1
|
Chen J, Takanami Y, Jansson J, Rossiter G. Practical considerations of promising zone design for interim sample size Re-estimation: An application to GRAPHITE for graft vs host disease. Contemp Clin Trials 2025; 148:107765. [PMID: 39603384 DOI: 10.1016/j.cct.2024.107765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 11/07/2024] [Accepted: 11/23/2024] [Indexed: 11/29/2024]
Abstract
BACKGROUND Sample size calculation and power estimate are an integral part of clinical trials. With accelerated development to address the unmet medical needs, the fast-paced development may lead to uncertainties in initial planning and assumptions of clinical trials. Promising zone design presents sponsors an opportunity to re-estimate the sample size based on the interim data to mitigate risks, reduce uncertainties, and increase probability of trial success. METHODS This paper aims to use the GRAPHITE trial (NCT03657160) as a real data application to showcase the practical considerations in implementation of promising zone design for interim sample size re-estimation (SSR), in light of sample size adaptation rules, maximum sample size allowed, multiplicity adjustment, and sponsor access to interim results. GRAPHITE is a phase 3 trial with vedolizumab for prophylaxis of acute graft vs host disease (aGvHD) after allogeneic hematopoietic stem cell transplant (allo-HSCT). The primary efficacy endpoint is lower intestinal aGVHD-free survival by Day +180 after allo-HSCT. A simulation study was conducted to demonstrate the evaluation of operating characteristics by various true underlying treatment effects at the design stage. CONCLUSION The application of promising zone design for interim SSR is novel and has successfully helped the sponsor achieve the balance between minimizing the risks and maintaining scientific integrity. This work aims to highlight the necessity of empirical guidance to gain better insights for clinical researchers in practice and is expected to facilitate the understanding and implementation of promising zone design for interim SSR in phase 3 trials.
Collapse
Affiliation(s)
- Jingjing Chen
- Takeda Development Center Americas, Inc., Cambridge, MA, USA.
| | | | - Johan Jansson
- Takeda Development Center Americas, Inc., Cambridge, MA, USA
| | | |
Collapse
|
2
|
Ehx G, Ritacco C, Baron F. Pathophysiology and preclinical relevance of experimental graft-versus-host disease in humanized mice. Biomark Res 2024; 12:139. [PMID: 39543777 PMCID: PMC11566168 DOI: 10.1186/s40364-024-00684-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Accepted: 11/06/2024] [Indexed: 11/17/2024] Open
Abstract
Graft-versus-host disease (GVHD) is a life-threatening complication of allogeneic hematopoietic cell transplantations (allo-HCT) used for the treatment of hematological malignancies and other blood-related disorders. Until recently, the discovery of actionable molecular targets to treat GVHD and their preclinical testing was almost exclusively based on modeling allo-HCT in mice by transplanting bone marrow and splenocytes from donor mice into MHC-mismatched recipient animals. However, due to fundamental differences between human and mouse immunology, the translation of these molecular targets into the clinic can be limited. Therefore, humanized mouse models of GVHD were developed to circumvent this limitation. In these models, following the transplantation of human peripheral blood mononuclear cells (PBMCs) into immunodeficient mice, T cells recognize and attack mouse organs, inducing GVHD. Thereby, humanized mice provide a platform for the evaluation of the effects of candidate therapies on GVHD mediated by human immune cells in vivo. Understanding the pathophysiology of this xenogeneic GVHD is therefore crucial for the design and interpretation of experiments performed with this model. In this article, we comprehensively review the cellular and molecular mechanisms governing GVHD in the most commonly used model of xenogeneic GVHD: PBMC-engrafted NOD/LtSz-PrkdcscidIL2rγtm1Wjl (NSG) mice. By re-analyzing public sequencing data, we also show that the clonal expansion and the transcriptional program of T cells in humanized mice closely reflect those in humans. Finally, we highlight the strengths and limitations of this model, as well as arguments in favor of its biological relevance for studying T-cell reactions against healthy tissues or cancer cells.
Collapse
Affiliation(s)
- Grégory Ehx
- Laboratory of Hematology, GIGA Institute, University of Liege, Liege, Belgium.
- Walloon Excellence in Life Sciences and Biotechnology (WELBIO) Department, WEL Research Institute, Wavre, Belgium.
| | - Caroline Ritacco
- Laboratory of Hematology, GIGA Institute, University of Liege, Liege, Belgium
| | - Frédéric Baron
- Laboratory of Hematology, GIGA Institute, University of Liege, Liege, Belgium
- Department of Medicine, Division of Hematology, CHU of Liege, University of Liege, Liege, Belgium
| |
Collapse
|
3
|
Maccioni F, La Rocca U, Milanese A, Busato L, Cleri A, Lopez M, Manganaro L, De Felice C, Di Gioia C, Vestri AR, Catalano C, Iori AP. Multi-parametric MRI in the diagnosis and scoring of gastrointestinal acute graft-versus-host disease. Eur Radiol 2023; 33:5911-5923. [PMID: 37071163 PMCID: PMC10415479 DOI: 10.1007/s00330-023-09563-7] [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: 05/26/2022] [Revised: 01/11/2023] [Accepted: 02/26/2023] [Indexed: 04/19/2023]
Abstract
OBJECTIVES Acute gastrointestinal graft-versus-host disease (GI-aGVHD) is a severe complication of allogeneic hematopoietic stem cell transplantation (HSCT). Diagnosis relies on clinical, endoscopic, and pathological investigations. Our purpose is to assess the value of magnetic resonance imaging (MRI) in the diagnosis, staging, and prediction of GI-aGVHD-related mortality. METHODS Twenty-one hematological patients who underwent MRI for clinical suspicion of acute GI-GVHD were retrospectively selected. Three independent radiologists, blinded to the clinical findings, reanalyzed MRI images. The GI tract was evaluated from stomach to rectum by analyzing fifteen MRI signs suggestive of intestinal and peritoneal inflammation. All selected patients underwent colonoscopy with biopsies. Disease severity was determined on the basis of clinical criteria, identifying 4 stages of increasing severity. Disease-related mortality was also assessed. RESULTS The diagnosis of GI-aGVHD was histologically confirmed with biopsy in 13 patients (61.9%). Using 6 major signs (diagnostic score), MRI showed 84.6% sensitivity and 100% specificity in identifying GI-aGVHD (AUC = 0.962; 95% confidence interval 0.891-1). The proximal, middle, and distal ileum were the segments most frequently affected by the disease (84.6%). Using all 15 signs of inflammation (severity score), MRI showed 100% sensitivity and 90% specificity for 1-month related mortality. No correlation with the clinical score was found. CONCLUSION MRI has proved to be an effective tool for diagnosing and scoring GI-aGVHD, with a high prognostic value. If larger studies will confirm these results, MRI could partly replace endoscopy, thus becoming the primary diagnostic tool for GI-aGVHD, being more complete, less invasive, and more easily repeatable. KEY POINTS • We have developed a new promising MRI diagnostic score for GI-aGVHD with a sensitivity of 84.6% and specificity of 100%; results are to be confirmed by larger multicentric studies. • This MRI diagnostic score is based on the six MRI signs most frequently associated with GI-aGVHD: small-bowel inflammatory involvement, bowel wall stratification on T2-w images, wall stratification on post-contrast T1-w images, ascites, and edema of retroperitoneal fat and declivous soft tissues. • A broader MRI severity score based on 15 MRI signs showed no correlation with clinical staging but high prognostic value (100% sensitivity, 90% specificity for 1-month related mortality); these results also need to be confirmed by larger studies.
Collapse
Affiliation(s)
- Francesca Maccioni
- Department of Radiological Sciences, Pathology and Oncology, Policlinico Umberto I Hospital, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy.
| | - Ursula La Rocca
- Department of Translational and Precision Medicine, Policlinico Umberto I Hospital, Sapienza University of Rome, Via Benevento 6, 00161, Rome, RM, Italy
| | - Alberto Milanese
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185, Rome, Italy
| | - Ludovica Busato
- Department of Radiological Sciences, Pathology and Oncology, Policlinico Umberto I Hospital, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy
| | - Arianna Cleri
- Department of Radiological Sciences, Pathology and Oncology, Policlinico Umberto I Hospital, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy
| | - Mariangela Lopez
- Department of Radiological Sciences, Pathology and Oncology, Policlinico Umberto I Hospital, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy
| | - Lucia Manganaro
- Department of Radiological Sciences, Pathology and Oncology, Policlinico Umberto I Hospital, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy
| | - Carlo De Felice
- Department of Radiological Sciences, Pathology and Oncology, Policlinico Umberto I Hospital, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy
| | - Cira Di Gioia
- Department of Radiological Sciences, Pathology and Oncology, Policlinico Umberto I Hospital, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy
| | - Anna Rita Vestri
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185, Rome, Italy
| | - Carlo Catalano
- Department of Radiological Sciences, Pathology and Oncology, Policlinico Umberto I Hospital, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy
| | - Anna Paola Iori
- Department of Translational and Precision Medicine, Policlinico Umberto I Hospital, Sapienza University of Rome, Via Benevento 6, 00161, Rome, RM, Italy
| |
Collapse
|
4
|
Mirfakhraie R, Ardakani MT, Hajifathali A, Karami S, Moshari MR, Hassani M, Firouz SM, Roshandel E. Highlighting the interaction between immunomodulatory properties of mesenchymal stem cells and signaling pathways contribute to Graft Versus Host Disease management. Transpl Immunol 2022; 71:101524. [PMID: 34990789 DOI: 10.1016/j.trim.2021.101524] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Revised: 12/29/2021] [Accepted: 12/29/2021] [Indexed: 12/11/2022]
Abstract
Background Allogeneic hematopoietic stem cell transplantation (Allo-HSCT) has been increasingly used as a therapeutic approach for hematological malignancies. Several potential strategies have been developed for treating or preventing allo-HSCT complications, specifically graft-versus-host disease (GVHD). GVHD could significantly affect the morbidity and mortality of patients after allo-HSCT. Curative treatment and prophylaxis regimens for GVHD could reduce GVHD incidence and improve survival rate. Among these therapeutic strategies, mesenchymal stem cell (MSCs) mediated immunomodulation has been explored widely in clinical trials. MSCs immunomodulation ability in GVHD correlates with the interactions of MSCs with innate and adaptive immune cells. However, signaling pathways responsible for MSCs' impact on GVHD regulation, like JAK/STAT, NOTCH, MAPK/ERK, and NFκβ signaling pathways, have not been clearly described yet. This review aims to illuminate the effect of MSCs-mediated immunomodulation in GVHD management after allo-HSCT representing the role of MSCs therapy on signaling pathways in GVHD. Conclusion MSCs could potentially modulate immune responses, prevent GVHD, and improve survival after allo-HSCT. Previous studies have investigated different signaling pathways' contributions to MSCs immunoregulatory ability. Accordingly, targeting signaling pathways components involved in MSCs related GVHD regulation is proven to be beneficial.
Collapse
Affiliation(s)
- Reza Mirfakhraie
- Hematopoietic Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maria Tavakoli Ardakani
- Department of Clinical Pharmacy, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abbas Hajifathali
- Hematopoietic Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Samira Karami
- Hematopoietic Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Mohammad Reza Moshari
- Department of Anesthesiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Hassani
- Department of General Surgery, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sahar Mashayekhi Firouz
- Department of Immunology, Afzalipour Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Elham Roshandel
- Hematopoietic Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
5
|
Sun X, He Q, Yang J, Wang A, Zhang F, Qiu H, Zhou K, Wang P, Ding X, Yuan X, Li H, Zhang Y, Song X. Preventive and Therapeutic Effects of a Novel JAK Inhibitor SHR0302 in Acute Graft-Versus-Host Disease. Cell Transplant 2021; 30:9636897211033778. [PMID: 34269100 PMCID: PMC8287347 DOI: 10.1177/09636897211033778] [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] [Indexed: 12/14/2022] Open
Abstract
Acute graft-versus-host disease (aGVHD) is one of the most common complications
of allogeneic hematopoietic stem cell transplantation (allo-HSCT). Janus kinase
(JAK) inhibitors are considered as reliable and promising agents for patients
with aGVHD. The prophylactic and therapeutic effects of SHR0302, a novel JAK
inhibitor, were evaluated in aGVHD mouse models. The overall survival (OS),
progression-free survival (PFS), bodyweight of mice, GVHD scores were observed
and recorded. The bone marrow and spleen samples of diseased model mice or
peripheral blood of patients were analyzed. SHR0302 could prevent and reverse
aGVHD in mouse models with preserving graft-versus-tumor effect. Functionally,
SHR0302 improved the OS and PFS, restored bodyweight, reduced GVHD scores, and
reduced immune cells infiltrated in target tissues. SHR0302 treatment also
enhanced the hematopoietic reconstruction compared to the control group.
Mechanistically, our results suggested that SHR0302 could inhibit the activation
of T cells and modulate the differentiation of helper T (Th) cells by reducing
Th1 and increasing regulatory T (Treg) cells. In addition, SHR0302 decreased the
expression of chemokine receptor CXCR3 on donor T cells and the secretion of
cytokines or chemokines including interleukin (IL)-6, interferon γ (IFN-γ),
tumor necrosis factor α (TNF-α), CXCL10, etc. thereby destroying the
IFN-γ/CXCR3/CXCL10 axis which promotes the progression of GVHD. Besides, SHR0302
decreased the phosphorylation of JAK and its downstream STATs, AKT and ERK1/2,
which ultimately regulated the activation, proliferation, and differentiation of
lymphocytes. Experiments on primary cells from aGVHD patients also confirmed the
results. In summary, our results indicated that JAK inhibitor SHR0302 might be
used as a novel agent for patients with aGVHD.
Collapse
Affiliation(s)
- Xi Sun
- Department of Hematology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Qiaomei He
- Department of Hematology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Jun Yang
- Department of Hematology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.,Engineering Technology Research Center of Cell Therapy and Clinical Translation, Shanghai Science and Technology Committee (STCSM), Shanghai, China
| | - Andi Wang
- Department of Hematology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Fang Zhang
- Department of Hematology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Huiying Qiu
- Department of Hematology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.,Engineering Technology Research Center of Cell Therapy and Clinical Translation, Shanghai Science and Technology Committee (STCSM), Shanghai, China
| | - Kun Zhou
- Department of Hematology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.,Engineering Technology Research Center of Cell Therapy and Clinical Translation, Shanghai Science and Technology Committee (STCSM), Shanghai, China
| | - Pengran Wang
- Department of Hematology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Xiaodan Ding
- Department of Hematology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Xiujie Yuan
- Department of Hematology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Huajun Li
- Jiangsu Hengrui Pharmaceuticals Co., Ltd., Shanghai, China
| | - Yan Zhang
- Department of Hematology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Xianmin Song
- Department of Hematology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.,Engineering Technology Research Center of Cell Therapy and Clinical Translation, Shanghai Science and Technology Committee (STCSM), Shanghai, China
| |
Collapse
|
6
|
Tecchio C, Cassatella MA. Uncovering the multifaceted roles played by neutrophils in allogeneic hematopoietic stem cell transplantation. Cell Mol Immunol 2021; 18:905-918. [PMID: 33203938 PMCID: PMC8115169 DOI: 10.1038/s41423-020-00581-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Accepted: 10/22/2020] [Indexed: 02/07/2023] Open
Abstract
Allogeneic hematopoietic stem cell transplantation (alloHSCT) is a life-saving procedure used for the treatment of selected hematological malignancies, inborn errors of metabolism, and bone marrow failures. The role of neutrophils in alloHSCT has been traditionally evaluated only in the context of their ability to act as a first line of defense against infection. However, recent evidence has highlighted neutrophils as key effectors of innate and adaptive immune responses through a wide array of newly discovered functions. Accordingly, neutrophils are emerging as highly versatile cells that are able to acquire different, often opposite, functional capacities depending on the microenvironment and their differentiation status. Herein, we review the current knowledge on the multiple functions that neutrophils exhibit through the different stages of alloHSCT, from the hematopoietic stem cell (HSC) mobilization in the donor to the immunological reconstitution that occurs in the recipient following HSC infusion. We also discuss the influence exerted on neutrophils by the immunosuppressive drugs delivered in the course of alloHSCT as part of graft-versus-host disease (GVHD) prophylaxis. Finally, the potential involvement of neutrophils in alloHSCT-related complications, such as transplant-associated thrombotic microangiopathy (TA-TMA), acute and chronic GVHD, and cytomegalovirus (CMV) reactivation, is also discussed. Based on the data reviewed herein, the role played by neutrophils in alloHSCT is far greater than a simple antimicrobial role. However, much remains to be investigated in terms of the potential functions that neutrophils might exert during a highly complex procedure such as alloHSCT.
Collapse
Affiliation(s)
- Cristina Tecchio
- Department of Medicine, Section of Hematology and Bone Marrow Transplant Unit, University of Verona, Verona, Italy.
| | | |
Collapse
|
7
|
Elgarten CW, Li Y, Getz KD, Hemmer M, Huang YSV, Hall M, Wang T, Kitko CL, Jagasia MH, Nishihori T, Murthy HS, Hashem H, Cairo MS, Sharma A, Hashmi SK, Askar M, Beitinjaneh A, Kelly MS, Auletta JJ, Badawy SM, Mavers M, Aplenc R, MacMillan ML, Spellman SR, Arora M, Fisher BT. Broad-Spectrum Antibiotics and Risk of Graft-versus-Host Disease in Pediatric Patients Undergoing Transplantation for Acute Leukemia: Association of Carbapenem Use with the Risk of Acute Graft-versus-Host Disease. Transplant Cell Ther 2020; 27:177.e1-177.e8. [PMID: 33718896 DOI: 10.1016/j.jtct.2020.10.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Variation in the gastrointestinal (GI) microbiota after hematopoietic cell transplantation (HCT) has been associated with acute graft-versus-host disease (aGVHD). Because antibiotics induce dysbiosis, we examined the association of broad-spectrum antibiotics with subsequent aGVHD risk in pediatric patients undergoing HCT for acute leukemia. We performed a retrospective analysis in a dataset merged from 2 sources: (1) the Center for International Blood and Marrow Transplant Research, an observational transplantation registry, and (2) the Pediatric Health Information Services, an administrative database from freestanding children's hospitals. We captured exposure to 3 classes of antibiotics used for empiric treatment of febrile neutropenia: (1) broad-spectrum cephalosporins, (2) antipseudomonal penicillins, and (3) carbapenems. The primary outcome was grade II-IV aGVHD; secondary outcomes were grade III-IV aGVHD and lower GI GVHD. The adjusted logistic regression model (full cohort) and time-to-event analysis (subcohort) included transplantation characteristics, GVHD risk factors, and adjunctive antibiotic exposures as covariates. The full cohort included 2550 patients at 36 centers; the subcohort included 1174 patients. In adjusted models, carbapenems were associated with an increased risk of grade II-IV aGVHD in the full cohort (adjusted odds ratio [aOR], 1.24; 95% confidence interval [CI], 1.02 to 1.51) and subcohort (sub hazard ratio [HR], 1.31; 95% CI, 0.99 to 1.72), as well as with an increased risk of grade III-IV aGVHD (subHR, 1.77; 95% CI, 1.25 to 2.52). Early carbapenem exposure (before day 0) especially impacted aGVHD risk. For antipseudomonal penicillins, the associations with aGVHD were in the direction of increased risk but were not statistically significant. There was no identified association between broad-spectrum cephalosporins and aGVHD. Carbapenems, more than other broad-spectrum antibiotics, should be used judiciously in pediatric HCT recipients to minimize aGVHD risk. Further research is needed to clarify the mechanism underlying this association.
Collapse
Affiliation(s)
- Caitlin W Elgarten
- Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, PA.,Center for Pediatric Clinical Effectiveness, Children's Hospital of Philadelphia, Philadelphia, PA
| | - Yimei Li
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia PA
| | - Kelly D Getz
- Center for Pediatric Clinical Effectiveness, Children's Hospital of Philadelphia, Philadelphia, PA.,Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia PA
| | | | - Yuan-Shung V Huang
- Department of Biomedical and Health Informatics, Children's Hospital of Philadelphia, Philadelphia, PA
| | | | - Tao Wang
- Division of Biostatistics, Institute for Health and Equity, Medical College of Wisconsin, Milwaukee, WI
| | - Carrie L Kitko
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN
| | | | - Taiga Nishihori
- Department of Blood and Marrow Transplantation, H. Lee Moffitt Cancer Center, Tampa, FL
| | - Hemant S Murthy
- Division of Hematology-Oncology, Blood and Marrow Transplantation Program, Mayo Clinic, Jacksonville, FL
| | - Hasan Hashem
- Division of Pediatric Hematology/Oncology and Bone Marrow Transplantation, King Hussein Cancer Center, Amman, Jordan
| | - Mitchell S Cairo
- Division of Pediatric Hematology, Oncology and Stem Cell Transplantation, Department of Pediatrics, New York Medical College, Valhalla, NY
| | - Akshay Sharma
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, Memphis, TN
| | - Shahrukh K Hashmi
- Department of Internal Medicine, Mayo Clinic, Rochester, MN.,Oncology Center, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Medhat Askar
- Department of Pathology and Laboratory Medicine, Baylor University Medical Center, Dallas, TX
| | - Amer Beitinjaneh
- Division of Transplantation and Cellular Therapy, University of Miami, Miami, FL
| | - Matthew S Kelly
- Division of Pediatric Infectious Diseases, Duke University Medical Center, Durham, NC
| | - Jeffery J Auletta
- Blood and Marrow Transplant Program and Host Defense Program, Divisions of Hematology/Oncology/Bone Marrow Transplant and Infectious Diseases, Nationwide Children's Hospital, Columbus, OH
| | - Sherif M Badawy
- Division of Hematology, Oncology and Stem Cell Transplant, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL.,Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Melissa Mavers
- Division of Stem Cell Transplantation and Regenerative Medicine, Department of Pediatrics, Bass Center for Childhood Cancer and Blood Diseases, Stanford University School of Medicine, Palo Alto, CA
| | - Richard Aplenc
- Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, PA.,Center for Pediatric Clinical Effectiveness, Children's Hospital of Philadelphia, Philadelphia, PA.,Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia PA
| | - Margaret L MacMillan
- Blood and Marrow Transplant Program, Department of Pediatrics, University of Minnesota, Minneapolis, MN
| | | | - Mukta Arora
- Division of Hematology, Oncology and Transplantation, Department of Medicine, University of Minnesota Medical Center, Minneapolis, MN
| | - Brian T Fisher
- Center for Pediatric Clinical Effectiveness, Children's Hospital of Philadelphia, Philadelphia, PA.,Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia PA.,Infectious Diseases, Children's Hospital of Philadelphia, Philadelphia, PA
| |
Collapse
|
8
|
Vandenhove B, Canti L, Schoemans H, Beguin Y, Baron F, Graux C, Kerre T, Servais S. How to Make an Immune System and a Foreign Host Quickly Cohabit in Peace? The Challenge of Acute Graft- Versus-Host Disease Prevention After Allogeneic Hematopoietic Cell Transplantation. Front Immunol 2020; 11:583564. [PMID: 33193397 PMCID: PMC7609863 DOI: 10.3389/fimmu.2020.583564] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 09/21/2020] [Indexed: 01/16/2023] Open
Abstract
Allogeneic hematopoietic cell transplantation (alloHCT) has been used as cellular immunotherapy against hematological cancers for more than six decades. Its therapeutic efficacy relies on the cytoreductive effects of the conditioning regimen but also on potent graft-versus-tumor (GVT) reactions mediated by donor-derived immune cells. However, beneficial GVT effects may be counterbalanced by acute GVHD (aGVHD), a systemic syndrome in which donor immune cells attack healthy tissues of the recipient, resulting in severe inflammatory lesions mainly of the skin, gut, and liver. Despite standard prophylaxis regimens, aGVHD still occurs in approximately 20–50% of alloHCT recipients and remains a leading cause of transplant-related mortality. Over the past two decades, advances in the understanding its pathophysiology have helped to redefine aGVHD reactions and clinical presentations as well as developing novel strategies to optimize its prevention. In this review, we provide a brief overview of current knowledge on aGVHD immunopathology and discuss current approaches and novel strategies being developed and evaluated in clinical trials for aGVHD prevention. Optimal prophylaxis of aGVHD would prevent the development of clinically significant aGVHD, while preserving sufficient immune responsiveness to maintain beneficial GVT effects and immune defenses against pathogens.
Collapse
Affiliation(s)
- Benoît Vandenhove
- Laboratory of Hematology, GIGA-I3, GIGA Institute, University of Liège, Liège, Belgium
| | - Lorenzo Canti
- Laboratory of Hematology, GIGA-I3, GIGA Institute, University of Liège, Liège, Belgium
| | - Hélène Schoemans
- Department of Clinical Hematology, University Hospitals Leuven, KU Leuven, Leuven, Belgium
| | - Yves Beguin
- Laboratory of Hematology, GIGA-I3, GIGA Institute, University of Liège, Liège, Belgium.,Department of Clinical Hematology, CHU of Liège, University of Liège, Liège, Belgium
| | - Frédéric Baron
- Laboratory of Hematology, GIGA-I3, GIGA Institute, University of Liège, Liège, Belgium.,Department of Clinical Hematology, CHU of Liège, University of Liège, Liège, Belgium
| | - Carlos Graux
- Department of Clinical Hematology, CHU UCL Namur (Godinne), Université Catholique de Louvain, Yvoir, Belgium
| | - Tessa Kerre
- Hematology Department, Ghent University Hospital, Ghent University, Ghent, Belgium
| | - Sophie Servais
- Laboratory of Hematology, GIGA-I3, GIGA Institute, University of Liège, Liège, Belgium.,Department of Clinical Hematology, CHU of Liège, University of Liège, Liège, Belgium
| |
Collapse
|
9
|
Leuci S, Coppola N, Blasi A, Ruoppo E, Bizzoca ME, Lo Muzio L, Marano L, Risitano AM, Mignogna MD. Oral Dysplastic Complications after HSCT: Single Case Series of Multidisciplinary Evaluation of 80 Patients. Life (Basel) 2020; 10:life10100236. [PMID: 33050268 PMCID: PMC7600275 DOI: 10.3390/life10100236] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 10/08/2020] [Indexed: 12/13/2022] Open
Abstract
Oral squamous cell carcinoma (OSCC) is the most common secondary solid malignancy after hematopoietic stem-cell transplantation (HSCT). OSCC following HSCT is frequently preceded by chronic graft-versus-host disease (cGVHD). The aim of this study was to describe a cohort of post-HSCT patients and to evaluate the onset of oral epithelial dysplasia and/or OSCC over time. In this retrospective cohort study, we present a cohort of hematological patients that underwent HSCT. Demographic variables, clinical hematological data, data regarding acute graft-versus-host disease (aGVHD) and cGVHD, and oral clinical features were analyzed. We focused on clinicopathological features of a subgroup of 22 patients with oral cGVHD and OSCC after HSCT. Among 80 included patients, 46 patients (57.5%) developed aGVHD and 39 patients (48.7%) developed cGVHD. Oral mucosa was involved in 17 patients with aGVHD (36.9%) and in 22 patients (56.4%) with cGVHD. Out of a total of 22 oral biopsies, roughly 40% revealed mild to moderate dysplasia, and 32% were OSCC. In the absence of international agreement on the best timing of oral follow-up after HSCT, it is mandatory to establish a close multidisciplinary evaluation in order to prevent the onset of HSCT-related OSCC and to reduce post-transplant mortality due to secondary tumors.
Collapse
Affiliation(s)
- Stefania Leuci
- Department of Neurosciences, Reproductive and Odontostomatological Sciences, Oral Medicine Unit, Federico II University of Naples, 80138 Naples, Italy; (S.L.); (A.B.); (E.R.); (M.D.M.)
| | - Noemi Coppola
- Department of Neurosciences, Reproductive and Odontostomatological Sciences, Oral Medicine Unit, Federico II University of Naples, 80138 Naples, Italy; (S.L.); (A.B.); (E.R.); (M.D.M.)
- Correspondence: ; Tel.: +39-3392602615
| | - Andrea Blasi
- Department of Neurosciences, Reproductive and Odontostomatological Sciences, Oral Medicine Unit, Federico II University of Naples, 80138 Naples, Italy; (S.L.); (A.B.); (E.R.); (M.D.M.)
| | - Elvira Ruoppo
- Department of Neurosciences, Reproductive and Odontostomatological Sciences, Oral Medicine Unit, Federico II University of Naples, 80138 Naples, Italy; (S.L.); (A.B.); (E.R.); (M.D.M.)
| | - Maria Eleonora Bizzoca
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy; (M.E.B.); (L.L.M.)
| | - Lorenzo Lo Muzio
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy; (M.E.B.); (L.L.M.)
| | - Luana Marano
- Department of Clinical Medicine and Surgery, Federico II University of Naples, 80138 Naples, Italy;
| | | | - Michele Davide Mignogna
- Department of Neurosciences, Reproductive and Odontostomatological Sciences, Oral Medicine Unit, Federico II University of Naples, 80138 Naples, Italy; (S.L.); (A.B.); (E.R.); (M.D.M.)
| |
Collapse
|
10
|
Vedolizumab for prevention of graft-versus-host disease after allogeneic hematopoietic stem cell transplantation. Blood Adv 2020; 3:4136-4146. [PMID: 31821456 DOI: 10.1182/bloodadvances.2019000893] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 10/31/2019] [Indexed: 12/21/2022] Open
Abstract
Acute graft-versus-host disease (aGVHD) remains a significant complication of allogeneic hematopoietic stem cell transplantation (allo-HSCT). Vedolizumab could help prevent aGVHD by inhibiting the migration of both naive and activated lymphocytes into gut-associated lymphoid tissues and the lamina propria. We carried out a phase 1b, open-label, dose-finding study in adults undergoing allo-HSCT to evaluate the tolerability, safety, and pharmacokinetics of vedolizumab, and its effectiveness in reducing aGVHD. IV vedolizumab was administered on day -1, +13, and +42 with respect to allo-HSCT, starting at 75 mg and with dose escalation guided by tolerability and pharmacokinetics. A total of 24 participants was enrolled, and no dose-limiting toxicities were observed in either the 75-mg cohort (n = 3) or the dose-escalated 300-mg cohort (n = 21). Treatment-emergent adverse events related to vedolizumab occurred in 8 participants. Overall, 4 deaths occurred during the 12 months following allo-HSCT. No participants in the 75-mg cohort developed modified Glucksberg grade II to IV aGVHD by 100 days after allo-HSCT. Four participants (19.0%) in the 300-mg cohort developed grade II to IV aGVHD by 100 days after allo-HSCT, including 3 participants who developed stage 1 aGVHD of the lower-intestinal tract. Vedolizumab IV 300 mg was well tolerated as aGVHD prevention, and the incidence of overall and lower-intestinal aGVHD was low. These findings support further evaluation of vedolizumab in this patient population. This trial was registered at www.clinicaltrials.gov as #NCT02728895.
Collapse
|
11
|
Khandelwal P, Fukuda T, Teusink-Cross A, Kashuba ADM, Lane A, Mehta PA, Marsh RA, Jordan MB, Grimley MS, Myers KC, Nelson AS, El-Bietar J, Chandra S, Bleesing JJ, Krupski MC, Davies SM. CCR5 inhibitor as novel acute graft versus host disease prophylaxis in children and young adults undergoing allogeneic stem cell transplant: results of the phase II study. Bone Marrow Transplant 2020. [PMID: 32273585 DOI: 10.1038/s41409–020–0888–3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
We report results of a phase II study of maraviroc to prevent acute graft versus host disease (GVHD) in children undergoing allogeneic hematopoietic stem cell transplant (HSCT). Oral maraviroc was added to standard GVHD prophylaxis of a calcineurin inhibitor with either mycophenolate mofetil, methotrexate or steroids from day -3 until day +30 after HSCT. Maraviroc trough levels were analyzed on day 0, +7, 14, and 21. We assessed functional CCR5 blockade by our previously described pharmacodynamic assay. In total, 17 patients were enrolled prospectively. No patient had liver GVHD by day +100. Four patients developed gastrointestinal (GI) GVHD (Grade II upper GI GVHD n = 2, grade III lower GI GVHD n = 2). No adverse effects of maraviroc were observed. Seven patients discontinued maraviroc at a median of day +14 (range day +1-day +29) due to study rules regarding hepatotoxicity (n = 5), renal function decline (n = 1) and withdrawal from study (n = 1). Maraviroc administration led to CCR5 inhibition but was limited by study rules defining hepatotoxicity, leading to frequent drug discontinuation. We cannot comment on the efficacy of maraviroc with our data but speculate that it could have a role in prevention of acute GI GVHD, with adequate compliance.
Collapse
Affiliation(s)
- Pooja Khandelwal
- Division of Bone Marrow Transplant and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| | - Tsuyoshi Fukuda
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Division of Clinical Pharmacology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Ashley Teusink-Cross
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Division of Pharmacy, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Angela D M Kashuba
- Division of Pharmacotherapy and Experimental Therapeutics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Adam Lane
- Division of Bone Marrow Transplant and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Parinda A Mehta
- Division of Bone Marrow Transplant and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Rebecca A Marsh
- Division of Bone Marrow Transplant and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Michael B Jordan
- Division of Bone Marrow Transplant and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Michael S Grimley
- Division of Bone Marrow Transplant and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Kasiani C Myers
- Division of Bone Marrow Transplant and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Adam S Nelson
- Division of Bone Marrow Transplant and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Javier El-Bietar
- Division of Bone Marrow Transplant and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Sharat Chandra
- Division of Bone Marrow Transplant and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Jacob J Bleesing
- Division of Bone Marrow Transplant and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Mary C Krupski
- Division of Bone Marrow Transplant and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Stella M Davies
- Division of Bone Marrow Transplant and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| |
Collapse
|
12
|
Boucault L, Lopez Robles MD, Thiolat A, Bézie S, Schmueck-Henneresse M, Braudeau C, Vimond N, Freuchet A, Autrusseau E, Charlotte F, Redjoul R, Beckerich F, Leclerc M, Piaggio E, Josien R, Volk HD, Maury S, Cohen JL, Anegon I, Guillonneau C. Transient antibody targeting of CD45RC inhibits the development of graft-versus-host disease. Blood Adv 2020; 4:2501-2515. [PMID: 32511714 PMCID: PMC7284095 DOI: 10.1182/bloodadvances.2020001688] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 04/24/2020] [Indexed: 02/06/2023] Open
Abstract
Allogeneic bone marrow transplantation (BMT) is a widely spread treatment of many hematological diseases, but its most important side effect is graft-versus-host disease (GVHD). Despite the development of new therapies, acute GVHD (aGVHD) occurs in 30% to 50% of allogeneic BMT and is characterized by the generation of effector T (Teff) cells with production of inflammatory cytokines. We previously demonstrated that a short anti-CD45RC monoclonal antibody (mAb) treatment in a heart allograft rat model transiently decreased CD45RChigh Teff cells and increased regulatory T cell (Treg) number and function allowing long-term donor-specific tolerance. Here, we demonstrated in rat and mouse allogeneic GVHD, as well as in xenogeneic GVHD mediated by human T cells in NSG mice, that both ex vivo depletion of CD45RChigh T cells and in vivo treatment with short-course anti-CD45RC mAbs inhibited aGVHD. In the rat model, we demonstrated that long surviving animals treated with anti-CD45RC mAbs were fully engrafted with donor cells and developed a donor-specific tolerance. Finally, we validated the rejection of a human tumor in NSG mice infused with human cells and treated with anti-CD45RC mAbs. The anti-human CD45RC mAbs showed a favorable safety profile because it did not abolish human memory antiviral immune responses, nor trigger cytokine release in in vitro assays. Altogether, our results show the potential of a prophylactic treatment with anti-human CD45RC mAbs in combination with rapamycin as a new therapy to treat aGVHD without abolishing the antitumor effect.
Collapse
Affiliation(s)
- Laetitia Boucault
- Centre de Recherche en Transplantation et Immunologie, Institut de Transplantation Urologie-Néphrologie (ITUN), Unité Mixte de Recherche (UMR) 1064, INSERM/Université de Nantes, Nantes, France
- Immunotherapy, Graft, Oncology (IGO), LabEx, Nantes, France
| | - Maria-Dolores Lopez Robles
- Centre de Recherche en Transplantation et Immunologie, Institut de Transplantation Urologie-Néphrologie (ITUN), Unité Mixte de Recherche (UMR) 1064, INSERM/Université de Nantes, Nantes, France
- Immunotherapy, Graft, Oncology (IGO), LabEx, Nantes, France
| | - Allan Thiolat
- Université Paris-Est Créteil, INSERM, Institut Mondor de Recherche Biomédicale (IMRB), Creteil, France
| | - Séverine Bézie
- Centre de Recherche en Transplantation et Immunologie, Institut de Transplantation Urologie-Néphrologie (ITUN), Unité Mixte de Recherche (UMR) 1064, INSERM/Université de Nantes, Nantes, France
- Immunotherapy, Graft, Oncology (IGO), LabEx, Nantes, France
| | - Michael Schmueck-Henneresse
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin/Berlin Institute of Health (BIH), Berlin, Germany
| | - Cécile Braudeau
- Centre de Recherche en Transplantation et Immunologie, Institut de Transplantation Urologie-Néphrologie (ITUN), Unité Mixte de Recherche (UMR) 1064, INSERM/Université de Nantes, Nantes, France
- Immunotherapy, Graft, Oncology (IGO), LabEx, Nantes, France
- Laboratoire d'Immunologie, Centre d'Immunomonitorage Nantes Atlantique (CIMNA), Centre Hospitalier Universitaire (CHU) Nantes, Nantes, France
| | - Nadège Vimond
- Centre de Recherche en Transplantation et Immunologie, Institut de Transplantation Urologie-Néphrologie (ITUN), Unité Mixte de Recherche (UMR) 1064, INSERM/Université de Nantes, Nantes, France
- Immunotherapy, Graft, Oncology (IGO), LabEx, Nantes, France
| | - Antoine Freuchet
- Centre de Recherche en Transplantation et Immunologie, Institut de Transplantation Urologie-Néphrologie (ITUN), Unité Mixte de Recherche (UMR) 1064, INSERM/Université de Nantes, Nantes, France
- Immunotherapy, Graft, Oncology (IGO), LabEx, Nantes, France
| | - Elodie Autrusseau
- Centre de Recherche en Transplantation et Immunologie, Institut de Transplantation Urologie-Néphrologie (ITUN), Unité Mixte de Recherche (UMR) 1064, INSERM/Université de Nantes, Nantes, France
- Immunotherapy, Graft, Oncology (IGO), LabEx, Nantes, France
| | - Frédéric Charlotte
- Service d'Anatomo-Pathologie, Hôpital Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| | - Rabah Redjoul
- AP-HP, Groupe Hospitalo-Universitaire Chenevier Mondor, Service d'Hematologie Clinique, Creteil, France
| | - Florence Beckerich
- AP-HP, Groupe Hospitalo-Universitaire Chenevier Mondor, Service d'Hematologie Clinique, Creteil, France
| | - Mathieu Leclerc
- AP-HP, Groupe Hospitalo-Universitaire Chenevier Mondor, Service d'Hematologie Clinique, Creteil, France
- Université Paris-Est Créteil, INSERM, IMRB, AP-HP, Groupe Hospitalo-Universitaire Chenevier Mondor, Service d'Hematologie Clinique, Creteil, France
| | - Eliane Piaggio
- Translational Research Department, Institut Curie Research Center, Paris Sciences & Lettres (PSL) Research University, U932, INSERM, Paris, France; and
| | - Regis Josien
- Centre de Recherche en Transplantation et Immunologie, Institut de Transplantation Urologie-Néphrologie (ITUN), Unité Mixte de Recherche (UMR) 1064, INSERM/Université de Nantes, Nantes, France
- Immunotherapy, Graft, Oncology (IGO), LabEx, Nantes, France
- Laboratoire d'Immunologie, Centre d'Immunomonitorage Nantes Atlantique (CIMNA), Centre Hospitalier Universitaire (CHU) Nantes, Nantes, France
| | - Hans-Dieter Volk
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin/Berlin Institute of Health (BIH), Berlin, Germany
| | - Sébastien Maury
- Université Paris-Est Créteil, INSERM, IMRB, AP-HP, Groupe Hospitalo-Universitaire Chenevier Mondor, Service d'Hematologie Clinique, Creteil, France
| | - José L Cohen
- Université Paris-Est Créteil, INSERM, IMRB, AP-HP, Groupe Hospitalo-Universitaire Chenevier Mondor, Centre d'Investigation Clinique Biotherapie, Creteil, France
| | - Ignacio Anegon
- Centre de Recherche en Transplantation et Immunologie, Institut de Transplantation Urologie-Néphrologie (ITUN), Unité Mixte de Recherche (UMR) 1064, INSERM/Université de Nantes, Nantes, France
- Immunotherapy, Graft, Oncology (IGO), LabEx, Nantes, France
| | - Carole Guillonneau
- Centre de Recherche en Transplantation et Immunologie, Institut de Transplantation Urologie-Néphrologie (ITUN), Unité Mixte de Recherche (UMR) 1064, INSERM/Université de Nantes, Nantes, France
- Immunotherapy, Graft, Oncology (IGO), LabEx, Nantes, France
| |
Collapse
|
13
|
Snyder KJ, Zitzer NC, Gao Y, Choe HK, Sell NE, Neidemire-Colley L, Ignaci A, Kale C, Devine RD, Abad MG, Pietrzak M, Wang M, Lin H, Zhang YW, Behbehani GK, Jackman JE, Garzon R, Vaddi K, Baiocchi RA, Ranganathan P. PRMT5 regulates T cell interferon response and is a target for acute graft-versus-host disease. JCI Insight 2020; 5:131099. [PMID: 32191634 DOI: 10.1172/jci.insight.131099] [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: 06/18/2019] [Accepted: 03/16/2020] [Indexed: 01/09/2023] Open
Abstract
Acute graft-versus-host disease (aGVHD) is a T cell-mediated immunological disorder and the leading cause of nonrelapse mortality in patients who receive allogeneic hematopoietic cell transplants. Based on recent observations that protein arginine methyltransferase 5 (PRMT5) and arginine methylation are upregulated in activated memory T cells, we hypothesized that PRMT5 is involved in the pathogenesis of aGVHD. Here, we show that PRMT5 expression and enzymatic activity were upregulated in activated T cells in vitro and in T cells from mice developing aGVHD after allogeneic transplant. PRMT5 expression was also upregulated in T cells of patients who developed aGVHD after allogeneic hematopoietic cell transplant compared with those who did not develop aGVHD. PRMT5 inhibition using a selective small-molecule inhibitor (C220) substantially reduced mouse and human allogeneic T cell proliferation and inflammatory IFN-γ and IL-17 cytokine production. Administration of PRMT5 small-molecule inhibitors substantially improves survival, reducing disease incidence and clinical severity in mouse models of aGVHD without adversely affecting engraftment. Importantly, we show that PRMT5 inhibition retained the beneficial graft-versus-leukemia effect by maintaining cytotoxic CD8+ T cell responses. Mechanistically, we show that PRMT5 inhibition potently reduced STAT1 phosphorylation as well as transcription of proinflammatory genes, including interferon-stimulated genes and IL-17. Additionally, PRMT5 inhibition deregulates the cell cycle in activated T cells and disrupts signaling by affecting ERK1/2 phosphorylation. Thus, we have identified PRMT5 as a regulator of T cell responses and as a therapeutic target in aGVHD.
Collapse
Affiliation(s)
- Katiri J Snyder
- Division of Hematology, Department of Internal Medicine, Comprehensive Cancer Center
| | - Nina C Zitzer
- Division of Hematology, Department of Internal Medicine, Comprehensive Cancer Center
| | - Yandi Gao
- Division of Hematology, Department of Internal Medicine, Comprehensive Cancer Center
| | - Hannah K Choe
- Division of Hematology, Department of Internal Medicine, Comprehensive Cancer Center
| | - Natalie E Sell
- Division of Hematology, Department of Internal Medicine, Comprehensive Cancer Center
| | | | - Anora Ignaci
- Division of Hematology, Department of Internal Medicine, Comprehensive Cancer Center
| | - Charuta Kale
- Division of Hematology, Department of Internal Medicine, Comprehensive Cancer Center
| | - Raymond D Devine
- Division of Hematology, Department of Internal Medicine, Comprehensive Cancer Center
| | | | - Maciej Pietrzak
- Department of Biomedical Informatics, The Ohio State University, Columbus, Ohio, USA
| | - Min Wang
- Prelude Therapeutics, Wilmington, Delaware, USA
| | - Hong Lin
- Prelude Therapeutics, Wilmington, Delaware, USA
| | | | - Gregory K Behbehani
- Division of Hematology, Department of Internal Medicine, Comprehensive Cancer Center
| | | | - Ramiro Garzon
- Division of Hematology, Department of Internal Medicine, Comprehensive Cancer Center
| | - Kris Vaddi
- Prelude Therapeutics, Wilmington, Delaware, USA
| | - Robert A Baiocchi
- Division of Hematology, Department of Internal Medicine, Comprehensive Cancer Center
| | - Parvathi Ranganathan
- Division of Hematology, Department of Internal Medicine, Comprehensive Cancer Center
| |
Collapse
|
14
|
CCR5 inhibitor as novel acute graft versus host disease prophylaxis in children and young adults undergoing allogeneic stem cell transplant: results of the phase II study. Bone Marrow Transplant 2020; 55:1552-1559. [PMID: 32273585 DOI: 10.1038/s41409-020-0888-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 03/23/2020] [Accepted: 03/25/2020] [Indexed: 11/09/2022]
Abstract
We report results of a phase II study of maraviroc to prevent acute graft versus host disease (GVHD) in children undergoing allogeneic hematopoietic stem cell transplant (HSCT). Oral maraviroc was added to standard GVHD prophylaxis of a calcineurin inhibitor with either mycophenolate mofetil, methotrexate or steroids from day -3 until day +30 after HSCT. Maraviroc trough levels were analyzed on day 0, +7, 14, and 21. We assessed functional CCR5 blockade by our previously described pharmacodynamic assay. In total, 17 patients were enrolled prospectively. No patient had liver GVHD by day +100. Four patients developed gastrointestinal (GI) GVHD (Grade II upper GI GVHD n = 2, grade III lower GI GVHD n = 2). No adverse effects of maraviroc were observed. Seven patients discontinued maraviroc at a median of day +14 (range day +1-day +29) due to study rules regarding hepatotoxicity (n = 5), renal function decline (n = 1) and withdrawal from study (n = 1). Maraviroc administration led to CCR5 inhibition but was limited by study rules defining hepatotoxicity, leading to frequent drug discontinuation. We cannot comment on the efficacy of maraviroc with our data but speculate that it could have a role in prevention of acute GI GVHD, with adequate compliance.
Collapse
|
15
|
Comparison of HLA-matched sibling and unrelated donor transplantation in adult patients with acquired severe aplastic anemia. Bone Marrow Transplant 2020; 55:1570-1579. [PMID: 32024992 DOI: 10.1038/s41409-020-0820-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 01/27/2020] [Accepted: 01/28/2020] [Indexed: 12/12/2022]
Abstract
The recent improvements in the outcomes of severe aplastic anemia (SAA) patients who received allogeneic stem cell transplantation (SCT) from unrelated donors (URD) suggest the possibility of its alternative first-line treatment. To address this issue, results of adult SAA patients receiving allogeneic SCT were compared between the following three donor-type groups: 8/8-matched sibling (MSD; n = 153), 8/8 well-matched unrelated (WM-URD; n = 72), and 6-7/8 partially matched unrelated (PM-URD; n = 33). Proportion of patients who experienced immunosuppressive treatment failures was significantly higher in the URD groups than in the MSD group (P< 0.01). The incidences of graft failure and transplant-related mortality, and graft-vs.-host disease-free, failure-free survival rates of the MSD, WM-URD, and PM-URD groups were 14.6, 0, and 0% (P< 0.01); 6.1, 10.3, and 21.7% (P= 0.03); and 76.7, 55.5, and 51.5% (P< 0.01), respectively. The overall survival (OS) rate of the MSD group (93.9%) was higher than that of the PM-URD (78.3%; P < 0.01) group, but not to that of the WM-URD (86.2%; P= 0.18) group. Our study showed comparable OS between the MSD group and WM-URD group, which suggest that the URD-SCT can be used as a first-line treatment for adult SAA patients with WM-URD.
Collapse
|
16
|
Can we prevent or treat graft-versus-host disease with cellular-therapy? Blood Rev 2020; 43:100669. [PMID: 32089398 DOI: 10.1016/j.blre.2020.100669] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2019] [Revised: 01/14/2020] [Accepted: 01/28/2020] [Indexed: 12/22/2022]
Abstract
Acute and chronic graft-versus-host disease (GvHD) are the most important causes of treatment-related morbidity and mortality after allogeneic hematopoietic cell transplants for various diseases. Corticosteroids are an effective therapy in only about one-half of affected individuals and new therapy options are needed. We discuss novel strategies to treat GvHD using cellular-therapy including adoptive transfer of regulatory T-cells (Tregs), mesenchymal stromal cells (MSCs), cells derived from placental tissues, invariant natural killer T-cells (iNKTs), and myeloid-derived suppressor cells (MDSCs).These strategies may be more selective than drugs in modulating GvHD pathophysiology, and may be safer and more effective than conventional pharmacologic therapies. Additionally, these therapies have not been observed to substantially compromise the graft-versus-tumor effect associated with allotransplants. Many of these strategies are effective in animal models but substantial data in humans are lacking.
Collapse
|
17
|
Pejčić A, Janković SM, Opančina V, Babić G, Milosavljević M. Drug-drug interactions in patients receiving hematopoietic stem cell transplantation. Expert Opin Drug Metab Toxicol 2018; 15:49-59. [PMID: 30479183 DOI: 10.1080/17425255.2019.1552256] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
INTRODUCTION Recipients of hematopoietic stem cell transplantation (HSCT) are exposed to numerous drugs in both pre- and post-transplantation period, which creates an opportunity for drug-drug interactions (DDIs); if clinically relevant DDIs happen, the risk of adverse treatment outcomes is increased. Areas covered: This review is focused on DDIs in recipients of HSCT that were observed and published as clinical trials, case series or case reports. Relevant publications were found by the systematic search of the following online databases: MEDLINE, SCOPUS, EBSCO, and SCINDEX. Expert opinion: The most important DDIs involve cytostatic or immunosuppressant drug on one side, and antimicrobial drugs on the other. The majority of clinically relevant interactions have pharmacokinetic character, involving drug metabolizing enzymes in the liver. Antifungal azoles inhibit metabolism of many cytostatic and immunosuppressant drugs at cytochromes and increase their plasma concentrations. Macrolide antibiotics and fluoroqunolones should be avoided in HSCT recipients, as they have much larger potential for DDIs than other antibiotic groups. HSCT recipients increasingly receive new immunomodulating drugs, and further observational studies are needed to reveal unsuspected DDIs with clinical relevance.
Collapse
Affiliation(s)
- Ana Pejčić
- a Faculty of Medical Sciences , University of Kragujevac , Kragujevac , Serbia
| | - Slobodan M Janković
- a Faculty of Medical Sciences , University of Kragujevac , Kragujevac , Serbia
| | - Valentina Opančina
- a Faculty of Medical Sciences , University of Kragujevac , Kragujevac , Serbia
| | - Goran Babić
- a Faculty of Medical Sciences , University of Kragujevac , Kragujevac , Serbia
| | - Miloš Milosavljević
- a Faculty of Medical Sciences , University of Kragujevac , Kragujevac , Serbia
| |
Collapse
|
18
|
Barrett J. Why is a 3-year NRM following allogeneic transplantation still stuck at approximately 20%? Best Pract Res Clin Haematol 2018; 31:414-419. [PMID: 30466759 DOI: 10.1016/j.beha.2018.09.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Whether and when to recommend an allogeneic stem cell transplant (SCT) for a patient with leukemia is a treatment decision that rests on determining whether the transplant or non-transplant option carries the greatest probability of 3-5-year survival. While SCT confers a greater possibility of leukemia cure, the decision to transplant has to be made in the light of the high chance of treatment-related mortality (TRM) that follows the allograft. Here we identify that current estimates of a 20% 3-year TRM hold largely true for a variety of leukemias, diverse types of conditioning regimen, and varied donor-recipient compatibility across a wide age-range. While there is a wide spectrum of causes of death in the first few months after SCT, they usually stem from a limited set of immediate post-transplant complications, including those induced by the conditioning regimen, post-transplant endovascular damage, gut dysbiosis, graft-versus-host disease, and immunodeficiency causing viral reactivation. As we better understand and improve preventative treatments for these initiating events there is a real expectation that TRM will continue to fall to levels well below 10% within the next decade.
Collapse
Affiliation(s)
- John Barrett
- Stem Cell Transplantation and Cellular Therapy Program, GW Cancer Center, George Washington University, Medical Faculty Associates, 2150 Pennsylvania Ave NW, Washington, DC 20037, USA.
| |
Collapse
|
19
|
In Vitro Th17-Polarized Human CD4 + T Cells Exacerbate Xenogeneic Graft-versus-Host Disease. Biol Blood Marrow Transplant 2018; 25:204-215. [PMID: 30326279 DOI: 10.1016/j.bbmt.2018.10.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2018] [Accepted: 10/08/2018] [Indexed: 12/14/2022]
Abstract
Acute graft-versus-host disease (aGVHD) is a severe complication of allogeneic hematopoietic stem cell transplantation. The role of Th17 cells in its pathophysiology remains a matter of debate. In this study, we assessed whether enrichment of human peripheral blood mononuclear cells (PBMCs) with in vitro Th17-polarized CD4+ T cells would exacerbate xenogeneic GVHD (xGVHD) into NOD-scid IL-2Rγ null (NSG) mice. Naive human CD4+ T cells were stimulated under Th17-skewing conditions for 8 to 10 days and then coinjected in NSG mice with fresh PBMCs from the same donor. We observed that Th17-polarized cells engrafted and migrated toward xGVHD target organs. They also acquired a double-expressing IL-17A+IFNγ+ profile in vivo. Importantly, cotransfer of Th17-polarized cells (1 × 106) with PBMCs (1 × 106) exacerbated xGVHD compared with transplantation of PBMCs alone (2 × 106). Furthermore, PBMC cotransfer with Th17-polarized cells was more potent for xGVHD induction than cotransfer with naive CD4+ T cells stimulated in nonpolarizing conditions (Th0 cells, 1 × 106 + 1 × 106 PBMCs) or with Th1-polarized cells (1 × 106 + 1 × 106 PBMCs). In summary, our results suggest that human Th17-polarized cells can cooperate with PBMCs and be pathogenic in the NSG xGVHD model.
Collapse
|
20
|
Bonifazi F, Olivieri J, Sessa M, Dan E, Sinigaglia B, Rizzi S, Motta MR, Bontadini A, Ulbar F, Giudice V, Papayannidis C, Curti A, Chiereghin A, Lazzarotto T, Cavo M, Arpinati M. Low-Dose Anti-T Lymphoglobulin as Prophylaxis for Graft-versus-Host Disease in Unrelated Donor Transplantations for Acute Leukemias and Myelodysplastic Syndromes. Biol Blood Marrow Transplant 2018; 24:2450-2458. [PMID: 30026107 DOI: 10.1016/j.bbmt.2018.07.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Accepted: 07/09/2018] [Indexed: 01/09/2023]
Abstract
Chronic graft-versus-host disease (cGVHD) is a major complication after stem cell transplantation (HSCT). Several randomized studies already demonstrated that anti-T lymphoglobulin (ATLG) is effective in preventing GVHD after myeloablative unrelated and HLA-identical sibling transplants. However, the issue of doses and the potential increase of relapses still remain unsolved. Here we report data on 190 patients with acute leukemia and myelodysplastic syndrome who underwent an unrelated HSCT with low-dose ATLG (15 to 30 mg/kg) given at an earlier timing (days -6 to -2). HSCT was performed from HLA 10/10 (n = 62, 33%), 9/10 (n = 91, 48%), 8/10 (n = 30, 16%), and <8/10 (n = 7, 4%) identical unrelated donor. Peripheral blood was the stem cell source in 42% (n = 80). Median follow-up was 51 months. Grades II to IV and III to IV acute GVHD were 26% and 9%, respectively, and 2-year overall and moderate to severe cGVHD were 23% and 14%, respectively. The 3-year incidences of relapse and nonrelapse mortality were 26% and 18%, respectively. The rates of 3-year overall survival (OS), disease-free survival (DFS), and GVHD-free and relapse-free survival (GRFS) were 60%, 56% and 44%, respectively. Factors such as younger donor, good performance status, and early disease were associated with better outcome in terms of OS, DFS, and GRFS. Our data indicate that doses of ATLG lower that those used in randomized clinical trials can be used for GVHD prevention, even in the adult setting, without clear increases in relapse and infections; these findings need to be further validated by a prospective randomized study.
Collapse
Affiliation(s)
- Francesca Bonifazi
- Department of Hematology "L. and A. Seràgnoli," University of Bologna, S. Orsola-Malpighi Hospital, Bologna, Italy.
| | - Jacopo Olivieri
- Department of Hematology, Transplant Unit and Cellular Therapies "C. Melzi" University Hospital, Udine, Italy
| | - Mariarosaria Sessa
- Department of Hematology "L. and A. Seràgnoli," University of Bologna, S. Orsola-Malpighi Hospital, Bologna, Italy
| | - Elisa Dan
- Department of Hematology "L. and A. Seràgnoli," University of Bologna, S. Orsola-Malpighi Hospital, Bologna, Italy
| | - Barbara Sinigaglia
- Department of Hematology "L. and A. Seràgnoli," University of Bologna, S. Orsola-Malpighi Hospital, Bologna, Italy
| | - Simonetta Rizzi
- Department of Hematology "L. and A. Seràgnoli," University of Bologna, S. Orsola-Malpighi Hospital, Bologna, Italy
| | - Maria Rosa Motta
- Department of Hematology "L. and A. Seràgnoli," University of Bologna, S. Orsola-Malpighi Hospital, Bologna, Italy
| | - Andrea Bontadini
- Department of Immunogenetics, S. Orsola-Malpighi Hospital, Bologna, Italy
| | - Francesca Ulbar
- Department of Hematology "L. and A. Seràgnoli," University of Bologna, S. Orsola-Malpighi Hospital, Bologna, Italy
| | - Valeria Giudice
- Apheresis Unit, University Hospital S. Orsola-Malpighi, Bologna, Italy
| | - Cristina Papayannidis
- Department of Hematology "L. and A. Seràgnoli," University of Bologna, S. Orsola-Malpighi Hospital, Bologna, Italy
| | - Antonio Curti
- Department of Hematology "L. and A. Seràgnoli," University of Bologna, S. Orsola-Malpighi Hospital, Bologna, Italy
| | - Angela Chiereghin
- Microbiology Unit, Laboratory of Virology, Department of Specialized, Experimental, and Diagnostic Medicine, St. Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy
| | - Tiziana Lazzarotto
- Microbiology Unit, Laboratory of Virology, Department of Specialized, Experimental, and Diagnostic Medicine, St. Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy
| | - Michele Cavo
- Department of Hematology "L. and A. Seràgnoli," University of Bologna, S. Orsola-Malpighi Hospital, Bologna, Italy
| | - Mario Arpinati
- Department of Hematology "L. and A. Seràgnoli," University of Bologna, S. Orsola-Malpighi Hospital, Bologna, Italy
| |
Collapse
|
21
|
Servais S, Baron F, Lechanteur C, Seidel L, Selleslag D, Maertens J, Baudoux E, Zachee P, Van Gelder M, Noens L, Kerre T, Lewalle P, Schroyens W, Ory A, Beguin Y. Infusion of bone marrow derived multipotent mesenchymal stromal cells for the treatment of steroid-refractory acute graft-versus-host disease: a multicenter prospective study. Oncotarget 2018; 9:20590-20604. [PMID: 29755674 PMCID: PMC5945536 DOI: 10.18632/oncotarget.25020] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Accepted: 03/17/2018] [Indexed: 12/12/2022] Open
Abstract
The prognosis of steroid-refractory acute graft-versus-host disease (aGVHD) remains poor and better treatments are urgently needed. Multipotent mesenchymal stromal cell (MSC)-based therapy emerged as a promising approach but response rates were highly variable across studies. We conducted a multicenter prospective study assessing the efficacy of 1-2 infusion(s) of cryopreserved, third-party donor bone marrow-derived MSCs for treating grade II-IV steroid-refractory or -dependent aGVHD in a series of 33 patients. MSCs were produced centrally and distributed to 8 hospitals throughout Belgium to be infused in 2 consecutive cohorts of patients receiving 1-2 or 3-4 × 106 MSCs/kg per dose, respectively. All patients received MSCs as the first rescue therapy after corticosteroids, with the exception for one patient who received prior treatment with mycophenolate mofetil (that was still ongoing by the time of MSC therapy). In these conditions, MSC therapy resulted in at least a partial response in 13 patients (40.6%) at day 30 and in 15 patients (46%) within 90 days after first MSC infusion. The corresponding complete response rates were 21.6% (7 patients) and 30% (10 patients), respectively. Only 5 patients achieved a sustained complete response, lasting for at least 1 month. The 1-year overall survival was 18.2% (95% CI: 8.82-37.5%). Higher response and survival rates were observed among patients receiving 3-4 × 106 MSCs/kg for first infusion, as compared with patients receiving 1-2 × 106 MSCs/ kg. Response and survival with MSC therapy for SR/SD-aGVHD remains to be optimized.
Collapse
Affiliation(s)
- Sophie Servais
- Department of Hematology, CHU of Liège, 4000 Liège, Belgium
- Laboratory of Cell and Gene Therapy, CHU of Liège, 4000 Liège, Belgium
| | - Frédéric Baron
- Department of Hematology, CHU of Liège, 4000 Liège, Belgium
- Laboratory of Cell and Gene Therapy, CHU of Liège, 4000 Liège, Belgium
| | - Chantal Lechanteur
- Department of Hematology, CHU of Liège, 4000 Liège, Belgium
- Laboratory of Cell and Gene Therapy, CHU of Liège, 4000 Liège, Belgium
| | - Laurence Seidel
- Department of biostatistics, SIMÉ, CHU of Liège, 4000 Liège, Belgium
| | | | - Johan Maertens
- Department of Hematology, AZ Gasthuisberg, 3000 Leuven, Belgium
| | - Etienne Baudoux
- Department of Hematology, CHU of Liège, 4000 Liège, Belgium
- Laboratory of Cell and Gene Therapy, CHU of Liège, 4000 Liège, Belgium
| | - Pierre Zachee
- Department of Hematology, ZNA Stuivenberg, 2060 Antwerp, Belgium
| | - Michel Van Gelder
- Department of Internal Medicine, Hematology Division, Maastricht University Medical Center, 6202 AZ Maastricht, The Nertherlands
| | - Lucien Noens
- Department of Hematology, UZ Gent, 9000 Ghent, Belgium
| | - Tessa Kerre
- Department of Hematology, UZ Gent, 9000 Ghent, Belgium
| | - Philippe Lewalle
- Department of Hematology, Institut Jules-Bordet, 1000 Brussels, Belgium
| | - Wilfried Schroyens
- Department of Hematology, Antwerp University Hospital, 2650 Edegem and University of Antwerp, 2610 Antwerp, Belgium
| | - Aurélie Ory
- Clinical Research Associate of the Belgian Hematology Society, CHU Sart-Tilman, 4000 Liège, Belgium
| | - Yves Beguin
- Department of Hematology, CHU of Liège, 4000 Liège, Belgium
- Laboratory of Cell and Gene Therapy, CHU of Liège, 4000 Liège, Belgium
| |
Collapse
|
22
|
Dillinger B, Ahmadi-Erber S, Soukup K, Halfmann A, Schrom S, Vanhove B, Steinberger P, Geyeregger R, Ladisch S, Dohnal AM. CD28 Blockade Ex Vivo Induces Alloantigen-Specific Immune Tolerance but Preserves T-Cell Pathogen Reactivity. Front Immunol 2017; 8:1152. [PMID: 28979262 PMCID: PMC5611377 DOI: 10.3389/fimmu.2017.01152] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 08/31/2017] [Indexed: 12/21/2022] Open
Abstract
Donor T-cells contribute to reconstitution of protective immunity after allogeneic hematopoietic stem cell transplantation (HSCT) but must acquire specific tolerance against recipient alloantigens to avoid life-threatening graft-versus-host disease (GvHD). Systemic immunosuppressive drugs may abrogate severe GvHD, but this also impedes memory responses to invading pathogens. Here, we tested whether ex vivo blockade of CD28 co-stimulation can enable selective T-cell tolerization to alloantigens by facilitating CD80/86-cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) signaling. Treatment of human allogeneic dendritic cell/T-cell co-cultures with a human CD28 blocking antibody fragment (α-huCD28) significantly abrogated subsequent allospecific immune responses, seen by decreased T-cell proliferation and of type 1 cytokine (IFN-γ and IL-2) expression. Allo-tolerization persisted after discontinuation of CD28 blockade and secondary alloantigen stimulation, as confirmed by enhanced CTLA-4 and PD-1 immune checkpoint signaling. However, T-cells retained reactivity to pathogens, supported by clonotyping of neo-primed and cross-reactive T-cells specific for Candida albicans or third-party antigens using deep sequencing analysis. In an MHC-mismatched murine model, we tolerized C57BL/6 T-cells by ex vivo exposure to a murine single chain Fv specific for CD28 (α-muCD28). Infusion of these cells, after α-muCD28 washout, into bone marrow-transplanted BALB/c mice caused allo-tolerance and did not induce GvHD-associated hepatic pathology. We conclude that selective CD28 blockade ex vivo can allow the generation of stably allo-tolerized T-cells that in turn do not induce graft-versus-host reactions while maintaining pathogen reactivity. Hence, CD28 co-stimulation blockade of donor T-cells may be a useful therapeutic approach to support the immune system after HSCT.
Collapse
Affiliation(s)
- Barbara Dillinger
- Tumor Immunology, Children's Cancer Research Institute (CCRI), St. Anna Kinderkrebsforschung e.V., Vienna, Austria
| | - Sarah Ahmadi-Erber
- Tumor Immunology, Children's Cancer Research Institute (CCRI), St. Anna Kinderkrebsforschung e.V., Vienna, Austria
| | - Klara Soukup
- Tumor Immunology, Children's Cancer Research Institute (CCRI), St. Anna Kinderkrebsforschung e.V., Vienna, Austria
| | - Angela Halfmann
- Tumor Immunology, Children's Cancer Research Institute (CCRI), St. Anna Kinderkrebsforschung e.V., Vienna, Austria
| | - Silke Schrom
- Tumor Immunology, Children's Cancer Research Institute (CCRI), St. Anna Kinderkrebsforschung e.V., Vienna, Austria
| | - Bernard Vanhove
- Centre de Recherche en Transplantation et Immunologie UMR 1064, INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France.,OSE Immunotherapeutics, Nantes, France
| | - Peter Steinberger
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Rene Geyeregger
- Clinical Cell Biology, Children's Cancer Research Institute (CCRI), St. Anna Kinderkrebsforschung e.V., Vienna, Austria.,Department of Pediatrics, Medical University of Vienna, Vienna, Austria
| | - Stephan Ladisch
- Center for Cancer and Immunology Research, Children's Research Institute, Children's National Medical Center, Washington, DC, United States
| | - Alexander Michael Dohnal
- Tumor Immunology, Children's Cancer Research Institute (CCRI), St. Anna Kinderkrebsforschung e.V., Vienna, Austria
| |
Collapse
|
23
|
Ehx G, Fransolet G, de Leval L, D'Hondt S, Lucas S, Hannon M, Delens L, Dubois S, Drion P, Beguin Y, Humblet-Baron S, Baron F. Azacytidine prevents experimental xenogeneic graft-versus-host disease without abrogating graft-versus-leukemia effects. Oncoimmunology 2017. [PMID: 28638744 DOI: 10.1080/2162402x.2017.1314425] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
The demethylating agent 5-azacytidine (AZA) has proven its efficacy in the treatment of myelodysplastic syndrome and acute myeloid leukemia. In addition, AZA can demethylate FOXP3 intron 1 (FOXP3i1) leading to the generation of regulatory T cells (Treg). Here, we investigated the impact of AZA on xenogeneic graft-vs.-host disease (xGVHD) and graft-vs.-leukemia effects in a humanized murine model of transplantation (human PBMCs-infused NSG mice), and described the impact of the drug on human T cells in vivo. We observed that AZA improved both survival and xGVHD scores. Further, AZA significantly decreased human T-cell proliferation as well as IFNγ and TNF-α serum levels, and reduced the expression of GRANZYME B and PERFORIN 1 by cytotoxic T cells. In addition, AZA significantly increased Treg frequency through hypomethylation of FOXP3i1 as well as increased Treg proliferation. The latter was subsequent to higher STAT5 signaling in Treg from AZA-treated mice, which resulted from higher IL-2 secretion by conventional T cells from AZA-treated mice itself secondary to demethylation of the IL-2 gene promoter by AZA. Importantly, Tregs harvested from AZA-treated mice were suppressive and stable over time since they persisted at high frequency in secondary transplant experiments. Finally, graft-vs.-leukemia effects (assessed by growth inhibition of THP-1 cells, transfected to express the luciferase gene) were not abrogated by AZA. In summary, our data demonstrate that AZA prevents xGVHD without abrogating graft-vs.-leukemia effects. These findings could serve as basis for further studies of GVHD prevention by AZA in acute myeloid leukemia patients offered an allogeneic transplantation.
Collapse
Affiliation(s)
- Grégory Ehx
- Groupe Interdisciplinaire de Génoprotéomique Appliquée (GIGA)-I3, University of Liège, Liège, Belgium
| | - Gilles Fransolet
- Groupe Interdisciplinaire de Génoprotéomique Appliquée (GIGA)-I3, University of Liège, Liège, Belgium
| | - Laurence de Leval
- Institute of Pathology, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Stéphanie D'Hondt
- de Duve Institute, Université catholique de Louvain, Brussels, Belgium
| | - Sophie Lucas
- de Duve Institute, Université catholique de Louvain, Brussels, Belgium
| | - Muriel Hannon
- Groupe Interdisciplinaire de Génoprotéomique Appliquée (GIGA)-I3, University of Liège, Liège, Belgium
| | - Loïc Delens
- Groupe Interdisciplinaire de Génoprotéomique Appliquée (GIGA)-I3, University of Liège, Liège, Belgium
| | - Sophie Dubois
- Groupe Interdisciplinaire de Génoprotéomique Appliquée (GIGA)-I3, University of Liège, Liège, Belgium
| | - Pierre Drion
- Experimental Surgery unit, GIGA & Credec, University of Liege, Liège, Belgium
| | - Yves Beguin
- Groupe Interdisciplinaire de Génoprotéomique Appliquée (GIGA)-I3, University of Liège, Liège, Belgium.,Department of Medicine, Division of Hematology, University of Liège, Liège, Belgium
| | - Stéphanie Humblet-Baron
- VIB Center for Brain & Disease Research, Leuven; KU Leuven, Department of Microbiology and Immunology, Leuven, Belgium
| | - Frédéric Baron
- Groupe Interdisciplinaire de Génoprotéomique Appliquée (GIGA)-I3, University of Liège, Liège, Belgium.,Department of Medicine, Division of Hematology, University of Liège, Liège, Belgium
| |
Collapse
|
24
|
Belle L, Fransolet G, Somja J, Binsfeld M, Delvenne P, Drion P, Hannon M, Beguin Y, Ehx G, Baron F. Limited Impact of Imatinib in a Murine Model of Sclerodermatous Chronic Graft-versus-Host Disease. PLoS One 2016; 11:e0167997. [PMID: 27942010 PMCID: PMC5152855 DOI: 10.1371/journal.pone.0167997] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Accepted: 11/23/2016] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Sclerodermatous chronic Graft-versus-Host Disease (scl-cGVHD) is one of the most severe form of cGVHD. The Platelet-derived Grotwth Factor (PDGF) and the Transforming Growth Factor-β (TGF-β) play a significant role in the fibrosing process occurring in scl-cGVHD. This prompted us to assess the impact of the PDGF-r and c-Abl tyrosine kinase inhibitor imatinib on scl-cGVHD. METHODS To assess the impact of imatinib on T cell subset proliferation in vivo, Balb/cJ recipient mice were lethally (7 Gy) irradiated and then injected with 10x106 bone marrow cells from B10.D2 mice on day 0. Fourteen days later, 70x106 carboxyfluorescein succinimidyl ester (CFSE)-labeled splenocytes from B10.D2 mice were infused and imatinib or sterile water was administered for 5 days. To induce severe scl-cGVHD, Balb/cJ mice were injected i.v. with 10.106 bone marrow cells and 70.106 splenocytes from B10.D2 donor mice after 7 Gy irradiation. Mice were then given sterile water or imatinib from day +7 after transplantation to the end of the experiment (day +52). RESULTS Imatinib decreased the proliferation of total T cells (P = 0.02), CD8+ T cells (P = 0.01), and of regulatory T cells (Tregs) (P = 0.02) in the spleen. In the severe scl-cGVHD model, imatinib-treated mice had significantly lower levels of PDGF-r phosphorylation than control mice on day 29 after transplantation (P = 0.008). However, scl-cGVHD scores were similar between vehicle- and imatinib-treated mice during the whole experiment, while there was a suggestion for less weight loss in imatinib-treated mice that reached statistical significance at day +52 following transplantation (P = 0.02). CONCLUSIONS Imatinib had a limited impact in murine scl-cGVHD despite significant inhibition of PDGF-r.
Collapse
Affiliation(s)
- Ludovic Belle
- Hematology Research Unit, GIGA-I³, University of Liège, Liège, Belgium
| | - Gilles Fransolet
- Hematology Research Unit, GIGA-I³, University of Liège, Liège, Belgium
- * E-mail:
| | - Joan Somja
- Department of Pathology, University of Liège, Liège, Belgium
| | - Marilène Binsfeld
- Hematology Research Unit, GIGA-I³, University of Liège, Liège, Belgium
| | | | | | - Muriel Hannon
- Hematology Research Unit, GIGA-I³, University of Liège, Liège, Belgium
| | - Yves Beguin
- Hematology Research Unit, GIGA-I³, University of Liège, Liège, Belgium
- Department of Medicine, Division of Hematology, CHU of Liège, Liège, Belgium
| | - Grégory Ehx
- Hematology Research Unit, GIGA-I³, University of Liège, Liège, Belgium
| | - Frédéric Baron
- Hematology Research Unit, GIGA-I³, University of Liège, Liège, Belgium
- Department of Medicine, Division of Hematology, CHU of Liège, Liège, Belgium
| |
Collapse
|