1
|
Fu L, Zhang J, Wang R, Yang B, Li H, Chen H, Ma J. Clinical characteristics of hepatitis-associated aplastic anemia in children. Ann Hematol 2024; 103:397-404. [PMID: 38082101 DOI: 10.1007/s00277-023-05566-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 11/22/2023] [Indexed: 01/23/2024]
Abstract
To understand the current situation of hepatitis-related aplastic anemia (HAAA) in children, we analyzed the patients with HAAA admitted to our hospital in the past 5 years to understand the disease characteristics and prognosis. The clinical data of patients with HAAA admitted to our hospital from February 2017 to May 2022 were retrospectively analyzed. A total of 81 patients with HAAA, 56 males and 25 females. The median onset age was 5.9 years. The median time from hepatitis to occurrence of hemocytopenia was 30 days, and the median follow-up time was 2.77 years. There were 23 cases (28.5%) of severe aplastic anemia (SAA), 50 cases of very severe aplastic anemia (VSAA), and 8 cases of non-severe aplastic anemia (NSAA). At the beginning of the disease, cytotoxic T lymphocyte (CTL) was higher than normal in 60% of patients, and the median CD4/CD8 ratio was 0.2. As of follow-up, 72 children survived, 4 were lost, and 5 died. Thirty-four cases were treated with immunosuppressive therapy (IST), with a median follow-up time of 0.97 years. The total reaction rate was 73.5% (25/34), the complete reaction rate was 67.6% (23/34), and the nonreaction rate was 26.5% (9/34). Multivariate analysis suggested that co-infection was an independent risk factor affecting the efficacy of IST at 6 months, with an OR value of 16.76, 95% CI (1.23, 227.95), P=0.034. No independent influencing factors were found at the end of follow-up. The proportion of CTL cells in peripheral blood of children with HAAA is relatively increased, and IST is effective in 73.5% of children. Co-infection may prolongs the time to response to IST.
Collapse
Affiliation(s)
- Lingling Fu
- Hematology Center, Beijing Key Laboratory of Pediatric Hematology Oncology; National Key Discipline of Pediatrics (Capital Medical University); Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045, China
| | - Jialu Zhang
- Hematology Center, Beijing Key Laboratory of Pediatric Hematology Oncology; National Key Discipline of Pediatrics (Capital Medical University); Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045, China
| | - Ruixin Wang
- Hematology Center, Beijing Key Laboratory of Pediatric Hematology Oncology; National Key Discipline of Pediatrics (Capital Medical University); Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045, China
| | - Bixi Yang
- Hematology Center, Beijing Key Laboratory of Pediatric Hematology Oncology; National Key Discipline of Pediatrics (Capital Medical University); Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045, China
| | - Hongmin Li
- Hematology Center, Beijing Key Laboratory of Pediatric Hematology Oncology; National Key Discipline of Pediatrics (Capital Medical University); Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045, China
| | - Hui Chen
- Hematologic Disease Laboratory, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, Beijing, 100045, China
| | - Jie Ma
- Hematology Center, Beijing Key Laboratory of Pediatric Hematology Oncology; National Key Discipline of Pediatrics (Capital Medical University); Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045, China.
| |
Collapse
|
2
|
Zhang X, Yang W, Yang D, Wei J, Zhang P, Feng S, Jiang E, Zhang L, He Y, Zhang F, Han M. Comparison of hematopoietic stem cell transplantation and immunosuppressive therapy as the first-line treatment option for patients with severe hepatitis−associated aplastic anemia. Front Immunol 2023; 14:1146997. [PMID: 37006284 PMCID: PMC10063874 DOI: 10.3389/fimmu.2023.1146997] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 03/06/2023] [Indexed: 03/19/2023] Open
Abstract
Hepatitis-associated aplastic anemia (HAAA) is a rare variant of acquired aplastic anemia characterized with a syndrome of bone marrow failure after hepatitis. We retrospectively analyzed the outcomes of consecutive severe HAAA patients who received immunosuppressive therapy (IST, n = 70), matched-sibling donor hematopoietic stem cell transplantation (MSD-HSCT, n = 26) or haploidentical-donor (HID) HSCT (n = 11) as the first-line treatment. In the IST group, the hematologic response (HR) rate was 55.71% at 6 months. In contrast, HSCT recipients exhibited significantly more rapid and sustained hematopoiesis (HR 76.92%, 96.15% and 96.15% at 3, 6 and 12months, respectively). The 5-year overall survival (OS) was not different among IST (83.7 ± 4.9%), MSD-HSCT (93.3 ± 6.4%) and HID-HSCT group (80.8 ± 12.3%). Compared with IST, MSD and HID-HSCT demonstrated a trend of superiority in the estimated 5-year failure-free survival rates (93.3 ± 6.4% vs 64.3 ± 6.0%, p = 0.05; 80.8 ± 12.3% vs 64.3 ± 6.0%, p = 0.57). In subsequent stratified analysis on age, we found that HID-HSCT showed its efficacy and safety among young patients. In sum, MSD-HSCT remains first-line treatment choice for HAAA, whereas HID-HSCT represents an alternative treatment choice in addition to IST for young patients (< 40 years) without a matched sibling donor.
Collapse
Affiliation(s)
- Xiaoyu Zhang
- Stem Cell Transplantation Center, State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Wenrui Yang
- Tianjin Institutes of Health Science, Tianjin, China
- Anemia Therapeutic Center, State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Donglin Yang
- Stem Cell Transplantation Center, State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Jialin Wei
- Tianjin Institutes of Health Science, Tianjin, China
- Anemia Therapeutic Center, State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Ping Zhang
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, United States
| | - Sizhou Feng
- Stem Cell Transplantation Center, State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Erlie Jiang
- Stem Cell Transplantation Center, State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Li Zhang
- Tianjin Institutes of Health Science, Tianjin, China
- Anemia Therapeutic Center, State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- *Correspondence: Li Zhang, ; Yi He,
| | - Yi He
- Stem Cell Transplantation Center, State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Fengkui Zhang
- Tianjin Institutes of Health Science, Tianjin, China
- Anemia Therapeutic Center, State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Mingzhe Han
- Stem Cell Transplantation Center, State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| |
Collapse
|
3
|
Zhang WJ, Wu LQ, Wang J, Lin SY, Wang B. Epstein–Barr-virus-associated hepatitis with aplastic anemia: A case report. World J Clin Cases 2022; 10:8242-8248. [PMID: 36159516 PMCID: PMC9403690 DOI: 10.12998/wjcc.v10.i23.8242] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 04/19/2022] [Accepted: 06/27/2022] [Indexed: 02/06/2023] Open
Abstract
ABSTRACT
BACKGROUND
Hepatitis-associated aplastic anemia (HAAA) is a rare condition. Patients with HAAA usually present with acute hepatitis, jaundice and significantly increased transaminase. After 1–2 mo, hepatitis gradually improves, but progressive hemocytopenia, bone marrow hematopoietic failure, and severe or extremely severe aplastic anemia are manifest. Most cases of HAAA are fulminant and usually lethal if left untreated. The literature on Epstein–Barr virus (EBV)-associated HAAA is sparse.
CASE SUMMARY
We report a 30-year-old man who was admitted to our hospital because of pale yellow urine and skin with a simultaneous decrease in peripheral blood ternary cells. We made a diagnosis of EBV-associated HAAA. The treatment strategy for this patient included eltrombopag, an immunosuppressive regimen of rabbit anti-human thymocyte immunoglobulin, cyclosporine, and supportive care. The patient was discharged in normal physical condition after five months. A hemogram performed on follow-up revealed that he had achieved a complete response.
CONCLUSION
Eltrombopag plus anti-thymocyte globubin and cyclosporine may be a therapeutic option for EBV-associated HAAA.Larger studies are warranted to confirm.
Collapse
Affiliation(s)
- Wan-Jun Zhang
- Department of Hematology, The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou 310006, Zhejiang Province, China
| | - Li-Qiang Wu
- Department of Hematology, The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou 310006, Zhejiang Province, China
| | - Jun Wang
- Department of Hematology, The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou 310006, Zhejiang Province, China
| | - Sheng-Yun Lin
- Department of Hematology, The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou 310006, Zhejiang Province, China
| | - Bo Wang
- Department of Hematology, The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou 310006, Zhejiang Province, China
| |
Collapse
|
4
|
Time and residual hematopoiesis are crucial for PNH clones escape in hepatitis-associated aplastic anemia. Ann Hematol 2021; 100:2435-2441. [PMID: 34269836 DOI: 10.1007/s00277-021-04553-5] [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: 07/01/2020] [Accepted: 05/02/2021] [Indexed: 10/20/2022]
Abstract
The presence of paroxysmal nocturnal hemoglobinuria (PNH) clones in aplastic anemia (AA) suggests immunopathogenesis, but when and how PNH clones emerge and proliferate are unclear. Hepatitis-associated aplastic anemia (HAAA) is a special variant of AA, contrarily to idiopathic AA, in HAAA the trigger for immune activation is clearer and represented by the hepatitis and thus serves as a good model for studying PNH clones. Ninety HAAA patients were enrolled, including 61 males and 29 females (median age 21 years). Four hundred three of idiopathic AA have been included as controls. The median time from hepatitis to cytopenia was 50 days (range 0-180 days) and from cytopenia to AA diagnosis was 26 days (range 2-370 days). PNH clones were detected in 8 HAAA patients (8.9%) at diagnosis and in 73 patients with idiopathic AA (IAA) (18.1%). PNH cells accounted for 4.2% (1.09-12.33%) of red cells and/or granulocytes and were more likely to be detected in patients with longer disease history and less severe disease. During follow-up, the cumulative incidence of PNH clones in HAAA increased to 18.9% (17/90). Nine HAAA patients newly developed PNH clones, including six immunosuppressive therapy (IST) nonresponders. The clone size was mostly stable during follow-up, and only 2 of 14 patients showed increased clone size without proof of hemolysis. In conclusion, PNH clones were infrequent in newly diagnosed HAAA, but their frequency increased to one that was similar to the IAA frequency during follow-up. These results suggest that the PNH clone selection/expansion process is dynamic and takes time to establish, confirming that retesting for PNH clones during follow-up is crucial.
Collapse
|
5
|
Abstract
BACKGROUND Seronegative autoimmune hepatitis (AIH) is a diagnostic challenge with unclear prognosis. This study describes the features and outcomes of seronegative AIH in children. PATIENTS AND METHODS Patients under 18 years of age, who had been diagnosed with AIH between April 2014 and April 2020, were retrospectively evaluated. Seronegative AIH was identified by the absence of the three conventional non-organ-specific autoantibodies (anti-nuclear antibody [ANA], anti-smooth muscle antibody [ASMA], and anti-liver kidney microsomal [anti-LKM] type 1 antibody), alongside the characteristic AIH liver histopathology and a positive response to immunosuppressive therapy in the absence of other liver diseases. RESULTS The study included 54 patients with AIH. 15 (27.77%) were seronegative at the time of diagnosis. 13 of the 15 seronegative patients presented with acute hepatitis or acute liver failure (ALF). Mean follow-up duration was 27.48 months in seronegative patients. Two seronegative patients had lymphocytopenia on admission, and, although the liver disease improved after corticosteroid treatment, they developed aplastic anemia (AA). Other seronegative patients responded well to immunosuppressive therapy. CONCLUSIONS Patients with seronegative AIH present frequently with acute hepatitis or ALF. AIH diagnosis can be confirmed by observing the effects of corticosteroid therapy in seronegative patients with characteristic AIH liver histopathological features. However, the presence of lymphocytopenia in seronegative patients is a sign of bone marrow failure.
Collapse
|
6
|
Fang L, Meikuang L, Ye G, Xiaojuan C, Wenyu Y, Min R, Lixian C, Weiqiang W, Zhibo H, Zhongchao H, Xiaofan Z. Successful Treatment of a 19-Month-Old Boy with Hepatitis Associated Aplastic Anemia by Infusion of Umbilical Cord-Derived Mesenchymal Stromal Cells: A Case Report. Cell Transplant 2021; 30:963689720977144. [PMID: 33525921 PMCID: PMC7863554 DOI: 10.1177/0963689720977144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Here we presented a case of a 19-month-old boy who developed severe aplastic anemia postacute hepatitis. He was treated successfully with the umbilical cord-derived mesenchymal stromal cells (UC-MSCs) infusion and cyclosporine A (CsA). The boy achieved both hematopoietic recovery and normal lymphocyte proportion. So far, his condition still remains stable. To our knowledge, there is a rare previous report on the utility of MSCs infusion for the treatment of hepatitis-associated aplastic anemia (HAAA). Considering the efficacy, safety, and strong operability, particularly for pediatric patient, the infusion of UC-MSCs combined with CsA could be an effective alternative for the treatment of HAAA.
Collapse
Affiliation(s)
- Liu Fang
- Institute of Hematology and Blood Diseases Hospital, 12501Peking Union Medical College and Chinese Academy of Medical Sciences, Tianjin, China
| | - Lim Meikuang
- National Engineering Research Center of Cell Products, Tianjin AmCellGene Engineering Co., Ltd, Tianjin, China
| | - Guo Ye
- Institute of Hematology and Blood Diseases Hospital, 12501Peking Union Medical College and Chinese Academy of Medical Sciences, Tianjin, China
| | - Chen Xiaojuan
- Institute of Hematology and Blood Diseases Hospital, 12501Peking Union Medical College and Chinese Academy of Medical Sciences, Tianjin, China
| | - Yang Wenyu
- Institute of Hematology and Blood Diseases Hospital, 12501Peking Union Medical College and Chinese Academy of Medical Sciences, Tianjin, China
| | - Ruan Min
- Institute of Hematology and Blood Diseases Hospital, 12501Peking Union Medical College and Chinese Academy of Medical Sciences, Tianjin, China
| | - Chang Lixian
- Institute of Hematology and Blood Diseases Hospital, 12501Peking Union Medical College and Chinese Academy of Medical Sciences, Tianjin, China
| | - Wang Weiqiang
- National Engineering Research Center of Cell Products, Tianjin AmCellGene Engineering Co., Ltd, Tianjin, China
| | - Han Zhibo
- Institute of Hematology and Blood Diseases Hospital, 12501Peking Union Medical College and Chinese Academy of Medical Sciences, Tianjin, China.,National Engineering Research Center of Cell Products, Tianjin AmCellGene Engineering Co., Ltd, Tianjin, China
| | - Han Zhongchao
- Institute of Hematology and Blood Diseases Hospital, 12501Peking Union Medical College and Chinese Academy of Medical Sciences, Tianjin, China.,National Engineering Research Center of Cell Products, Tianjin AmCellGene Engineering Co., Ltd, Tianjin, China
| | - Zhu Xiaofan
- Institute of Hematology and Blood Diseases Hospital, 12501Peking Union Medical College and Chinese Academy of Medical Sciences, Tianjin, China
| |
Collapse
|
7
|
Alshaibani A, Dufour C, Risitano A, de Latour R, Aljurf M. Hepatitis-associated aplastic anemia. Hematol Oncol Stem Cell Ther 2020; 15:8-12. [PMID: 33197413 DOI: 10.1016/j.hemonc.2020.10.001] [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: 10/15/2020] [Accepted: 10/24/2020] [Indexed: 02/08/2023] Open
Abstract
Hepatitis-associated aplastic anemia (HAAA) is a rare illness, characterized by onset of pancytopenia with a hypoplastic bone marrow that traditionally occurs within 6 months of an increase in serum aminotransferases. HAAA is observed in 1% to 5% of all newly diagnosed cases of acquired aplastic anemia. Several hepatitis viruses have been linked to the disease, but in many cases no specific virus is detected. The exact pathophysiology is unknown; however, immune destruction of hematopoietic stem cells is believed to be the underlying mechanism. HAAA is a potentially lethal disease if left untreated. Management includes immunosuppression with antithymocyte globulin and cyclosporine and allogeneic hematopoietic stem cell transplantation.
Collapse
Affiliation(s)
- Alfadel Alshaibani
- King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia.
| | - Carlo Dufour
- Hematology-Oncology-HSCT Pole, G.Gaslini IRCCS Children Hospital, Genova, Italy.
| | - Antonio Risitano
- Department of Clinical Medicine and Surgery, Bone Marrow Transplant Center, Federico II University of Naples, Naples, Italy.
| | - Regis de Latour
- Saint Louis Hospital, Paris Diderot University, Paris, France.
| | - Mahmoud Aljurf
- King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia.
| |
Collapse
|
8
|
Chapin CA, Alonso EM. Reply. J Pediatr 2019; 214:244-245. [PMID: 31351681 DOI: 10.1016/j.jpeds.2019.06.055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 06/21/2019] [Indexed: 11/19/2022]
Affiliation(s)
- Catherine A Chapin
- Northwestern University Feinberg School of Medicine and Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois
| | - Estella M Alonso
- Northwestern University Feinberg School of Medicine and Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois
| |
Collapse
|
9
|
Chapin CA, Burn T, Meijome T, Loomes KM, Melin-Aldana H, Kreiger PA, Whitington PF, Behrens EM, Alonso EM. Indeterminate pediatric acute liver failure is uniquely characterized by a CD103 + CD8 + T-cell infiltrate. Hepatology 2018; 68:1087-1100. [PMID: 29603342 DOI: 10.1002/hep.29901] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 02/28/2018] [Accepted: 03/23/2018] [Indexed: 12/16/2022]
Abstract
UNLABELLED The cause of pediatric acute liver failure (PALF) is unknown in up to 40% of cases. Evidence suggests that aberrant immune system activation may play a role. We hypothesized that indeterminate PALF cases would exhibit a unique pattern of hepatic inflammation. This was a retrospective and prospective study of PALF cases due to indeterminate (iPALF), autoimmune hepatitis, or known diagnosis (dPALF) etiology. Liver tissue sections were stained with immunohistochemical markers for cytotoxic T-cells (cluster of differentiation 8 [CD8]), perforin, and tissue resident memory T-cells (CD103) and scored as minimal, moderate, or dense. Lymphocytes were isolated from liver tissue for T-cell receptor beta sequencing and flow-cytometric studies. Thirty-three iPALF, 9 autoimmune hepatitis, and 14 dPALF cases were included. Dense hepatic infiltrates of CD8+ T-cells were found in 27 (82%) iPALF cases compared to 1 (7%) dPALF case (P < 0.0001). Perforin staining was dense or moderate in 19 (73%) of 26 iPALF cases compared to minimal in all 7 dPALF cases (P = 0.004); 16 (62%) of 26 iPALF cases had dense CD103 staining compared to none of the 6 dPALF cases (P = 0.001). T-cell receptor beta sequencing of iPALF cases demonstrated increased clonality compared to dPALF and control cases. Flow cytometry and immunohistochemistry revealed that iPALF intrahepatic leukocytes were predominantly tissue resident memory CD8+ T-cells. CONCLUSION Indeterminate PALF is characterized by a dense CD8+ T-cell hepatic infiltrate consistent with expansion of a tissue resident memory T-cell phenotype; CD8+ T-cells are a biomarker of immune dysregulation in iPALF and may be used to better identify and define this group. (Hepatology 2018).
Collapse
Affiliation(s)
- Catherine A Chapin
- Department of Pediatrics, Northwestern University, Feinberg School of Medicine, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL
| | - Thomas Burn
- Department of Pediatrics, University of Pennsylvania, Perelman School of Medicine, The Children's Hospital of Philadelphia, Philadelphia, PA
| | - Tomas Meijome
- Department of Pediatrics, University of Pennsylvania, Perelman School of Medicine, The Children's Hospital of Philadelphia, Philadelphia, PA
| | - Kathleen M Loomes
- Department of Pediatrics, University of Pennsylvania, Perelman School of Medicine, The Children's Hospital of Philadelphia, Philadelphia, PA
| | - Hector Melin-Aldana
- Department of Pathology and Laboratory Medicine, Northwestern University, Feinberg School of Medicine, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL
| | - Portia A Kreiger
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Perelman School of Medicine, The Children's Hospital of Philadelphia, Philadelphia, PA
| | - Peter F Whitington
- Department of Pediatrics, Northwestern University, Feinberg School of Medicine, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL
| | - Edward M Behrens
- Department of Pediatrics, University of Pennsylvania, Perelman School of Medicine, The Children's Hospital of Philadelphia, Philadelphia, PA
| | - Estella M Alonso
- Department of Pediatrics, Northwestern University, Feinberg School of Medicine, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL
| |
Collapse
|
10
|
Seronegative autoimmune hepatitis in children: Spectrum of disorders. Dig Liver Dis 2016; 48:785-91. [PMID: 27079745 DOI: 10.1016/j.dld.2016.03.015] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Revised: 03/06/2016] [Accepted: 03/21/2016] [Indexed: 12/11/2022]
Abstract
BACKGROUND A few children with acute or chronic liver disease display histological features compatible with autoimmune hepatitis, but lack specific serological markers. AIM To describe features, management and outcome of childhood seronegative autoimmune hepatitis. METHODS From 1988 to 2010, 38 children were included under the following criteria: negative virological studies, no serum autoantibodies, exclusion of other causes of liver diseases, and liver histology compatible with autoimmune hepatitis. RESULTS Four groups were identified: (1) 12 with increased serum gamma globulin concentrations; (2) 10 with normal or low serum gamma globulins and no combined blood disease; (3) 10 with combined aplastic anemia; and (4) 6 with peripheral thrombocytopenia with/without neutropenia. Immunosuppressive treatment was associated with aminotransferases normalization in all but one child who required liver transplantation. Relapses occurred in 10 children. Lymphocytopenia was found at the time of the diagnosis of hepatitis in 13 children, 12 in groups 3 or 4. All 38 children are alive after 4-17 years, 18 still under immunosuppression. CONCLUSIONS Childhood seronegative autoimmune hepatitis includes a spectrum of disorders. Early liver histology is recommended and, if compatible with autoimmune hepatitis, immunosuppressive treatment should be started. Initial lymphocytopenia may indicate future hematological complication.
Collapse
|
11
|
Yang WR, Jing LP, Zhou K, Peng GX, Li Y, Ye L, Li Y, Li JP, Fan HH, Song L, Zhao X, Yang Y, Zhang FK, Zhang L. [Hepatitis-associated aplastic anaemia: clinical characteristics and immunosuppressive therapy outcomes]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2016; 37:399-404. [PMID: 27210875 PMCID: PMC7348316 DOI: 10.3760/cma.j.issn.0253-2727.2016.05.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Indexed: 01/01/2023]
Abstract
OBJECTIVE To analyze the clinical characteristics and to evaluate immunosuppressive therapy (IST) response and survival in hepatitis-associated aplastic anemia (HAAA). METHODS We retrospectively analyzed clinical characteristics, IST response, long-term survival and clonal evolution in 41 HAAA patients, and compared those with age and bone marrow failure matched idiopathic aplastic anemia (IAA) patients. RESULTS The prevalence of HAAA among cases of SAA was 4.34% (41/944). The proportion of VSAA in HAAA cases was significantly higher than IAA (65.9% vs 39.4%, P=0.001). There was no significant difference in the prevalence of hemorrhage and infections between HAAA and IAA patients, but the duration of infection persistence in HAAA group was much longer than IAA group [21 (4-100) d vs 13 (3-139) d, P=0.048]. The absolute counts of CD3(+) T-cell, CD3(+)CD4(+)T-cell, CD3(+)CD8(+)T-cell and ratio of CD4(+) T-cell/CD8(+) T-cell in HAAA were significant lower than that in IAA patients. However, the percentage of CD3(+)CD8(+)T-cell in HAAA was significant higher than that in IAA (P <0.05). The total response in HAAA and IAA patients treated with IST were 34.1% vs 34.1% (P=1.000), 56.1% vs 53.7% (P=0.787), and 73.2% vs 68.3% (P=0.558) at 3, 6, 12 months after IST, respectively. There were no significant difference in 5-year overall survival and event-free survival between HAAA and IAA patients (90% vs 87.1%, P=0.700; 71.9% vs 62.4%, P=0.450). CONCLUSION HAAA was a rare distinct variant of aplastic anemia with more severe bone marrow failure and more severe imbalance of the T cell immune system than IAA. Treatment outcomes were comparable in patients with HAAA and IAA.
Collapse
Affiliation(s)
- W R Yang
- Institute of Hematology and Blood Diseases Hospital, CAMS & PUMC, Tianjin 300020, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Babushok DV, Grignon A, Li Y, Atienza J, Xie HM, Lam H, Hartung H, Bessler M, Olson TS. Disrupted lymphocyte homeostasis in hepatitis-associated acquired aplastic anemia is associated with short telomeres. Am J Hematol 2016; 91:243-7. [PMID: 26615915 PMCID: PMC4724330 DOI: 10.1002/ajh.24256] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Accepted: 11/24/2015] [Indexed: 12/13/2022]
Abstract
Hepatitis‐associated aplastic anemia (HAA) is a variant of acquired aplastic anemia (AA) in which immune‐mediated bone marrow failure (BMF) develops following an acute episode of seronegative hepatitis. Dyskeratosis congenita (DC) is an inherited BMF syndrome characterized by the presence of short telomeres, mucocutaneous abnormalities, and cancer predisposition. While both conditions may cause BMF and hepatic impairment, therapeutic approaches are distinct, making it imperative to establish the correct diagnosis. In clinical practice, lymphocyte telomere lengths (TL) are used as a first‐line screen to rule out inherited telomeropathies before initiating treatment for AA. To evaluate the reliability of TL in the HAA population, we performed a retrospective analysis of TL in 10 consecutively enrolled HAA patients compared to 19 patients with idiopathic AA (IAA). HAA patients had significantly shorter telomeres than IAA patients (P = 0.009), including four patients with TL at or below the 1st percentile for age‐matched controls. HAA patients had no clinical features of DC and did not carry disease‐causing mutations in known genes associated with inherited telomere disorders. Instead, short TLs were significantly correlated with severe lymphopenia and skewed lymphocyte subsets, features characteristic of HAA. Our results indicate the importance of caution in the interpretation of TL measurements in HAA, because, in this patient population, short telomeres have limited specificity. Am. J. Hematol. 91:243–247, 2016. © 2015 The Authors. American Journal of Hematology Published by Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Daria V. Babushok
- Comprehensive Bone Marrow Failure Center, Division of Hematology, Department of PediatricsChildren's Hospital of PhiladelphiaPhiladelphia Pennsylvania
- Division of Hematology–Oncology, Department of MedicineHospital of the University of PennsylvaniaPhiladelphia Pennsylvania
| | - Anne‐Laure Grignon
- Comprehensive Bone Marrow Failure Center, Division of Hematology, Department of PediatricsChildren's Hospital of PhiladelphiaPhiladelphia Pennsylvania
| | - Yimei Li
- Department of Biostatistics and EpidemiologyPerelman School of Medicine at the University of PennsylvaniaPhiladelphia Pennsylvania
| | - Jamie Atienza
- Comprehensive Bone Marrow Failure Center, Division of Hematology, Department of PediatricsChildren's Hospital of PhiladelphiaPhiladelphia Pennsylvania
| | - Hongbo M. Xie
- Division of Health and Biomedical InformaticsChildren's Hospital of PhiladelphiaPhiladelphia Pennsylvania
| | - Ho‐Sun Lam
- Comprehensive Bone Marrow Failure Center, Division of Hematology, Department of PediatricsChildren's Hospital of PhiladelphiaPhiladelphia Pennsylvania
| | - Helge Hartung
- Comprehensive Bone Marrow Failure Center, Division of Hematology, Department of PediatricsChildren's Hospital of PhiladelphiaPhiladelphia Pennsylvania
| | - Monica Bessler
- Comprehensive Bone Marrow Failure Center, Division of Hematology, Department of PediatricsChildren's Hospital of PhiladelphiaPhiladelphia Pennsylvania
| | - Timothy S. Olson
- Comprehensive Bone Marrow Failure Center, Division of Hematology, Department of PediatricsChildren's Hospital of PhiladelphiaPhiladelphia Pennsylvania
- Division of Oncology, Department of PediatricsChildren's Hospital of Philadelphia and University of PennsylvaniaPhiladelphia Pennsylvania
| |
Collapse
|
13
|
Ahrenhoerster LS, Leuthner TC, Tate ER, Lakatos PA, Laiosa MD. Developmental exposure to 2,3,7,8 tetrachlorodibenzo-p-dioxin attenuates later-life Notch1-mediated T cell development and leukemogenesis. Toxicol Appl Pharmacol 2015; 283:99-108. [PMID: 25585350 DOI: 10.1016/j.taap.2014.12.017] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Revised: 11/26/2014] [Accepted: 12/31/2014] [Indexed: 01/04/2023]
Abstract
Over half of T cell acute lymphoblastic leukemia (T-ALL) patients have activating mutations in the Notch gene. Moreover, the contaminant 2,3,7,8 tetrachlorodibenzo-p-dioxin (TCDD) is a known carcinogen that mediates its toxicity through the aryl hydrocarbon receptor (AHR), and crosstalk between activated AHR and Notch signaling pathways has previously been observed. Given the importance of Notch signaling in thymocyte development and T-ALL disease progression, we hypothesized that the activated AHR potentiates disease initiation and progression in an in vivo model of Notch1-induced thymoma. This hypothesis was tested utilizing adult and developmental exposure paradigms to TCDD in mice expressing a constitutively active Notch1 transgene (Notch(ICN-TG)). Following exposure of adult Notch(ICN-TG) mice to a single high dose of TCDD, we observed a significant increase in the efficiency of CD8 thymocyte generation. We next exposed pregnant mice to 3μg/kg of TCDD throughout gestation and lactation to elucidate effects of developmental AHR activation on later-life T cell development and T-ALL-like thymoma susceptibility induced by Notch1. We found that the vehicle-exposed Notch(ICN-TG) offspring have a peripheral T cell pool heavily biased toward the CD4 lineage, while TCDD-exposed Notch(ICN-TG) offspring were biased toward the CD8 lineage. Furthermore, while the vehicle-exposed NotchICN-TG mice showed increased splenomegaly and B to T cell ratios indicative of disease, mice developmentally exposed to TCDD were largely protected from disease. These studies support a model where developmental AHR activation attenuates later-life Notch1-dependent impacts on thymocyte development and disease progression.
Collapse
Affiliation(s)
- Lori S Ahrenhoerster
- Joseph J. Zilber School of Public Health, University of Wisconsin-Milwaukee, Milwaukee, WI 53211, United States
| | - Tess C Leuthner
- Joseph J. Zilber School of Public Health, University of Wisconsin-Milwaukee, Milwaukee, WI 53211, United States
| | - Everett R Tate
- Joseph J. Zilber School of Public Health, University of Wisconsin-Milwaukee, Milwaukee, WI 53211, United States
| | - Peter A Lakatos
- Joseph J. Zilber School of Public Health, University of Wisconsin-Milwaukee, Milwaukee, WI 53211, United States
| | - Michael D Laiosa
- Joseph J. Zilber School of Public Health, University of Wisconsin-Milwaukee, Milwaukee, WI 53211, United States.
| |
Collapse
|
14
|
McKenzie RB, Berquist WE, Nadeau KC, Louie CY, Chen SF, Sibley RK, Glader BE, Wong WB, Hofmann LV, Esquivel CO, Cox KL. Novel protocol including liver biopsy to identify and treat CD8+ T-cell predominant acute hepatitis and liver failure. Pediatr Transplant 2014; 18:503-9. [PMID: 24930635 DOI: 10.1111/petr.12296] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/24/2014] [Indexed: 12/21/2022]
Abstract
In the majority of children with ALF, the etiology is unknown and liver transplantation is often needed for survival. A patient case prompted us to consider that immune dysregulation may be the cause of indeterminate acute hepatitis and liver failure in children. Our study includes nine pediatric patients treated under a multidisciplinary clinical protocol to identify and treat immune-mediated acute liver injury. Patients with evidence of inflammation and no active infection on biopsy received treatment with intravenous immune globulin and methylprednisolone. Seven patients had at least one positive immune marker before or after treatment. All patients had a CD8+ T-cell predominant liver injury that completely or partially responded to immune therapy. Five of the nine patients recovered liver function and did not require liver transplantation. Three of these patients subsequently developed bone marrow failure and were treated with either immunosuppression or stem cell transplant. This series highlights the importance of this tissue-based approach to diagnosis and treatment that may improve transplant-free survival. Further research is necessary to better characterize the immune injury and to predict the subset of patients at risk for bone marrow failure who may benefit from earlier and stronger immunosuppressive therapy.
Collapse
Affiliation(s)
- Rebecca B McKenzie
- Division of Gastroenterology, Hepatology & Nutrition, Department of Pediatrics, Stanford School of Medicine, Stanford, CA, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
The clinical and immune characteristics of patients with hepatitis-associated aplastic anemia in China. PLoS One 2014; 9:e98142. [PMID: 24845454 PMCID: PMC4028298 DOI: 10.1371/journal.pone.0098142] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Accepted: 04/29/2014] [Indexed: 01/10/2023] Open
Abstract
Hepatitis-associated aplastic anemia (HAAA) is a variant of severe aplastic anemia (SAA) in which bone marrow failure follows an acute attack of hepatitis. Its pathogenesis is poorly understood. We investigated the prevalence of HAAA among cases of newly diagnosed SAA presenting to our hospital between January 1998 and February 2013, and analyzed the clinical and immune characteristics of HAAA and non-hepatitis-associated SAA (non-HASAA) patients. The prevalence of HAAA among cases of SAA was 3.8% (36/949), and the majority of patients (33/36) were seronegative for a known hepatitis virus. Compared with non-HASAA patients, HAAA patients had a larger proportion of CD8+ T cells, a lower ratio of CD4+/CD8+ T cells, and a smaller proportion of CD4+CD25+ regulatory T cells. There was no significant difference in peripheral blood count, bone marrow cellularity, or the number of blood transfusions received between HAAA and non-HASAA patients. HAAA patients had a higher early infection rate and more infection-related mortality in the first 2 years after diagnosis than non-HASAA patients, and their 2-year survival rate was lower. The results demonstrate that HAAA patients have a more severe T cell imbalance and a poorer prognosis than non-HASAA patients.
Collapse
|
16
|
de Carvalho LG, Marchevsky RS, dos Santos DRL, de Oliveira JM, de Paula VS, Lopes LM, Van der Poel WHM, González JE, Munné MS, Moran J, Cajaraville ACRA, Pelajo-Machado M, Cruz OG, Pinto MA. Infection by Brazilian and Dutch swine hepatitis E virus strains induces haematological changes in Macaca fascicularis. BMC Infect Dis 2013; 13:495. [PMID: 24148233 PMCID: PMC3870956 DOI: 10.1186/1471-2334-13-495] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2013] [Accepted: 10/18/2013] [Indexed: 01/29/2023] Open
Abstract
BACKGROUND Hepatitis E virus (HEV) has been described as an emerging pathogen in Brazil and seems to be widely disseminated among swine herds. An autochthonous human case of acute hepatitis E was recently reported. To obtain a better understanding of the phenotypic profiles of both human and swine HEV strains, a experimental study was conducted using the animal model, Macaca fascicularis. METHODS Six cynomolgus monkeys (Macaca fascicularis) were inoculated intravenously with swine HEV genotype 3 that was isolated from naturally and experimentally infected pigs in Brazil and the Netherlands. Two other monkeys were inoculated with HEV genotype 3 that was recovered from Brazilian and Argentinean patients with locally acquired acute and fulminant hepatitis E. The haematological, biochemical, and virological parameters of all animals were monitored for 67 days. RESULTS Subclinical hepatitis was observed in all monkeys after inoculation with HEV genotype 3 that was recovered from the infected swine and human patients. HEV RNA was detected in the serum and/or faeces of 6 out of the 8 cynomolgus monkeys between 5 and 53 days after inoculation. The mild inflammation of liver tissues and elevations of discrete liver enzymes were observed. Seroconversions to anti-HEV IgM and/or IgG were detected in 7 animals. Reactivities to anti-HEV IgA were also detected in the salivary samples of 3 animals. Interestingly, all of the infected monkeys showed severe lymphopenia and a trend toward monocytosis, which coincided with elevations in alanine aminotransferase and antibody titres. CONCLUSIONS The ability of HEV to cross the species barrier was confirmed for both the swine (Brazilian and Dutch) and human (Argentinean) strains, thus reinforcing the zoonotic risk of hepatitis E in South America. Cynomolgus monkeys that were infected with HEV genotype 3 developed subclinical hepatitis that was associated with haematological changes. Haematological approaches should be considered in future studies of HEV infection.
Collapse
Affiliation(s)
- Lilian G de Carvalho
- Centre for Laboratory Animal Breeding, Department of Primatology, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
- Laboratory of Technological Development in Virology, Oswaldo Cruz Institute/Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Renato S Marchevsky
- Laboratory of Neurovirulence, Institute of Technology on Immunobiologicals, Bio-Manguinhos, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Debora RL dos Santos
- Laboratory of Veterinary Viruses, Department of Veterinary Microbiology and Immunology, UFRRJ, Rio de Janeiro, Brazil
| | - Jaqueline M de Oliveira
- Laboratory of Technological Development in Virology, Oswaldo Cruz Institute/Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Vanessa S de Paula
- Laboratory of Technological Development in Virology, Oswaldo Cruz Institute/Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Leilane M Lopes
- Laboratory of Technological Development in Virology, Oswaldo Cruz Institute/Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Wilhelmus HM Van der Poel
- Central Veterinary, Institute of Wageningen University and Research Centre, Wageningen, The Netherlands
| | - Jorge E González
- National Reference Laboratory in Viral Hepatitis, National Institute of Infectious Diseases, Buenos Aires, Argentina
| | - Maria S Munné
- National Reference Laboratory in Viral Hepatitis, National Institute of Infectious Diseases, Buenos Aires, Argentina
| | - Julio Moran
- Dr. Julio Moran Laboratories, Ebmatingen, Zurich, Switzerland
| | - Ana Carolina R A Cajaraville
- Laboratory of Virological Technology, Institute of Technology on Immunobiologicals, Bio-Manguinhos, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Marcelo Pelajo-Machado
- Laboratory of Pathology, Oswaldo Cruz Institute/Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Oswaldo G Cruz
- Programme of Scientific Computation, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Marcelo A Pinto
- Laboratory of Technological Development in Virology, Oswaldo Cruz Institute/Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| |
Collapse
|