalho
- Life and Health Sciences Research Institute (ICVS), University of Minho, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Guimarães/Braga, Portugal
| | - Toni Gabaldón
- Barcelona Supercomputing Centre (BSC-CNS), Barcelona, Spain .,Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain.,Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| | - Cristina Cunha
- Life and Health Sciences Research Institute (ICVS), University of Minho, Braga, Portugal .,ICVS/3B's - PT Government Associate Laboratory, Guimarães/Braga, Portugal
| |
Collapse
|
40
|
Shi Z, Zhang M. Emerging Roles for the Gut Microbiome in Lymphoid Neoplasms. CLINICAL MEDICINE INSIGHTS-ONCOLOGY 2021; 15:11795549211024197. [PMID: 34211309 PMCID: PMC8216388 DOI: 10.1177/11795549211024197] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Accepted: 05/18/2021] [Indexed: 12/15/2022]
Abstract
Lymphoid neoplasms encompass a heterogeneous group of malignancies with a predilection for immunocompromised individuals, and the disease burden of lymphoid neoplasms has been rising globally over the last decade. At the same time, mounting studies delineated a crucial role of the gut microbiome in the aetiopathogenesis of various diseases. Orchestrated interactions between myriad microorganisms and the gastrointestinal mucosa establish a defensive barrier for a range of physiological processes, especially immunity and metabolism. These findings provide new perspectives to harness our knowledge of the gut microbiota for preclinical and clinical studies of lymphoma. Here, we review recent findings that support a role for the gut microbiota in the development of lymphoid neoplasms and pinpoint relevant molecular mechanisms. Accordingly, we propose the microbiota-gut-lymphoma axis as a promising target for clinical translation, including auxiliary diagnosis, novel prevention and treatment strategies, and predicting clinical outcomes and treatment-related adverse effects of the disease in the future. This review will reveal a fascinating avenue of research in the microbiota-mediated lymphoma field.
Collapse
Affiliation(s)
- Zhuangzhuang Shi
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Lymphoma Diagnosis and Treatment Centre of Henan Province, Zhengzhou, China
| | - Mingzhi Zhang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Lymphoma Diagnosis and Treatment Centre of Henan Province, Zhengzhou, China
| |
Collapse
|
41
|
Classen AY, Henze L, von Lilienfeld-Toal M, Maschmeyer G, Sandherr M, Graeff LD, Alakel N, Christopeit M, Krause SW, Mayer K, Neumann S, Cornely OA, Penack O, Weißinger F, Wolf HH, Vehreschild JJ. Primary prophylaxis of bacterial infections and Pneumocystis jirovecii pneumonia in patients with hematologic malignancies and solid tumors: 2020 updated guidelines of the Infectious Diseases Working Party of the German Society of Hematology and Medical Oncology (AGIHO/DGHO). Ann Hematol 2021; 100:1603-1620. [PMID: 33846857 PMCID: PMC8116237 DOI: 10.1007/s00277-021-04452-9] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 02/04/2021] [Indexed: 12/11/2022]
Abstract
Hematologic and oncologic patients with chemo- or immunotherapy-related immunosuppression are at substantial risk for bacterial infections and Pneumocystis jirovecii pneumonia (PcP). As bacterial resistances are increasing worldwide and new research reshapes our understanding of the interactions between the human host and bacterial commensals, administration of antibacterial prophylaxis has become a matter of discussion. This guideline constitutes an update of the 2013 published guideline of the Infectious Diseases Working Party (AGIHO) of the German Society for Hematology and Medical Oncology (DGHO). It gives an overview about current strategies for antibacterial prophylaxis in cancer patients while taking into account the impact of antibacterial prophylaxis on the human microbiome and resistance development. Current literature published from January 2012 to August 2020 was searched and evidence-based recommendations were developed by an expert panel. All recommendations were discussed and approved in a consensus conference of the AGIHO prior to publication. As a result, we present a comprehensive update and extension of our guideline for antibacterial and PcP prophylaxis in cancer patients.
Collapse
Affiliation(s)
- Annika Y Classen
- Faculty of Medicine and University Hospital Cologne, Department I for Internal Medicine, University of Cologne, Herderstr. 52-54, 50931, Cologne, Germany
- German Centre for Infection Research (DZIF), partner site Bonn-Cologne, Cologne, Germany
| | - Larissa Henze
- Department of Medicine, Clinic III - Hematology, Oncology, Palliative Medicine, Rostock University Medical Center, Rostock, Germany
| | - Marie von Lilienfeld-Toal
- Department of Hematology and Oncology, Clinic for Internal Medicine II, University Hospital Jena, Jena, Germany
| | - Georg Maschmeyer
- Hematology, Oncology and Palliative Care, Klinikum Ernst von Bergmann, Potsdam, Germany
| | - Michael Sandherr
- Specialist Clinic for Haematology and Oncology, Medical Care Center Penzberg, Penzberg, Germany
| | - Luisa Durán Graeff
- Faculty of Medicine and University Hospital Cologne, Department I for Internal Medicine, University of Cologne, Herderstr. 52-54, 50931, Cologne, Germany
- German Centre for Infection Research (DZIF), partner site Bonn-Cologne, Cologne, Germany
| | - Nael Alakel
- Department I of Internal Medicine, Hematology and Oncology, University Hospital Dresden, Dresden, Germany
| | - Maximilian Christopeit
- Department of Internal Medicine II, Hematology, Oncology, Clinical Immunology and Rheumatology, University Hospital Tübingen, Tübingen, Germany
| | - Stefan W Krause
- Department of Medicine 5 - Hematology and Oncology, University Hospital Erlangen, Erlangen, Germany
| | - Karin Mayer
- Medical Clinic III for Oncology, Hematology, Immunooncology and Rheumatology, University Hospital Bonn (UKB), Bonn, Germany
| | - Silke Neumann
- Interdisciplinary Center for Oncology, Wolfsburg, Germany
| | - Oliver A Cornely
- Faculty of Medicine and University Hospital Cologne, Department I for Internal Medicine, University of Cologne, Herderstr. 52-54, 50931, Cologne, Germany
- German Centre for Infection Research (DZIF), partner site Bonn-Cologne, Cologne, Germany
- Faculty of Medicine and University Hospital Cologne, Chair Translational Research, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Clinical Trials Centre Cologne (ZKS Köln), University of Cologne, Cologne, Germany
| | - Olaf Penack
- Medical Department for Hematology, Oncology and Tumor Immunology, Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin, Germany
| | - Florian Weißinger
- Department for Internal Medicine, Hematology/Oncology, and Palliative Care, Evangelisches Klinikum Bethel v. Bodelschwinghsche Stiftungen Bethel, Bielefeld, Germany
| | - Hans-Heinrich Wolf
- Department IV of Internal Medicine, University Hospital Halle, Halle, Germany
| | - Jörg Janne Vehreschild
- Faculty of Medicine and University Hospital Cologne, Department I for Internal Medicine, University of Cologne, Herderstr. 52-54, 50931, Cologne, Germany.
- German Centre for Infection Research (DZIF), partner site Bonn-Cologne, Cologne, Germany.
- Department of Internal Medicine, Hematology/Oncology, Goethe University Frankfurt, Frankfurt am Main, Germany.
| |
Collapse
|
42
|
Yuzefpolskaya M, Lumish HS, Javaid A, Cagliostro B, Mondellini GM, Bohn B, Sweat A, Onat D, Braghieri L, Takeda K, Naka Y, Sayer GT, Uriel N, Aaron JG, Montassier E, Demmer RT, Colombo PC. Association of preoperative infections, nasal Staphylococcus aureus colonization and gut microbiota with left ventricular assist device outcomes. Eur J Heart Fail 2021; 23:1404-1415. [PMID: 33964186 DOI: 10.1002/ejhf.2215] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 03/29/2021] [Accepted: 05/04/2021] [Indexed: 01/05/2023] Open
Abstract
AIMS Infections are common following left ventricular assist device (LVAD) implantation and predict adverse events. Infections are frequent prior to LVAD implantation although their impact on postoperative outcomes remains unknown. Gut and nasal microbial imbalance may predispose to mucosal colonization with pathogens. Herein, we investigated the predictive role of pre-LVAD infections, and explored the association of nasal Staphylococcus aureus (SA) colonization and gut microbiota, on postoperative outcomes. METHODS AND RESULTS Overall, 254 LVAD patients were retrospectively categorized based on pre-LVAD infection status: Group 1, bacterial/fungal bloodstream infection (BSI); Group 2, other bacterial/fungal; Group 3, viral; and Group 4, no infection. In a subset of patients, nasal SA colonization (n = 140) and pre-LVAD stool (n = 25) were analysed using 16S rRNA sequencing. A total of 75 (29%) patients had a pre-LVAD infection [Group 1: 22 (29%); Group 2: 41 (55%); Group 3: 12 (16%)]. Pre-LVAD BSIs were independent predictors of 1-year postoperative mortality and infections [Group 1 vs. 4: hazard ratio (HR) 2.70, P = 0.036 vs. HR 1.8, P = 0.046]. In an unadjusted analysis, pre-LVAD infections other than BSIs, INTERMACS profile ≤2, higher serum creatinine, lower serum albumin and nasal SA colonization were also significantly associated with postoperative infections. Patients with early post-LVAD infections exhibited decreased microbial diversity (P < 0.05). CONCLUSIONS Pre-LVAD infections are common. BSIs independently predict postoperative mortality and infections. Additional studies are needed to confirm our findings that pre-LVAD SA nasal colonization and gut microbial composition can help stratify patients' risk for infectious complications after LVAD implantation.
Collapse
Affiliation(s)
- Melana Yuzefpolskaya
- Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Heidi S Lumish
- Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Azka Javaid
- Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Barbara Cagliostro
- Division of Cardiac Surgery, Department of Surgery, Columbia University Irving Medical Center, New York, NY, USA
| | - Giulio M Mondellini
- Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Bruno Bohn
- Division of Epidemiology and Community Health, University of Minnesota, Minneapolis, MN, USA
| | - Austin Sweat
- Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Duygu Onat
- Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Lorenzo Braghieri
- Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Koji Takeda
- Division of Epidemiology and Community Health, University of Minnesota, Minneapolis, MN, USA
| | - Yoshifumi Naka
- Division of Epidemiology and Community Health, University of Minnesota, Minneapolis, MN, USA
| | - Gabriel T Sayer
- Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Nir Uriel
- Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Justin G Aaron
- Division of Infectious Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Emmanuel Montassier
- Department of Computer Science and Engineering, University of Minnesota, Minneapolis, MN, USA.,Université de Nantes, Microbiotas Hosts Antibiotics and Bacterial Resistances (MiHAR), and Department of Emergency Medicine, CHU de Nantes, Nantes, France
| | - Ryan T Demmer
- Division of Epidemiology and Community Health, University of Minnesota, Minneapolis, MN, USA.,Department of Epidemiology, Mailman School of Public Health, Columbia University Irving Medical Center, New York, NY, USA
| | - Paolo C Colombo
- Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| |
Collapse
|
43
|
Zinter MS, Lindemans CA, Versluys BA, Mayday MY, Sunshine S, Reyes G, Sirota M, Sapru A, Matthay MA, Kharbanda S, Dvorak CC, Boelens JJ, DeRisi JL. The pulmonary metatranscriptome prior to pediatric HCT identifies post-HCT lung injury. Blood 2021; 137:1679-1689. [PMID: 33512420 PMCID: PMC7995292 DOI: 10.1182/blood.2020009246] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 12/15/2020] [Indexed: 02/07/2023] Open
Abstract
Lung injury after pediatric allogeneic hematopoietic cell transplantation (HCT) is a common and disastrous complication that threatens long-term survival. To develop strategies to prevent lung injury, novel tools are needed to comprehensively assess lung health in HCT candidates. Therefore, this study analyzed biospecimens from 181 pediatric HCT candidates who underwent routine pre-HCT bronchoalveolar lavage (BAL) at the University Medical Center Utrecht between 2005 and 2016. BAL fluid underwent metatranscriptomic sequencing of microbial and human RNA, and unsupervised clustering and generalized linear models were used to associate microbiome gene expression data with the development of post-HCT lung injury. Microbe-gene correlations were validated using a geographically distinct cohort of 18 pediatric HCT candidates. The cumulative incidence of post-HCT lung injury varied significantly according to 4 pre-HCT pulmonary metatranscriptome clusters, with the highest incidence observed in children with pre-HCT viral enrichment and innate immune activation, as well as in children with profound microbial depletion and concomitant natural killer/T-cell activation (P < .001). In contrast, children with pre-HCT pulmonary metatranscriptomes containing diverse oropharyngeal taxa and lacking inflammation rarely developed post-HCT lung injury. In addition, activation of epithelial-epidermal differentiation, mucus production, and cellular adhesion were associated with fatal post-HCT lung injury. In a separate validation cohort, associations among pulmonary respiratory viral load, oropharyngeal taxa, and pulmonary gene expression were recapitulated; the association with post-HCT lung injury needs to be validated in an independent cohort. This analysis suggests that assessment of the pre-HCT BAL fluid may identify high-risk pediatric HCT candidates who may benefit from pathobiology-targeted interventions.
Collapse
Affiliation(s)
- Matt S Zinter
- Division of Critical Care Medicine and
- Division of Allergy, Immunology, and Bone Marrow Transplantation, Department of Pediatrics, School of Medicine, University of California, San Francisco, CA
| | - Caroline A Lindemans
- Department of Pediatric Stem Cell Transplantation, University Medical Center Utrecht, Utrecht, The Netherlands
- Department of Hematopoietic Cell Transplantation, Princess Maxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Birgitta A Versluys
- Department of Pediatric Stem Cell Transplantation, University Medical Center Utrecht, Utrecht, The Netherlands
- Department of Hematopoietic Cell Transplantation, Princess Maxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Madeline Y Mayday
- Graduate Program in Experimental Pathology, and Yale Stem Cell Center, Department of Pathology, Yale University, New Haven, CT
| | - Sara Sunshine
- Department of Biochemistry and Biophysics, School of Medicine
| | | | - Marina Sirota
- Bakar Computational Health Sciences Institute, and
- Department of Pediatrics, School of Medicine, University of California, San Francisco, CA
| | - Anil Sapru
- Division of Critical Care Medicine, Department of Pediatrics, School of Medicine, University of California, Los Angeles, CA
| | - Michael A Matthay
- Department of Medicine and
- Department of Anesthesiology, Cardiovascular Research Institute, School of Medicine, University of California, San Francisco, CA
| | - Sandhya Kharbanda
- Division of Allergy, Immunology, and Bone Marrow Transplantation, Department of Pediatrics, School of Medicine, University of California, San Francisco, CA
| | - Christopher C Dvorak
- Division of Allergy, Immunology, and Bone Marrow Transplantation, Department of Pediatrics, School of Medicine, University of California, San Francisco, CA
| | - Jaap J Boelens
- Department of Pediatric Stem Cell Transplantation and Cellular Therapies, Memorial Sloan Kettering Cancer Center, New York, NY; and
| | - Joseph L DeRisi
- Department of Biochemistry and Biophysics, School of Medicine
- Chan Zuckerberg Biohub, San Francisco, CA
| |
Collapse
|
44
|
Chang CC, Hayase E, Jenq RR. The role of microbiota in allogeneic hematopoietic stem cell transplantation. Expert Opin Biol Ther 2021; 21:1121-1131. [PMID: 33412949 DOI: 10.1080/14712598.2021.1872541] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Allogeneic hematopoietic stem cell transplantation (Allo-HSCT) is commonly performed to treat a variety of benign and malignant hematological diseases. Acute graft-versus-host disease (GVHD) is a major life-threatening complication that often occurs following allo-HSCT. Recently, improvements in methods to characterize the microbiota have led to a greater appreciation for how frequently and profoundly an alteration in microbial composition, or dysbiosis, can occur in allo-HSCT recipients to better decipher the complex interplay between microbiota and allo-HSCT outcomes. AREAS COVERED This article reviews the current knowledge of the microbiota's impact on allo-HSCT outcomes, including effects of microbiota-derived metabolites, and crosstalk between commensals and the allogeneic immune response. This article also summarizes the effects of HSCT and transplant-related procedures on microbiota, and recent developments in interventional strategies. EXPERT OPINION A growing body of literature indicates that the composition of the intestinal microbiota can function as a predictive biomarker for the risk and severity of acute GVHD, as well as overall survival, in patients undergoing allo-HSCT. Mechanisms underpinning these associations, however, are not well understood, and clinical strategies that modulate the microbiome to improve outcomes have yet to be fully developed. There is an unmet need to determine mechanisms to improve the efficacy of allo-HSCT.
Collapse
Affiliation(s)
- Chia-Chi Chang
- Department of Genomic Medicine, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Eiko Hayase
- Department of Genomic Medicine, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Robert R Jenq
- Department of Genomic Medicine, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Department of Stem Cell Transplantation and Cellular Therapy, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
45
|
D'Angelo CR, Sudakaran S, Callander NS. Clinical effects and applications of the gut microbiome in hematologic malignancies. Cancer 2020; 127:679-687. [PMID: 33369893 DOI: 10.1002/cncr.33400] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 07/20/2020] [Accepted: 11/22/2020] [Indexed: 12/16/2022]
Abstract
The gut microbiome and its effects on host immunity have exciting implications for cancer prognosis and therapy. Examples in allogeneic hematopoietic stem cell transplantation (allo-SCT) demonstrate the role of the gut microbiome as a biomarker for clinical outcomes, and animal models demonstrate how microbiota manipulation may augment therapeutic responses. There are multiple mechanisms that gut microbiota may have in affecting distant tumor environments, including control of cytokine release, dendritic cell activation, and T-cell lymphocyte stimulation. Recently, there has been a marked interest in understanding interactions between host and microbiome in hematologic malignancies. This review summarizes the current understanding of the gut microbiome and its impact on leukemia, lymphoma, multiple myeloma, and allo-SCT and highlights several broad methods for targeting the gut microbiome in therapeutic trials.
Collapse
Affiliation(s)
- Christopher R D'Angelo
- Division of Hematology/Oncology, Department of Medicine, University of Nebraska Medical Center, Omaha, Nebraska
| | - Sailendharan Sudakaran
- Microbiome Hub, Wisconsin Institute of Discovery, University of Wisconsin, Madison, Wisconsin
| | - Natalie S Callander
- Section of Hematology/Oncology and Bone Marrow Transplantation, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| |
Collapse
|
46
|
Kumari R, Palaniyandi S, Strattan E, Hildebrandt GC. Microbiome: an emerging new frontier in graft‑versus‑host disease. Inflamm Res 2020; 70:1-5. [PMID: 33185704 PMCID: PMC7661320 DOI: 10.1007/s00011-020-01419-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 10/06/2020] [Accepted: 10/26/2020] [Indexed: 11/27/2022] Open
Affiliation(s)
- Reena Kumari
- Division of Hematology & Blood and Marrow Transplantation, Markey Cancer Center, University of Kentucky, Lexington, KY 40536-093 USA
| | - Senthilnathan Palaniyandi
- Division of Hematology & Blood and Marrow Transplantation, Markey Cancer Center, University of Kentucky, Lexington, KY 40536-093 USA
| | - Ethan Strattan
- Division of Hematology & Blood and Marrow Transplantation, Markey Cancer Center, University of Kentucky, Lexington, KY 40536-093 USA
- Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky, Lexington, KY 40536-093 USA
| | - Gerhard C. Hildebrandt
- Division of Hematology & Blood and Marrow Transplantation, Markey Cancer Center, University of Kentucky, Lexington, KY 40536-093 USA
- Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky, Lexington, KY 40536-093 USA
| |
Collapse
|
47
|
Adelman MW, Woodworth MH, Langelier C, Busch LM, Kempker JA, Kraft CS, Martin GS. The gut microbiome's role in the development, maintenance, and outcomes of sepsis. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2020; 24:278. [PMID: 32487252 PMCID: PMC7266132 DOI: 10.1186/s13054-020-02989-1] [Citation(s) in RCA: 181] [Impact Index Per Article: 36.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 05/12/2020] [Indexed: 12/15/2022]
Abstract
The gut microbiome regulates a number of homeostatic mechanisms in the healthy host including immune function and gut barrier protection. Loss of normal gut microbial structure and function has been associated with diseases as diverse as Clostridioides difficile infection, asthma, and epilepsy. Recent evidence has also demonstrated a link between the gut microbiome and sepsis. In this review, we focus on three key areas of the interaction between the gut microbiome and sepsis. First, prior to sepsis onset, gut microbiome alteration increases sepsis susceptibility through several mechanisms, including (a) allowing for expansion of pathogenic intestinal bacteria, (b) priming the immune system for a robust pro-inflammatory response, and (c) decreasing production of beneficial microbial products such as short-chain fatty acids. Second, once sepsis is established, gut microbiome disruption worsens and increases susceptibility to end-organ dysfunction. Third, there is limited evidence that microbiome-based therapeutics, including probiotics and selective digestive decontamination, may decrease sepsis risk and improve sepsis outcomes in select patient populations, but concerns about safety have limited uptake. Case reports of a different microbiome-based therapy, fecal microbiota transplantation, have shown correlation with gut microbial structure restoration and decreased inflammatory response, but these results require further validation. While much of the evidence linking the gut microbiome and sepsis has been established in pre-clinical studies, clinical evidence is lacking in many areas. To address this, we outline a potential research agenda for further investigating the interaction between the gut microbiome and sepsis.
Collapse
Affiliation(s)
- Max W Adelman
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, 49 Jesse Hill Jr. Drive, Atlanta, GA, 30303, USA.
| | - Michael H Woodworth
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, 49 Jesse Hill Jr. Drive, Atlanta, GA, 30303, USA
| | - Charles Langelier
- Division of Infectious Diseases, Department of Medicine, University of California, San Francisco, CA, USA
| | - Lindsay M Busch
- Critical Care Medicine Department, National Institutes of Health Clinical Center, Bethesda, MD, USA
| | - Jordan A Kempker
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Colleen S Kraft
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, 49 Jesse Hill Jr. Drive, Atlanta, GA, 30303, USA.,Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Greg S Martin
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA.,Emory Critical Care Center, Emory Healthcare, Atlanta, GA, USA
| |
Collapse
|
48
|
Gavriilaki M, Sakellari I, Anagnostopoulos A, Gavriilaki E. The Impact of Antibiotic-Mediated Modification of the Intestinal Microbiome on Outcomes of Allogeneic Hematopoietic Cell Transplantation: Systematic Review and Meta-Analysis. Biol Blood Marrow Transplant 2020; 26:1738-1746. [PMID: 32447043 DOI: 10.1016/j.bbmt.2020.05.011] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 05/13/2020] [Indexed: 01/02/2023]
Abstract
Accumulating evidence points toward a protective role of intestinal microbiota diversity in allogeneic hematopoietic cell transplantation (allo-HCT). The purpose of this systematic review and meta-analysis is to determine the effect of antibiotic-mediated disruption of microbiota on main allo-HCT outcomes (graft-versus-host disease [GVHD], treatment-related mortality [TRM], overall survival [OS]). Following literature search, 2 reviewers screened eligible studies and assessed risk of bias (RoB). Meta-analysis was performed using Review Manager Software. Among 443 screened references, 18 were eligible for meta-analysis. In studies with genomic markers, grade II to IV acute GVHD was significantly reduced in patients not receiving gut decontamination (GD) (odds ratio [OR], 1.56; 95% confidence interval [CI], 1.20 to 2.04). In subgroup analysis, prophylaxis with systemic antibiotics conferred an increased risk of acute GVHD (OR, 1.65; 95% CI, 1.08 to 2.53). When we incorporated RoB, we found a positive correlation of intestinal GVHD with GD (OR, 1.77; 95% CI, 1.29 to 2.44). Patients with higher microbiota diversity presented increased OS (risk ratio [RR], 1.58; 95% CI, 1.19 to 2.09) and lower TRM (RR, 0.45; 95% CI, 0.26 to 0.76). Our findings confirm that GD and prophylaxis with systemic antibiotics increase acute and intestinal GVHD. Importantly, our meta-analysis was the first to show a significant effect of microbiota diversity on TRM and OS.
Collapse
Affiliation(s)
- Maria Gavriilaki
- Laboratory of Clinical Neurophysiology, AHEPA Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece.
| | - Ioanna Sakellari
- Hematology Department-BMT Unit, G. Papanicolaou Hospital, Thessaloniki, Greece
| | | | - Eleni Gavriilaki
- Hematology Department-BMT Unit, G. Papanicolaou Hospital, Thessaloniki, Greece
| |
Collapse
|
49
|
Moghnieh R, Tamim H, Abyad A, Jadayel M, Awad L, Abdallah D, Haddad N, Berberi J, Alwazir R, Taher R, Jisr T, Lakkis R, Zahran K, Ibrahim A. Pre-engraftment infectious complications and patient outcomes after allogeneic hematopoietic cell transplantation: a single-center experience from Lebanon. Infection 2020; 48:385-401. [PMID: 32170652 DOI: 10.1007/s15010-020-01407-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Accepted: 03/02/2020] [Indexed: 12/17/2022]
Abstract
BACKGROUND Infectious complications are significant causes of morbidity and mortality after allogeneic hematopoietic cell transplantation (allo-HCT). They occur variably over different periods, with scant data reported from Lebanon and neighboring countries. In this study, we described the pre-engraftment neutropenic phase, febrile episodes, and peri-transplant medical complications in patients undergoing allo-HCT at a tertiary-care hospital. METHODS This is a retrospective chart review of patients who underwent allo-HCT between 2007 and 2016 at Makassed General Hospital in Beirut, Lebanon. Data were extracted from medical records, the HCT registry, and medical laboratory logbooks. RESULTS One hundred and six patients were included, 75% having hematologic malignancies and 13% aplastic anemia. None received antibacterial prophylaxis with fluoroquinolones. Yet from conditioning chemotherapy till the say before HCT, 32% of the patients received broad-spectrum antibiotics (BSA) due to fever or infection. At the day of cell infusion, 41.5% of the patients were on BSA. Neutrophil engraftment failure was recorded in 8% of the patients. The cumulative incidence of pre-engraftment bacteremia and Gram-negative bacteremia was 14.3 and 7.1%, respectively. Aplastic anemia was an independent risk factor for pre-engraftment bacteremia [hazard ratio (HR) = 3.86, 95% confidence interval (CI) (1.29-11.5), P = 0.02]. The cumulative incidence of pre-engraftment pneumonia was 11.2%. Patient age significantly increased the risk of pre-engraftment pneumonia [HR = 12.35, 95% CI (1.27-120.50), P = 0.03]. Six-month post-transplant mortality reached 17% in our cohort. Myelodysplastic syndrome was the only significant parameter increasing the risk of death [HR = 3.40, 95% CI (1.05-10.98), P = 0.04]. CONCLUSION The cumulative incidence of pre-engraftment bacteremia and pneumonia was 14.3% and 11.2% respectively in this cohort. Aplastic anemia predicted for the occurrence of bacteremia, increasing patient age contributed to the occurrence of pneumonia, and myelodysplastic syndrome increased the risk of death.
Collapse
Affiliation(s)
- Rima Moghnieh
- Division of Infectious Diseases, Department of Internal Medicine, Makassed General Hospital, Beirut, Lebanon. .,Division of Infectious Diseases, Department of Internal Medicine, Hôtel Dieu de France, Beirut, Lebanon.
| | - Hani Tamim
- Department of Internal Medicine, American University of Beirut, Beirut, Lebanon
| | - Amin Abyad
- Division of Hematology/Oncology, Department of Internal Medicine, Makassed General Hospital, Beirut, Lebanon
| | - Marwa Jadayel
- School of Pharmacy, Beirut Arab University, Beirut, Lebanon
| | - Lyn Awad
- Pharmacy Department, Makassed General Hospital, Beirut, Lebanon
| | - Dania Abdallah
- Pharmacy Department, Makassed General Hospital, Beirut, Lebanon
| | - Nicholas Haddad
- Infectious Disease and Residency Program, Internal Medicine, Central Michigan University, Saginaw, MI, 48602, USA
| | | | - Rawya Alwazir
- Faculty of Medicine, Lebanese University, Beirut, Lebanon
| | - Rida Taher
- Faculty of Medicine, Lebanese University, Beirut, Lebanon
| | - Tamima Jisr
- Department of Laboratory Medicine, Makassed General Hospital, Beirut, Lebanon
| | - Rawad Lakkis
- Faculty of Arts and Sciences, American University of Beirut, Beirut, Lebanon
| | - Kamal Zahran
- Division of Hematology/Oncology, Middle East Institute of Health, Bsalim, Lebanon
| | - Ahmad Ibrahim
- Division of Hematology/Oncology, Department of Internal Medicine, Makassed General Hospital, Beirut, Lebanon
| |
Collapse
|
50
|
Niederman MS, Nair GB, Matt U, Herold S, Pennington K, Crothers K, Cummings M, Schluger NW. Update in Lung Infections and Tuberculosis 2018. Am J Respir Crit Care Med 2020; 200:414-422. [PMID: 31042415 DOI: 10.1164/rccm.201903-0606up] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
- Michael S Niederman
- 1Division of Pulmonary and Critical Care Medicine, Weill Cornell Medical College, New York Presbyterian/Weill Cornell Medical Center, New York, New York
| | - Girish Balachandran Nair
- 2Division of Pulmonary and Critical Care Medicine, Beaumont Health, William Beaumont School of Medicine, Oakland University, Royal Oak, Michigan
| | - Ulrich Matt
- 3Department of Internal Medicine II, Universities of Giessen and Marburg Lung Center, member of the German Center for Lung Research, Justus Liebig University, Giessen, Germany
| | - Susanne Herold
- 3Department of Internal Medicine II, Universities of Giessen and Marburg Lung Center, member of the German Center for Lung Research, Justus Liebig University, Giessen, Germany
| | - Kelly Pennington
- 4Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota
| | - Kristina Crothers
- 5Pulmonary, Critical Care and Sleep Medicine, VA Puget Sound Health Care System, Seattle, Washington.,6University of Washington, Seattle, Washington; and
| | | | - Neil W Schluger
- 7Columbia University Irving Medical Center, New York, New York
| |
Collapse
|