1
|
Pollack S, Plonsky M, Tibi R, Libinson-Zebegret I, Yakobov R, Eisenstein I, Magen D. Prevention of post-transplant lymphoproliferative disorder in pediatric kidney transplant recipients. Pediatr Nephrol 2024:10.1007/s00467-024-06522-2. [PMID: 39373867 DOI: 10.1007/s00467-024-06522-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 08/26/2024] [Accepted: 08/28/2024] [Indexed: 10/08/2024]
Abstract
BACKGROUND Post-transplant lymphoproliferative disorder (PTLD) is a devastating complication of immunosuppressive treatment in both solid organ transplantations (SOT) and hematopoietic stem cell transplantations (HSCT). Epstein-Barr virus (EBV) infection precedes PTLD in 90% of patients. Rituximab, a monoclonal anti-CD20 antibody, depletes B-lymphocytes, which are the ultimate reservoir for EBV. Although rituximab therapy is commonly used as a preventive measure for PTLD in high-risk HSCT, it is not established in SOT. METHODS Pediatric kidney transplant recipients (PKTR) underwent routine EBV-PCR surveillance. Patients with increasing viral loads, despite immunosuppressive dose reduction, were managed with preventive rituximab therapy. RESULTS Between 2012 and 2023, we identified eight episodes of asymptomatic EBV-PCR-positive blood tests in seven out of 65 PKTR (11%) under our care. EBV DNAemia emerged 120-720 days post-transplantation. Five of seven patients with EBV DNAemia (71%) were EBV-seronegative prior to transplantation. All five patients did not respond to MMF dose reduction and were therefore treated with preventive rituximab therapy. Following this treatment, EBV PCR clearance was observed in all patients with only minimal complications. CONCLUSIONS PKTR who are EBV-naïve prior to transplantation are expected to have a higher prevalence of EBV DNAemia. We found that PKTR who were EBV seronegative prior to transplantation were less likely to achieve EBV clearance in response to immunosuppression dose reduction. We suggest that rituximab therapy in PKTR may be safe and effective in EBV clearance and PTLD prevention.
Collapse
Affiliation(s)
- Shirley Pollack
- Technion Faculty of Medicine, Haifa, Israel.
- Pediatric Nephrology Institute, Ruth Children's Hospital, Rambam Health Care Campus, Haifa, Israel.
| | - Moran Plonsky
- Technion Faculty of Medicine, Haifa, Israel
- Pediatric Nephrology Institute, Ruth Children's Hospital, Rambam Health Care Campus, Haifa, Israel
| | - Rami Tibi
- Pediatric Nephrology Institute, Ruth Children's Hospital, Rambam Health Care Campus, Haifa, Israel
| | - Irina Libinson-Zebegret
- Pediatric Nephrology Institute, Ruth Children's Hospital, Rambam Health Care Campus, Haifa, Israel
| | - Renata Yakobov
- Pediatric Nephrology Institute, Ruth Children's Hospital, Rambam Health Care Campus, Haifa, Israel
| | - Israel Eisenstein
- Pediatric Nephrology Institute, Ruth Children's Hospital, Rambam Health Care Campus, Haifa, Israel
| | - Daniella Magen
- Technion Faculty of Medicine, Haifa, Israel
- Pediatric Nephrology Institute, Ruth Children's Hospital, Rambam Health Care Campus, Haifa, Israel
| |
Collapse
|
2
|
Furlano PL, Böhmig GA, Puchhammer-Stöckl E, Vietzen H. Mechanistic Understanding of EBV+Lymphoproliferative Disease Development After Transplantation. Transplantation 2024; 108:1867-1881. [PMID: 39166902 DOI: 10.1097/tp.0000000000004919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/23/2024]
Abstract
Posttransplant lymphoproliferative disorders (PTLDs) are among the most common malignant complications after transplantation, leading to a drastic reduction in patient survival rates. The majority of PTLDs are tightly linked to Epstein-Barr virus (EBV+PTLDs) and are the result of an uncontrolled proliferation of EBV-infected cells. However, although EBV infections are a common finding in transplant recipients, most patients with high EBV loads will never develop EBV+PTLD. Natural killer cells and EBV-specific CD8+ T lymphocytes are critical for controlling EBV-infected cells, and the impairment of these cytotoxic immune responses facilitates the unfettered proliferation of EBV-infected cells. Recent years have seen a considerable increase in available literature aiming to describe novel risk factors associated with the development of EBV+PTLD, which may critically relate to the strength of EBV-specific natural killer cell and EBV-CD8+ T lymphocyte responses. The accumulation of risk factors and the increased risk of developing EBV+PTLD go hand in hand. On the one hand, most of these risk factors, such as the level of immunosuppression or the EBV donor and recipient serologic mismatch, and distinct genetic risk factors are host related and affect cytotoxic EBV-specific immune responses. On the other hand, there is growing evidence that distinct EBV variants may have an increased malignant potential and are thus more likely to induce EBV+PTLD. Here, we aim to review, from a mechanistic point of view, the risk factors for EBV+PTLD in the host and the infecting EBV variants that may explain why only a minority of transplant recipients develop EBV+PTLD.
Collapse
Affiliation(s)
| | - Georg A Böhmig
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | | | - Hannes Vietzen
- Center for Virology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
3
|
Cheng J, Wistinghausen B. Clinicopathologic Spectrum of Pediatric Posttransplant Lymphoproliferative Diseases Following Solid Organ Transplant. Arch Pathol Lab Med 2024; 148:1052-1062. [PMID: 38051286 DOI: 10.5858/arpa.2023-0323-ra] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/22/2023] [Indexed: 12/07/2023]
Abstract
CONTEXT.— Posttransplant lymphoproliferative disorder (PTLD) remains a significant complication in pediatric patients undergoing solid organ transplant (SOT). The majority involve Epstein-Barr virus (EBV)-driven CD20+ B-cell proliferations, which respond to reduction of immunosuppression and anti-CD20-directed immunotherapy. Owing to the low overall incidence, prospective studies of pediatric PTLD are scarce, leading to a lack of comprehensive understanding of this disorder in pediatric populations. This review aims to bridge this knowledge gap by providing a comprehensive analysis of the clinical, morphologic, and molecular genetic features of PTLD in children, adolescents, and young adults after SOT. OBJECTIVE.— To examine the clinical features, pathogenesis, and classification of pediatric PTLDs after SOT. DATA SOURCES.— Personal experiences and published works in PubMed. CONCLUSIONS.— PTLD includes a broad and heterogeneous spectrum of disorders, ranging from nonmalignant lymphoproliferations to lymphomas. While most pediatric PTLDs are EBV+, an increasing number of EBV- PTLDs have been recognized. The pathologic classification of PTLDs has evolved in recent decades, reflecting advancements in understanding the underlying pathobiology. Nevertheless, there remains a great need for further research to elucidate the biology, identify patients at higher risk for aggressive disease, and establish optimal treatment strategies for relapsed/refractory disease.
Collapse
Affiliation(s)
- Jinjun Cheng
- From the Department of Pathology and Laboratory Medicine (Cheng), Center for Cancer and Blood Disorders (Wistinghausen), and Center for Cancer and Immunology Research (Cheng, Wistinghausen), Children's National Hospital, Washington, District of Columbia
| | - Birte Wistinghausen
- From the Department of Pathology and Laboratory Medicine (Cheng), Center for Cancer and Blood Disorders (Wistinghausen), and Center for Cancer and Immunology Research (Cheng, Wistinghausen), Children's National Hospital, Washington, District of Columbia
| |
Collapse
|
4
|
El-amir Z, Jamil A, Solanki D, Mansuri U, Mathew M, Singh J, Aslam S, Akram H, Mandal S, Kichloo A. Admissions for posttransplant lymphoproliferative disorder: a 9-year longitudinal analysis of the National (Nationwide) Inpatient Sample. Proc AMIA Symp 2024; 37:804-812. [PMID: 39165820 PMCID: PMC11332645 DOI: 10.1080/08998280.2024.2375688] [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: 02/28/2024] [Revised: 05/14/2024] [Accepted: 06/24/2024] [Indexed: 08/22/2024] Open
Abstract
Background Posttransplant lymphoproliferative disorder (PTLD), a term that encompasses a wide array of malignancies that occur after transplant, can be one of the most devastating transplant complications. While there have been major advancements in care, especially after the landmark PTLD-1 trial in 2012, there is a paucity of information on hospitalizations for PTLD and the changes in hospitalizations over time. Methods This retrospective cohort study used the National Inpatient Sample to identify hospitalizations for PTLD that occurred between 2009 and 2018. We extracted data for hospitalizations with a primary or secondary diagnosis of PTLD and examined a range of variables, including age, gender, race, hospital type, hospital location, and disposition status. We also collected data on hospital region, median household income, insurance status, and bed size. Results There was a statistically significant increase in the number of hospitalizations from 2009 to 2019 and an increasing rate of hospitalizations over the study period. Hypertension, electrolyte imbalances, renal failure, and anemia were among the most common comorbidities. We found an increased mortality rate, but this was not statistically significant. Conclusion Our study provides insight into the changes in hospitalizations for PTLD over nearly a decade, showing an increase in hospitalizations and reports of comorbidities.
Collapse
Affiliation(s)
- Zain El-amir
- Department of Internal Medicine, Michigan Medicine, Ann Arbor, Michigan, USA
| | - Abdur Jamil
- Division of Internal Medicine, Samaritan Medical Center, Watertown, New York, USA
| | | | - Uvesh Mansuri
- Department of Medicine, Medstar Harbor Hospital, Baltimore, Maryland, USA
| | | | - Jagmeet Singh
- Division of Nephrology, Guthrie Robert Packer Hospital, Sayre, Pennsylvania, USA
| | - Shehroz Aslam
- Division of Internal Medicine, Samaritan Medical Center, Watertown, New York, USA
| | - Hamza Akram
- Division of Internal Medicine, Samaritan Medical Center, Watertown, New York, USA
| | - Shobha Mandal
- Hospitalist Department, Guthrie Robert Packer Hospital, Sayre, Pennsylvania, USA
| | - Asim Kichloo
- Division of Internal Medicine, Samaritan Medical Center, Watertown, New York, USA
| |
Collapse
|
5
|
Nikiforow S, Whangbo JS, Reshef R, Tsai DE, Bunin N, Abu-Arja R, Mahadeo KM, Weng WK, Van Besien K, Loeb D, Nasta SD, Nemecek ER, Zhao W, Sun Y, Galderisi F, Wahlstrom J, Mehta A, Gamelin L, Dinavahi R, Prockop S. Tabelecleucel for EBV+ PTLD after allogeneic HCT or SOT in a multicenter expanded access protocol. Blood Adv 2024; 8:3001-3012. [PMID: 38625984 PMCID: PMC11215195 DOI: 10.1182/bloodadvances.2023011626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 03/27/2024] [Accepted: 03/31/2024] [Indexed: 04/18/2024] Open
Abstract
ABSTRACT Patients with Epstein-Barr virus (EBV)-positive posttransplant lymphoproliferative disease (EBV+ PTLD) in whom initial treatment fails have few options and historically low median overall survival (OS) of 0.7 months after allogeneic hematopoietic cell transplant (HCT) and 4.1 months after solid organ transplant (SOT). Tabelecleucel is an off-the-shelf, allogeneic EBV-specific cytotoxic T-lymphocyte immunotherapy for EBV+ PTLD. Previous single-center experience showed responses in patients with EBV+ PTLD after HCT or SOT. We now report outcomes from a multicenter expanded access protocol in HCT (n = 14) and SOT (n = 12) recipients treated with tabelecleucel for EBV+ PTLD that was relapsed/refractory (R/R) to rituximab with/without chemotherapy. The investigator-assessed objective response rate was 65.4% overall (including 38.5% with a complete and 26.9% with a partial response), 50.0% in HCT, and 83.3% in SOT. The estimated 1- and 2-year OS rates were both 70.0% (95% confidence interval [CI], 46.5-84.7) overall, both 61.5% (95% CI, 30.8-81.8) in HCT, and both 81.5% (95% CI, 43.5-95.1) in SOT (median follow-up: 8.2, 2.8, and 22.5 months, respectively). Patients responding to tabelecleucel had higher 1- and 2-year OS rates (94.1%) than nonresponders (0%). Treatment was well tolerated, with no reports of tumor flare, cytokine release syndrome, or rejection of marrow and SOT. Results demonstrate clinically meaningful outcomes across a broad population treated with tabelecleucel, indicating a potentially transformative and accessible treatment advance for R/R EBV+ PTLD after HCT or SOT. This trial was registered at www.ClinicalTrials.gov as #NCT02822495.
Collapse
Affiliation(s)
- Sarah Nikiforow
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Jennifer S. Whangbo
- VOR Bio, Cambridge, MA
- Department of Pediatrics, Boston Children’s Hospital, Boston, MA
| | - Ran Reshef
- Blood and Marrow Transplantation and Cell Therapy Program, Columbia University Irving Medical Center, New York, NY
| | - Donald E. Tsai
- Department of Medicine, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA
| | - Nancy Bunin
- Division of Pediatric Hematology/Oncology and Blood and Marrow Transplant, Children’s Hospital of Philadelphia, Philadelphia, PA
| | - Rolla Abu-Arja
- Division of Pediatric Hematology/Oncology and Blood and Marrow Transplant, Nationwide Children’s Hospital, Columbus, OH
| | - Kris Michael Mahadeo
- Division of Pediatric Transplant and Cellular Therapy, Duke University Medical Center, Durham, NC
| | - Wen-Kai Weng
- BMT-Cellular Therapy, Department of Medicine, Stanford University, School of Medicine, Stanford, CA
| | - Koen Van Besien
- Department of Medicine, University Hospitals Seidman Cancer Center, Cleveland, OH
| | - David Loeb
- Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA
| | - Sunita Dwivedy Nasta
- Department of Medicine, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA
| | - Eneida R. Nemecek
- Pediatric Transplant & Cellular Therapy, Oregon Health and Science University, Portland, OR
| | | | - Yan Sun
- Atara Biotherapeutics, Thousand Oaks, CA
| | | | | | | | | | | | - Susan Prockop
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- VOR Bio, Cambridge, MA
- Department of Pediatrics, Boston Children’s Hospital, Boston, MA
- Dana-Farber Cancer Institute/Boston Children's Hospital Cancer and Blood Disorders Center, Boston, MA
| |
Collapse
|
6
|
Tajima T, Martinez OM, Bernstein D, Boyd SD, Gratzinger D, Lum G, Sasaki K, Tan B, Twist CJ, Weinberg K, Armstrong B, Desai DM, Mazariegos GV, Chin C, Fishbein TM, Tekin A, Venick RS, Krams SM, Esquivel CO. Epstein-Barr virus-associated post-transplant lymphoproliferative disorders in pediatric transplantation: A prospective multicenter study in the United States. Pediatr Transplant 2024; 28:e14763. [PMID: 38682750 PMCID: PMC11115376 DOI: 10.1111/petr.14763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 03/26/2024] [Accepted: 04/08/2024] [Indexed: 05/01/2024]
Abstract
BACKGROUND Epstein-Barr virus (EBV)-associated post-transplant lymphoproliferative disorders (PTLD) is the most common malignancy in children after transplant; however, difficulties for early detection may worsen the prognosis. METHODS The prospective, multicenter, study enrolled 944 children (≤21 years of age). Of these, 872 received liver, heart, kidney, intestinal, or multivisceral transplants in seven US centers between 2014 and 2019 (NCT02182986). In total, 34 pediatric EBV+ PTLD (3.9%) were identified by biopsy. Variables included sex, age, race, ethnicity, transplanted organ, EBV viral load, pre-transplant EBV serology, immunosuppression, response to chemotherapy and rituximab, and histopathological diagnosis. RESULTS The uni-/multivariable competing risk analyses revealed the combination of EBV-seropositive donor and EBV-naïve recipient (D+R-) was a significant risk factor for PTLD development (sub-hazard ratio: 2.79 [1.34-5.78], p = .006) and EBV DNAemia (2.65 [1.72-4.09], p < .001). Patients with D+R- were significantly more associated with monomorphic/polymorphic PTLD than those with the other combinations (p = .02). Patients with monomorphic/polymorphic PTLD (n = 21) had significantly more EBV DNAemia than non-PTLD patients (p < .001) and an earlier clinical presentation of PTLD than patients with hyperplasias (p < .001), within 6-month post-transplant. Among non-liver transplant recipients, monomorphic/polymorphic PTLD were significantly more frequent than hyperplasias in patients ≥5 years of age at transplant (p = .01). CONCLUSIONS D+R- is a risk factor for PTLD and EBV DNAemia and associated with the incidence of monomorphic/polymorphic PTLD. Intensive follow-up of EBV viral load within 6-month post-transplant, especially for patients with D+R- and/or non-liver transplant recipients ≥5 years of age at transplant, may help detect monomorphic/polymorphic PTLD early in pediatric transplant.
Collapse
Affiliation(s)
- Tetsuya Tajima
- Division of Abdominal Transplantation, Department of Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Olivia M Martinez
- Division of Abdominal Transplantation, Department of Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Daniel Bernstein
- Department of Pediatrics, Stanford University School of Medicine, Stanford, California, USA
| | - Scott D Boyd
- Department of Pathology, Stanford University School of Medicine, Stanford, California, USA
| | - Dita Gratzinger
- Department of Pathology, Stanford University School of Medicine, Stanford, California, USA
| | - Grant Lum
- Division of Abdominal Transplantation, Department of Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Kazunari Sasaki
- Division of Abdominal Transplantation, Department of Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Brent Tan
- Department of Pathology, Stanford University School of Medicine, Stanford, California, USA
| | - Clare J Twist
- Department of Pediatric Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Kenneth Weinberg
- Department of Pediatrics, Stanford University School of Medicine, Stanford, California, USA
| | - Brian Armstrong
- Rho Federal Systems Division, Rho, Durham, North Carolina, USA
| | - Dev M Desai
- Division of Surgical Transplantation, University of Texas (UT) Southwestern Medical Center, Dallas, Texas, USA
| | - George V Mazariegos
- Department of Pediatrics, University of Pittsburgh Medical Center (UPMC) Children's Hospital, Pittsburgh, Pennsylvania, USA
| | - Clifford Chin
- Department of Pediatrics and Cincinnati Children's Hospital, University of Cincinnati, Cincinnati, Ohio, USA
| | - Thomas M Fishbein
- Department of Surgery and Pediatrics, MedStar Georgetown University Hospital, Washington, DC, USA
| | - Akin Tekin
- Department of Surgery, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Robert S Venick
- Department of Pediatric Gastroenterology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Sheri M Krams
- Division of Abdominal Transplantation, Department of Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Carlos O Esquivel
- Division of Abdominal Transplantation, Department of Surgery, Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
7
|
Amjad W, Hamaad Rahman S, Schiano TD, Jafri SM. Epidemiology and Management of Infections in Liver Transplant Recipients. Surg Infect (Larchmt) 2024; 25:272-290. [PMID: 38700753 DOI: 10.1089/sur.2023.346] [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] [Indexed: 07/13/2024] Open
Abstract
Background: Improvements in liver transplant (LT) outcomes are attributed to advances in surgical techniques, use of potent immunosuppressants, and rigorous pre-LT testing. Despite these improvements, post-LT infections remain the most common complication in this population. Bacteria constitute the most common infectious agents, while fungal and viral infections are also frequently encountered. Multi-drug-resistant bacterial infections develop because of polymicrobial overuse and prolonged hospital stays. Immediate post-LT infections are commonly caused by viruses. Conclusions: Appropriate vaccination, screening of both donor and recipients before LT and antiviral prophylaxis in high-risk individuals are recommended. Antimicrobial drug resistance is common in high-risk LT and associated with poor outcomes; epidemiology and management of these cases is discussed. Additionally, we also discuss the effect of coronavirus disease 2019 (COVID-19) infection and monkeypox in the LT population.
Collapse
Affiliation(s)
- Waseem Amjad
- Gastroenterology and Hepatology, University of Maryland, Baltimore, Maryland, USA
| | | | - Thomas D Schiano
- Recanati-Miller Transplantation Institute, Division of Liver Diseases, Mount Sinai Medical Center, New York, New York, USA
| | - Syed-Mohammed Jafri
- Gastroenterology and Hepatology, Henry Ford Hospital, Detroit, Michigan, USA
| |
Collapse
|
8
|
Ergisi M, Ooi B, Salim O, Papalois V. Post-transplant lymphoproliferative disorders following kidney transplantation: A literature review with updates on risk factors, prognostic indices, screening strategies, treatment and analysis of donor type. Transplant Rev (Orlando) 2024; 38:100837. [PMID: 38430887 DOI: 10.1016/j.trre.2024.100837] [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] [Revised: 01/31/2024] [Accepted: 02/19/2024] [Indexed: 03/05/2024]
Abstract
Post-transplant lymphoproliferative disorders (PTLD) is a devastating complication of kidney transplantation with an insidious presentation and potential to disseminate aggressively. This review delineates the risk factors, prognostic indexes, screening, current management algorithm and promising treatment strategies for PTLD. Kidneys from both extended criteria donors (ECD) and living donors (LD) are being increasingly used to expand the donor pool. This review also delineates whether PTLD outcomes vary based on these donor sources. While Epstein-Barr virus (EBV) is a well-known risk factor for PTLD development, the use of T-cell depleting induction agents has been increasingly implicated in aggressive, monomorphic forms of PTLD. Research regarding maintenance therapy is sparse. The international prognostic index seems to be the most validate prognostic tool. Screening for PTLD is controversial, as annual PET-CT is most sensitive but costly, while targeted monitoring of EBV-seronegative patients was more economically feasible, is recommended by the American Society of Transplantation, but is limited to a subset of the population. Other screening strategies such as using Immunoglobulin/T-cell receptor require further validation. A risk-stratified approach is taken in the treatment of PTLD. The first step is the reduction of immunosuppressants, after which rituximab and chemotherapy may be introduced if unsuccessful. Some novel treatments have also shown potential benefit in studies: brentuximab vedotin, chimeric antigen receptor T-cell therapy and EBV-specific cytotoxic T lymphocytes. Analysis of LD v DD recipients show no significant difference in incidence and mortality of PTLD but did reveal a shortened time to development of PTLD from transplant. Analysis of SCD vs ECD recipients show a higher incidence of PTLD in the ECD group, which might be attributed to longer time on dialysis for these patients, age, and the pro-inflammatory nature of these organs. However, incidence of PTLD overall is still extremely low. Efforts should be focused on optimising recipients instead. Minimising the use of T-cell depleting therapy while encouraging research on the effect of new immunosuppressants on PTLD, screening for EBV status are essential, while enabling shared decision-making during counselling when choosing kidney donor types and individualised risk tailoring are strongly advocated.
Collapse
Affiliation(s)
- Mehmet Ergisi
- Norfolk and Norwich University Hospital, Norfolk and Norwich University Hospitals NHS Foundation Trust, Department of Medicine, Norwich, United Kingdom.
| | - Bryan Ooi
- Department of Medicine, Imperial College London, London, United Kingdom.
| | - Omar Salim
- Isle of Wight NHS Trust, Parkhurst Road, Newport, United Kingdom
| | - Vassilios Papalois
- Hammersmith Hospital, Imperial College Healthcare NHS Trust, Department of Transplant and General Surgery, London, United Kingdom.
| |
Collapse
|
9
|
Kim IC, Kim SH, Youn JC, Kim D, Lee S, Kim H, Kim JJ, Jung MH, Rossano JW, Cherikh WS, Kobashigawa JA, Stehlik J. Temporal Trends, Risk Factors, and Clinical Outcomes of De Novo Lymphoproliferative Disorders After Heart Transplantation. JACC. HEART FAILURE 2024; 12:395-405. [PMID: 38326002 DOI: 10.1016/j.jchf.2023.10.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 10/02/2023] [Accepted: 10/17/2023] [Indexed: 02/09/2024]
Abstract
BACKGROUND Post-transplant lymphoproliferative disorder (PTLD) is an important cause of morbidity and mortality in heart transplant (HTx) recipients. However, previous studies of PTLD after HTx are limited to single-center analyses or extrapolated from all solid organ transplantations. OBJECTIVES The authors analyzed the temporal trends, risk factors, and clinical outcome of de novo PTLD specifically after HTx. METHODS Using multi-institutional, multinational data from the International Society for Heart and Lung Transplantation Thoracic Organ Transplant Registry, the authors evaluated the real-world data of PTLD after HTx, transplanted between January 2000 and June 2015. Multivariable analysis was done to identify risk factors for PTLD development after HTx. RESULTS Among 28,136 HTx recipients, 1,069 (3.8%) developed PTLD within 10 years of transplantation. PTLD showed a bimodal age pattern with peak incidence in patients of pediatric age and late adulthood at transplantation. The early transplant era (2000-2007 vs 2008-2015), male recipient, and EBV donor-positive-recipient-negative match were independent risk factors of PTLD development within 3 years of transplantation, whereas maintenance therapy with cyclosporine vs tacrolimus at initial discharge was associated with a lower incidence. PTLD development within 3 years of transplantation was significantly associated with mortality (HR: 2.42 [95% CI: 2.01-2.91]; P < 0.001). Survival after PTLD diagnosis was higher in the recent transplant era. CONCLUSIONS PTLD is relatively rare, but potentially fatal, post-transplant malignancy. PTLD incidence and mortality after HTx have decreased in the recent era. Strategies to minimize the risk of PTLD, and ensure early diagnosis and effective treatment are likely to improve outcomes in HTx.
Collapse
Affiliation(s)
- In-Cheol Kim
- Division of Cardiology, Department of Internal Medicine, Keimyung University Dongsan Hospital, Daegu, Republic of Korea
| | - Sang Hyun Kim
- Division of Cardiology, Department of Internal Medicine, Seoul St. Mary's Hospital, Catholic Research Institute for Intractable Cardiovascular Disease, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jong-Chan Youn
- Division of Cardiology, Department of Internal Medicine, Seoul St. Mary's Hospital, Catholic Research Institute for Intractable Cardiovascular Disease, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.
| | - Darae Kim
- Division of Cardiology, Department of Medicine, Heart Vascular Stroke Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Seonhwa Lee
- Division of Cardiology, Department of Internal Medicine, Keimyung University Dongsan Hospital, Daegu, Republic of Korea
| | - Hyungseop Kim
- Division of Cardiology, Department of Internal Medicine, Keimyung University Dongsan Hospital, Daegu, Republic of Korea
| | - Jin-Jin Kim
- Division of Cardiology, Department of Internal Medicine, Seoul St. Mary's Hospital, Catholic Research Institute for Intractable Cardiovascular Disease, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Mi-Hyang Jung
- Division of Cardiology, Department of Internal Medicine, Seoul St. Mary's Hospital, Catholic Research Institute for Intractable Cardiovascular Disease, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Joseph W Rossano
- The Cardiac Center, The Children's Hospital of Philadelphia, Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Wida S Cherikh
- United Network for Organ Sharing, Richmond, Virginia, USA; ISHLT Transplant Registry, Dallas, Texas, USA
| | - Jon A Kobashigawa
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Josef Stehlik
- Division of Cardiovascular Medicine, University of Utah School of Medicine, Salt Lake City, Utah, USA.
| |
Collapse
|
10
|
Hasegawa Y, Obara H, Kikuchi T, Uno S, Tsujikawa H, Yamada Y, Hori S, Eguchi S, Kitagawa Y. Malignant lymphoma after liver transplantation for liver cirrhosis caused by human immunodeficiency virus and hepatitis C virus co-infection. J Infect Chemother 2023; 29:1160-1163. [PMID: 37543174 DOI: 10.1016/j.jiac.2023.08.002] [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: 06/08/2023] [Revised: 07/25/2023] [Accepted: 08/02/2023] [Indexed: 08/07/2023]
Abstract
Here, we describe a rare case of malignant lymphoma after liver transplantation for liver cirrhosis caused by human immunodeficiency virus (HIV) and hepatitis C virus (HCV) co-infection. A male patient was diagnosed with hemophilia A at 8 months of age. Since then, he had been receiving blood products, which led to HIV and HCV co-infection. His HIV viral load was suppressed with antiretroviral therapy, and a sustained virologic response was achieved for HCV using direct-acting antivirals. However, his decompensated liver cirrhosis progressed, and deceased donor liver transplantation was performed. A post-transplant lymphoproliferative disorder (PTLD) developed 105 days after liver transplantation, with enlarged para-aortic and hilar lymph nodes, a right renal mass, and masses in the small and large intestines. Histopathological examination confirmed monomorphic PTLD (diffuse large B-cell lymphoma). Against the treatment (reduction of immunosuppression, rituximab, and chemotherapy), the response was poor, and the patient died 94 days after the outbreak of PTLD. Both transplantation and HIV infection are risk factors for lymphoproliferative diseases. To the best of our knowledge, this is one of the very few reports of PTLD in a patient with HIV/HCV co-infection.
Collapse
Affiliation(s)
- Yasushi Hasegawa
- Department of Surgery, Keio University School of Medicine, Japan
| | - Hideaki Obara
- Department of Surgery, Keio University School of Medicine, Japan.
| | - Taku Kikuchi
- Division of Hematology, Department of Medicine, Keio University School of Medicine, Japan
| | - Shunsuke Uno
- Department of Infectious Diseases, Keio University School of Medicine, Japan
| | - Hanako Tsujikawa
- Department of Pathology, Keio University School of Medicine, Japan
| | - Yohei Yamada
- Department of Pediatric Surgery, Keio University School of Medicine, Japan
| | - Shutaro Hori
- Department of Surgery, Keio University School of Medicine, Japan
| | - Susumu Eguchi
- Department of Surgery, Nagasaki University Graduate School of Biomedical Sciences, Japan
| | - Yuko Kitagawa
- Department of Surgery, Keio University School of Medicine, Japan
| |
Collapse
|
11
|
Kolijn PM, Langerak AW. Immune dysregulation as a leading principle for lymphoma development in diverse immunological backgrounds. Immunol Lett 2023; 263:46-59. [PMID: 37774986 DOI: 10.1016/j.imlet.2023.08.007] [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: 05/22/2023] [Revised: 07/28/2023] [Accepted: 08/10/2023] [Indexed: 10/01/2023]
Abstract
Lymphoma is a heterogeneous group of malignancies arising from lymphocytes, which poses a significant challenge in terms of diagnosis and treatment due to its diverse subtypes and underlying mechanisms. This review aims to explore the shared and distinct features of various forms of lymphoma predisposing conditions, with a focus on genetic, immunological and molecular aspects. While diseases such as autoimmune disorders, inborn errors of immunity and iatrogenic immunodeficiencies are biologically and immunologically distinct, each of these diseases results in profound immune dysregulation and a predisposition to lymphoma development. Interestingly, the increased risk is often skewed towards a particular subtype of lymphoma. Patients with inborn errors of immunity in particular present with extreme forms of lymphoma predisposition, providing a unique opportunity to study the underlying mechanisms. External factors such as chronic infections and environmental exposures further modulate the risk of lymphoma development. Common features of conditions predisposing to lymphoma include: persistent inflammation, recurrent DNA damage or malfunctioning DNA repair, impaired tumor surveillance and viral clearance, and dysregulation of fundamental cellular processes such as activation, proliferation and apoptosis. Our growing understanding of the underlying mechanisms of lymphomagenesis provides opportunities for early detection, prevention and tailored treatment of lymphoma development.
Collapse
Affiliation(s)
- P Martijn Kolijn
- Laboratory Medical Immunology, Department of Immunology, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Anton W Langerak
- Laboratory Medical Immunology, Department of Immunology, Erasmus Medical Center, Rotterdam, the Netherlands.
| |
Collapse
|
12
|
Leivonen SK, Friman T, Autio M, Vaittinen S, Jensen AW, D'Amore F, Hamilton-Dutoit SJ, Holte H, Beiske K, Kovanen PE, Räty R, Leppä S. Characterization and clinical impact of the tumor microenvironment in post-transplant aggressive B-cell lymphomas. Haematologica 2023; 108:3044-3057. [PMID: 37259566 PMCID: PMC10620595 DOI: 10.3324/haematol.2023.282831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 05/23/2023] [Indexed: 06/02/2023] Open
Abstract
Post-transplant lymphoproliferative disorders (PTLD) are iatrogenic immune deficiency-associated lymphoid/plasmacytic proliferations developing due to immunosuppression in solid organ or hematopoietic stem cell allograft patients. PTLD are characterized by abnormal proliferation of lymphoid cells and have a heterogeneous clinical behavior. We profiled expression of >700 tumor microenvironment (TME)-related genes in 75 post-transplant aggressive B-cell lymphomas (PTABCL). Epstein-Barr virus (EBV)-positive PT-ABCL clustered together and were enriched for type I interferon pathway and antiviral-response genes. Additionally, a cytotoxicity gene signature associated with EBV-positivity and favorable overall survival (OS) (hazard ratio =0.61; P=0.019). In silico immunophenotyping revealed two subgroups with distinct immune cell compositions. The inflamed subgroup with higher proportions of immune cells had better outcome compared to noninflamed subgroup (median OS >200.0 vs. 15.2 months; P=0.006). In multivariable analysis with EBV status, International Prognostic Index, and rituximab-containing treatment, inflamed TME remained as an independent predictor for favorable outcome. We also compared TME between post-transplant and immunocompetent host diffuse large B-cell lymphomas (n=75) and discovered that the proportions of T cells were lower in PT-diffuse large B-cell lymphomas. In conclusion, we provide a comprehensive phenotypic characterization of PT-ABCL, highlighting the importance of immune cell composition of TME in determining the clinical behavior and prognosis of PT-ABCL.
Collapse
Affiliation(s)
- Suvi-Katri Leivonen
- Applied Tumor Genomics Research Program, Medical Faculty, University of Helsinki, Helsinki, Finland; Department of Oncology, Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland; iCAN Digital Precision Cancer Medicine Flagship, Helsinki
| | - Terhi Friman
- Department of Hematology, Helsinki University Hospital Comprehensive Cancer Center and University of Helsinki
| | - Matias Autio
- Applied Tumor Genomics Research Program, Medical Faculty, University of Helsinki, Helsinki, Finland; Department of Oncology, Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland; iCAN Digital Precision Cancer Medicine Flagship, Helsinki
| | - Samuli Vaittinen
- Department of Pathology, Turku University Hospital, University of Turku, Turku
| | | | | | | | - Harald Holte
- Department of Oncology, Oslo University Hospital, Oslo, Norway
| | - Klaus Beiske
- Department of Pathology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Panu E Kovanen
- Department of Pathology, University of Helsinki, and HUSLAB, Helsinki University Hospital, Helsinki
| | - Riikka Räty
- Department of Hematology, Helsinki University Hospital Comprehensive Cancer Center and University of Helsinki
| | - Sirpa Leppä
- Applied Tumor Genomics Research Program, Medical Faculty, University of Helsinki, Helsinki, Finland; Department of Oncology, Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland; iCAN Digital Precision Cancer Medicine Flagship, Helsinki.
| |
Collapse
|
13
|
Shang F, Wang Y, Shi Z, Deng Z, Ma J. Development of a Signature Based on Eight Metastatic-Related Genes for Prognosis of GC Patients. Mol Biotechnol 2023; 65:1796-1808. [PMID: 36790659 PMCID: PMC10518294 DOI: 10.1007/s12033-023-00671-9] [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: 10/08/2022] [Accepted: 01/14/2023] [Indexed: 02/16/2023]
Abstract
Gastric cancer (GC) has been a common tumor type with high mortality. Distal metastasis is one of the main causes of death in GC patients, which is also related to poor prognosis. The mRNA profiles and clinical information of GC patients were downloaded from The Cancer Genome Atlas and Gene Expression Omnibus databases. Univariate Cox and LASSO Cox analyses were used to screen the optimal metastasis-related genes (MRGs) to establish a prognostic Risk Score model for GC patients. The nomogram was used to visualize the Risk Score and predict the 1-, 3-, 5-year survival rate. The immune cell infiltration was analyzed by CIBERSORT and the ratio of immune-stromal component was calculated by the ESTIMATE algorithm. A total of 142 differentially expressed genes were identified between metastatic and non-metastatic GC samples. The optimal 8 genes, comprising GAMT (guanidinoacetate N-methyltransferase), ABCB5 (ATP-binding cassette subfamily B member 5), ITIH3 (inter-alpha-trypsin inhibitor heavy chain 3), GDF3 (growth differentiation factor 3), VSTM2L (V-set and transmembrane domain-containing 2 like), CIDEA (cell death inducing DFFA like effector a), NPTX1 (neuronal pentraxin-1), and UMOD (uromodulin), were further screened to establish a prognostic Risk Score, which proved to be an independent prognostic factor. Patients in high-risk group had a poor prognosis. There were significant differences in the proportion of 11 tumor-infiltrating immune cells between high-risk and low-risk subgroups. In addition, the StromalScore, ImmuneScore, and ESTIMATEScore in high-risk group were higher than those in low-risk group, indicating that the tumor microenvironment of the high-risk group was more complex. A Risk Score model based on eight metastasis-related genes could clearly distinguish the prognosis of GC patients. The poor prognosis of patients with high-Risk Score might be associated with the complex tumor microenvironments.
Collapse
Affiliation(s)
- Fanjing Shang
- Department of General Surgery, People's Hospital of Ningxia Hui Autonomous Region, No. 301 Zhengyuan North Road, Jinfeng District, Yinchuan, 750001, Ningxia, China
| | - Yafei Wang
- Department of General Surgery, People's Hospital of Ningxia Hui Autonomous Region, No. 301 Zhengyuan North Road, Jinfeng District, Yinchuan, 750001, Ningxia, China
| | - Zixu Shi
- Department of General Surgery, People's Hospital of Ningxia Hui Autonomous Region, No. 301 Zhengyuan North Road, Jinfeng District, Yinchuan, 750001, Ningxia, China
| | - Zhidong Deng
- Department of General Surgery, People's Hospital of Ningxia Hui Autonomous Region, No. 301 Zhengyuan North Road, Jinfeng District, Yinchuan, 750001, Ningxia, China
| | - Jianwen Ma
- Department of General Surgery, People's Hospital of Ningxia Hui Autonomous Region, No. 301 Zhengyuan North Road, Jinfeng District, Yinchuan, 750001, Ningxia, China.
| |
Collapse
|
14
|
Amengual JE, Pro B. How I treat posttransplant lymphoproliferative disorder. Blood 2023; 142:1426-1437. [PMID: 37540819 PMCID: PMC10731918 DOI: 10.1182/blood.2023020075] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 07/03/2023] [Accepted: 07/21/2023] [Indexed: 08/06/2023] Open
Abstract
Posttransplant lymphoproliferative disorder (PTLD) is an important and potentially life-threatening complication of solid organ transplant and hematopoietic stem cell transplant (HSCT). Given the heterogeneity of PTLD and the risk of infectious complications in patients with immunosuppression, the treatment of this disease remains challenging. Monomorphic PTLD and lymphoma of B-cell origin account for the majority of cases. Treatment strategies for PTLD consist of response-adapted, risk-stratified methods using immunosuppression reduction, immunotherapy, and/or chemotherapy. With this approach, ∼25% of the patients do not need chemotherapy. Outcomes for patients with high risk or those who do not respond to frontline therapies remain dismal, and novel treatments are needed in this setting. PTLD is associated with Epstein-Barr virus (EBV) infection in 60% to 80% of cases, making EBV-directed therapy an attractive treatment modality. Recently, the introduction of adoptive immunotherapies has become a promising option for refractory cases; hopefully, these treatment strategies can be used as earlier lines of therapy in the future.
Collapse
Affiliation(s)
- Jennifer E. Amengual
- Division of Hematology and Oncology, Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY
| | - Barbara Pro
- Division of Hematology and Oncology, Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY
| |
Collapse
|
15
|
Ibrahim U, Bassil C, Chavez JC, Khimani F, Jain MD, Locke FL, Osman K, Lazaryan A. CAR T Cell Therapy for Post-Transplant Lymphoproliferative Disorder After Solid Organ Transplantation: A Safe and Feasible Therapy for an Orphan Disease. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2023; 23:772-778. [PMID: 37263876 DOI: 10.1016/j.clml.2023.05.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 05/01/2023] [Accepted: 05/07/2023] [Indexed: 06/03/2023]
Affiliation(s)
- Uroosa Ibrahim
- Bone Marrow Transplant and Cellular Therapy/Division of Hematology and Oncology, Icahn School of Medicine at Mount Sinai, Tisch Cancer Institute, New York, NY.
| | - Claude Bassil
- Department of Onconephrology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Julio C Chavez
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Farhad Khimani
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Michael D Jain
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Frederick L Locke
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Keren Osman
- Bone Marrow Transplant and Cellular Therapy/Division of Hematology and Oncology, Icahn School of Medicine at Mount Sinai, Tisch Cancer Institute, New York, NY
| | - Aleksandr Lazaryan
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| |
Collapse
|
16
|
Coradine TLC, Guida LDOC, Fraiman P, Santos AJ, Pedroso JL, Barsottini OGP. Post-transplant lymphoproliferative disorder myeloradiculopathy. Pract Neurol 2023; 23:408-410. [PMID: 37147122 DOI: 10.1136/pn-2023-003696] [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] [Accepted: 04/12/2023] [Indexed: 05/07/2023]
Abstract
A 56-year-old woman developed progressive subacute lower limb weakness with sensory and autonomic abnormalities. She had received a living-donor kidney transplantation 21 years before for end-stage chronic kidney disease and took mycophenolate mofetil and prednisolone. MR scan of the spinal cord showed bilateral cauda equina gadolinium enhancement and MR scan of the brain showed enhancing nodular hyperintensities in the internal capsule and globus pallidus. Cerebrospinal fluid (CSF) showed a pleocytosis with extremely low glucose, and positive DNA-PCR for Epstein-Barr virus. Her condition worsened despite empirically guided antimicrobial treatment. CSF immunophenotyping later identified mature, clonal B lymphocytes of large size, expressing CD19, CD20, CD200 antigens, and kappa light chain immunoglobulin, with absent CD5 and CD10 expression. We diagnosed a myeloradiculopathy from a monomorphic post-transplant lymphoproliferative disorder. This condition occurs after kidney transplantation and falls on the lymphoma spectrum. We review its clinical features, diagnosis and management.
Collapse
Affiliation(s)
| | | | - Pedro Fraiman
- Departament of Neurology and Neurosurgery, General Neurology Unit, Universidade Federal de São Paulo, Sao Paulo, Brazil
| | - Adrialdo José Santos
- Departament of Neurology and Neurosurgery, Neuro-Oncology Unit, Universidade Federal de São Paulo, São Paulo, Brazil
| | - José Luiz Pedroso
- Departament of Neurology and Neurosurgery, General Neurology Unit, Universidade Federal de São Paulo, Sao Paulo, Brazil
| | - Orlando G P Barsottini
- Departament of Neurology and Neurosurgery, General Neurology Unit, Universidade Federal de São Paulo, Sao Paulo, Brazil
| |
Collapse
|
17
|
Hu J, Yu YW, Han DS, Li XJ, Zhang YQ, Cai HL, Xiao YH, Zheng X. Case Report: Non-negligible Epstein-Barr virus-associated posttransplant lymphoproliferative disorders in a lung transplant recipient. Front Immunol 2023; 14:1244534. [PMID: 37781359 PMCID: PMC10540203 DOI: 10.3389/fimmu.2023.1244534] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 08/24/2023] [Indexed: 10/03/2023] Open
Abstract
Background Posttransplant lymphoproliferative disorders (PTLDs) are uncommon but serious complications in patients following solid organ transplantation. Primary Epstein-Barr virus (EBV) infection is a risk factor for the development of PTLD, especially early-onset PTLD, in EBV-negative recipients. To date, however, there are no specific guidelines on the threshold of EBV-DNA load for therapeutic intervention, the source for measurement (e.g., blood, bronchoalveolar fluid), or the use of antiviral agents as prophylaxis for early PTLD prevention in EBV-mismatched patients. Methods The present study describes a 56-year-old male lung transplant recipient diagnosed with EBV-associated PTLD. Results This patient had a history of invasive fungal disease and Mucor and Aspergillus fumigatus infections in the early post-transplant period, necessitating antifungal therapy throughout the course of the disease. The patient was EBV-positive 15 days after transplantation, with lung CT showing multiple bilateral nodules of varying sizes beginning 98 days after transplantation. A lung biopsy showed PTLD, and next-generation sequencing (NGS) revealed EBV. This patient, however, did not receive any antiviral therapy for early PTLD prevention or any PTLD-related treatment. He died 204 days after lung transplantation. Conclusion The present study describes a lung transplant recipient who developed EBV-associated PTLD, a non-negligible disease, after solid organ transplantation. Monitoring EBV-DNA load is important, as a sudden increase may be a sensitive indicator of PTLD. An earlier diagnosis may increase the likelihood of successful treatment.
Collapse
Affiliation(s)
- Juan Hu
- Intensive Care Unit, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yong-Wei Yu
- Intensive Care Unit, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Dong-Sheng Han
- Centre of Clinical Laboratory, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xue-Jie Li
- Department of Pathology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yi-Qi Zhang
- Intensive Care Unit, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hong-Liu Cai
- Intensive Care Unit, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yong-Hong Xiao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xia Zheng
- Intensive Care Unit, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
18
|
Franco A, Hernandez D, Zarraga S, Fructuoso AS, Crespo M, Mazuecos A, Corte CD, Benot AR, Ruiz JC, Beneyto I. Lymphoproliferative disorders after renal transplantation along 2 decades: a large longitudinal study of 21.546 recipients. Nefrologia 2023; 43:427-434. [PMID: 37813738 DOI: 10.1016/j.nefroe.2023.09.001] [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: 12/09/2021] [Revised: 02/17/2022] [Accepted: 02/20/2022] [Indexed: 10/11/2023] Open
Abstract
INTRODUCTION Post transplant lymphoproliferative disorders (PTLD) are heterogeneous lymphoid proliferations in recipients of solid organs which seem to be related to Epstein Barr Virus (EBV). The use of antilymphocyte antibodies, EBV seronegativity in the recipient,acute rejection and CMV infection have been identified as classical risk factors. MATERIAL Y METHODS We have studied in a retrospective observational study, the incidence of PTLD in a period of 22 years, its relationship with EBV, presence of classical risk factors and outcome in 21546 simple adult renal transplant recipients from cadaveric and living donors, transplanted in 21 hospitals from 1990 to 2009. RESULTS A total of 275 recipients developed PTLD (1,2%),195 males (70,9%), 80 females (29,1%) aged 59.2 (p25 44.7 p75 68)years. Two hundred forty-five (89.0%) were 1st transplant recipients and 269 (97,8%) from cadaveric donors. EBV in the tissue was reported in 94 out of the 155 studied recipients (60.6%) and 86.0% of the proliferations were due to B lymphocytes. PTLD median appearance after transplant were 42.months (p25, 75, 12, 77, 5). One hundred eighty-eight recipients out of 275 patients (68.3%) had any classical risk factor and the use of antilymphocyte antibodies was the most frequent. During the follow-up, 172 patients died (62,5%) and 103 (37,5%) had a complete remission. The main cause of death was PTLD progression (n = 91, 52,9%), followed by sepsis (n = 24, 13,9%). The follow-up period post-transplant of the recipients was between 3 and 22 years. The incidence was 0,14% during the first year post-trasplant and 0.98% the cumulative incidence at 10 years. Patient survival after diagnosis was 51%, 44% and 39% after 1, 2 and 5 years, respectively. Finally, overall graft survival was 48%, 39% and 33% at the same periods. CONCLUSION PTLD has a low incidence in renal transplant recipients. Most of the proliferations are due to B lymphocytes and seem to have a close relationship with EBV. PTLD can develop in the absence of classical risk factors. The prognosis is poor, mainly due to PTLD progression, but the survivors can even maintain their grafts.
Collapse
Affiliation(s)
- Antonio Franco
- Servicio de Nefrología, Hospital General de Alicante, Alicante, Spain.
| | | | - Sofia Zarraga
- Servicio de Nefrologia, Hospital de Cruces, Baracaldo, Vizcaya, Spain
| | | | - Marta Crespo
- Servicio de Nefrologia, Hospital del Mar, Barcelona, Spain
| | | | | | | | - Juan Carlos Ruiz
- Servicio de Nefrologia, Hospital Marque de Valdecilla, Santander, Cantabria, Spain
| | | |
Collapse
|
19
|
Okamoto Y, Shikano S. Emerging roles of a chemoattractant receptor GPR15 and ligands in pathophysiology. Front Immunol 2023; 14:1179456. [PMID: 37457732 PMCID: PMC10348422 DOI: 10.3389/fimmu.2023.1179456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Accepted: 06/19/2023] [Indexed: 07/18/2023] Open
Abstract
Chemokine receptors play a central role in the maintenance of immune homeostasis and development of inflammation by directing leukocyte migration to tissues. GPR15 is a G protein-coupled receptor (GPCR) that was initially known as a co-receptor for human immunodeficiency virus (HIV) and simian immunodeficiency virus (SIV), with structural similarity to other members of the chemoattractant receptor family. Since the discovery of its novel function as a colon-homing receptor of T cells in mice a decade ago, GPR15 has been rapidly gaining attention for its involvement in a variety of inflammatory and immune disorders. The recent identification of its natural ligand C10orf99, a chemokine-like polypeptide strongly expressed in gastrointestinal tissues, has established that GPR15-C10orf99 is a novel signaling axis that controls intestinal homeostasis and inflammation through the migration of immune cells. In addition, it has been demonstrated that C10orf99-independent functions of GPR15 and GPR15-independent activities of C10orf99 also play significant roles in the pathophysiology. Therefore, GPR15 and its ligands are potential therapeutic targets. To provide a basis for the future development of GPR15- or GPR15 ligand-targeted therapeutics, we have summarized the latest advances in the role of GPR15 and its ligands in human diseases as well as the molecular mechanisms that regulate GPR15 expression and functions.
Collapse
Affiliation(s)
| | - Sojin Shikano
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL, United States
| |
Collapse
|
20
|
McKenna M, Epperla N, Ghobadi A, Liu J, Lazaryan A, Ibrahim U, Jacobson CA, Naik SG, Nastoupil L, Chowdhury SM, Voorhees TJ, Jacobs MT, Farooq U, Osman K, Olszewski AJ, Ahmed S, Evens AM. Real-world evidence of the safety and survival with CD19 CAR-T cell therapy for relapsed/refractory solid organ transplant-related PTLD. Br J Haematol 2023. [PMID: 37129856 DOI: 10.1111/bjh.18828] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 04/11/2023] [Accepted: 04/12/2023] [Indexed: 05/03/2023]
Abstract
The use of CD19 chimeric antigen receptor T-cell (CAR-T) therapy for relapsed/refractory solid organ transplantation (SOT)-related post-transplant lymphoproliferative disorder (PTLD) is not well studied. We conducted a multicentre, retrospective analysis of adults with relapsed/refractory SOT-associated PTLD. Among 22 relapsed/refractory SOT-PTLD patients, the pathology was monomorphic B cell. Prior SOTs included 14 kidney (64%), three liver (14%), two heart (9%), one intestinal (5%), one lung (5%), and one pancreas after kidney transplant (5%). The median time from SOT to PTLD diagnosis was 107 months. Pre-CAR-T bridging therapy was used in 55% of patients, and immunosuppression was stopped completely before CAR-T infusion in 64%. Eighteen (82%) patients experienced cytokine release syndrome: one (5%) each grade (G) 3 and G4. The immune effector cell-associated neurotoxicity syndrome was observed in 16 (73%) patients: six (27%) G3 and two (9%) G4. The overall response rate was 64% (55% complete response). Three patients (14%) experienced allograft rejection after CAR-T. The two-year progression-free survival and overall survival rates were 35% and 58%, respectively. Additionally, the achievement of CR post-CAR-T was strongly associated with survival. Collectively, the safety and efficacy of CD19 CAR-T therapy in relapsed/refractory SOT-related PTLD appeared similar to pivotal CAR-T data, including approximately one-third of patients achieving sustained remission.
Collapse
Affiliation(s)
- Marshall McKenna
- Division of Blood Disorders, Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey, USA
| | - Narendranath Epperla
- Division of Hematology, Ohio State University Comprehensive Cancer Center, Columbus, Ohio, USA
| | - Armin Ghobadi
- Siteman Cancer Center, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Jieqi Liu
- Division of Blood Disorders, Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey, USA
| | - Aleksandr Lazaryan
- Blood and Marrow Transplant and Cellular Immunotherapy, Moffitt Cancer Center, Tampa, Florida, USA
| | - Uroosa Ibrahim
- Department of Hematology and Oncology, Bone Marrow Transplantation and Cellular Therapy Program, Mount Sinai Hospital, New York, New York, USA
| | - Caron A Jacobson
- Division of Hematologic Malignancies, Harvard Medical School, Dana Faber Cancer Institute, Boston, Massachusetts, USA
| | - Seema G Naik
- Penn State Cancer Institute, Penn State Hershey Medical Center, Hershey, Pennsylvania, USA
| | - Loretta Nastoupil
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Sayan Mullick Chowdhury
- Division of Hematology, Ohio State University Comprehensive Cancer Center, Columbus, Ohio, USA
| | - Timothy J Voorhees
- Division of Hematology, Ohio State University Comprehensive Cancer Center, Columbus, Ohio, USA
| | - Miriam T Jacobs
- Siteman Cancer Center, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Umar Farooq
- Division of Hematology, Oncology and Blood & Marrow Transplantation, University of Iowa Hospital and Clinics, Iowa City, Iowa, USA
| | - Keren Osman
- Department of Hematology and Oncology, Bone Marrow Transplantation and Cellular Therapy Program, Mount Sinai Hospital, New York, New York, USA
| | - Adam J Olszewski
- Lifespan Cancer Institute, Alpert Medical School of Brown University, Providence, Rhode Island, USA
| | - Sairah Ahmed
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Andrew M Evens
- Division of Blood Disorders, Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey, USA
| |
Collapse
|
21
|
Eiz-Vesper B, Ravens S, Maecker-Kolhoff B. αβ and γδ T-cell responses to Epstein-Barr Virus: insights in immunocompetence, immune failure and therapeutic augmentation in transplant patients. Curr Opin Immunol 2023; 82:102305. [PMID: 36963323 DOI: 10.1016/j.coi.2023.102305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 02/19/2023] [Accepted: 02/21/2023] [Indexed: 03/26/2023]
Abstract
Epstein-Barr Virus (EBV) is a human gamma herpes virus, which causes several diseases in immunocompetent (mononucleosis, chronic fatigue syndrome, gastric cancer, endemic Burkitt's lymphoma, head and neck cancer) and immunosuppressed (post-transplant lymphoproliferative disease, EBV-associated soft tissue tumors) patients. It elicits a complex humoral and cellular immune response with both innate and adaptive immune components. Substantial progress has been made in understanding the interplay of immune cells in EBV-associated diseases in recent years, and several therapeutic approaches have been developed to augment cellular immunity toward EBV for control of EBV-associated malignancy. This review will focus on recent developments in immunosuppressed transplant recipients.
Collapse
Affiliation(s)
- Britta Eiz-Vesper
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, Germany; CRC900 Microbial persistence and its control; German Center for Infection Research (DZIF)
| | - Sarina Ravens
- CRC900 Microbial persistence and its control; Institute of Immunology, Hannover Medical School, Germany
| | - Britta Maecker-Kolhoff
- CRC900 Microbial persistence and its control; German Center for Infection Research (DZIF); Department of Pediatric Hematology and Oncology, Hannover Medical School, Germany.
| |
Collapse
|
22
|
Afify ZAM, Taj MM, Orjuela-Grimm M, Srivatsa K, Miller TP, Edington HJ, Dalal M, Robles J, Ford JB, Ehrhardt MJ, Ureda TJ, Rubinstein JD, McCormack S, Rivers JM, Chisholm KM, Kavanaugh MK, Bukowinski AJ, Friehling ED, Ford MC, Reddy SN, Marks LJ, Smith CM, Mason CC. Multicenter study of pediatric Epstein-Barr virus-negative monomorphic post solid organ transplant lymphoproliferative disorders. Cancer 2023; 129:780-789. [PMID: 36571557 PMCID: PMC11200327 DOI: 10.1002/cncr.34600] [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: 09/20/2022] [Revised: 10/21/2022] [Accepted: 11/07/2022] [Indexed: 12/27/2022]
Abstract
BACKGROUND Pediatric Epstein-Barr virus-negative monomorphic post solid organ transplant lymphoproliferative disorder [EBV(-)M-PTLD] comprises approximately 10% of M-PTLD. No large multi-institutional pediatric-specific reports on treatment and outcome are available. METHODS A multi-institutional retrospective review of solid organ recipients diagnosed with EBV(-)M-PTLD aged ≤21 years between 2001 and 2020 in 12 centers in the United States and United Kingdom was performed, including demographics, staging, treatment, and outcomes data. RESULTS Thirty-six patients were identified with EBV(-)M-PTLD. Twenty-three (63.9%) were male. Median age (range) at transplantation, diagnosis of EBV(-)M-PTLD, and interval from transplant to PTLD were 2.2 years (0.1-17), 14 years (3.0-20), and 8.5 years (0.6-18.3), respectively. Kidney (n = 17 [47.2%]) and heart (n = 13 [36.1%]) were the most commonly transplanted organs. Most were Murphy stage III (n = 25 [69.4%]). Lactate dehydrogenase was elevated in 22/34 (64.7%) and ≥2 times upper limit of normal in 11/34 (32.4%). Pathological diagnoses included diffuse large B-cell lymphoma (n = 31 [86.1%]) and B-non-Hodgkin lymphoma (B-NHL) not otherwise specified (NOS) (n = 5 [13.9%]). Of nine different regimens used, the most common were: pediatric mature B-NHL-specific regimen (n = 13 [36.1%]) and low-dose cyclophosphamide, prednisone, and rituximab (n = 9 [25%]). Median follow-up from diagnosis was 3.0 years (0.3-11.0 years). Three-year event-free survival (EFS) and overall survival (OS) were 64.8% and 79.9%, respectively. Of the seven deaths, six were from progressive disease. CONCLUSIONS EFS and OS were comparable to pediatric EBV(+) PTLD, but inferior to mature B-NHL in immunocompetent pediatric patients. The wide range of therapeutic regimens used directs our work toward developing an active multi-institutional registry to design prospective studies. PLAIN LANGUAGE SUMMARY Pediatric Epstein-Barr virus-negative monomorphic post solid organ transplant lymphoproliferative disorders (EBV(-)M-PTLD) have comparable outcomes to EBV(+) PTLD, but are inferior to diffuse large B-cell lymphoma in immunocompetent pediatric patients. The variety of treatment regimens used highlights the need to develop a pediatric PTLD registry to prospectively evaluate outcomes. The impact of treatment regimen on relapse risk could not be assessed because of small numbers. In the intensive pediatric B-non-Hodgkin lymphoma chemoimmunotherapy group, 11 of 13 patients remain alive in complete remission after 0.6 to 11 years.
Collapse
Affiliation(s)
- Zeinab A. M. Afify
- Primary Children’s Hospital, University of Utah, Salt Lake City, Utah, USA
| | - Mary M. Taj
- The Royal Marsden Hospital, Sutton, London, UK
| | | | | | - Tamara P. Miller
- Emory University School of Medicine and Children’s Healthcare of Atlanta, Atlanta, Georgia, USA
| | - Holly J. Edington
- Emory University School of Medicine and Children’s Healthcare of Atlanta, Atlanta, Georgia, USA
- Novant Health Presbyterian Medical Center, Charlotte, North Carolina, USA
| | - Mansi Dalal
- University of Florida Health Science Center, Gainesville, Florida, USA
| | - Joanna Robles
- Duke University School of Medicine, Durham, North Carolina, USA
| | - James B. Ford
- University of Nebraska Medical Center, Omaha, Nebraska, USA
| | | | - Tonya J. Ureda
- St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Jeremy D. Rubinstein
- Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
- University of Cincinnati, College of Medicine, Cincinnati, Ohio, USA
| | - Sarah McCormack
- Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | | | | | - Madison K. Kavanaugh
- Department of Pediatrics, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Andrew J. Bukowinski
- Department of Pediatrics, University of Pittsburgh, School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Erika D. Friehling
- Department of Pediatrics, University of Pittsburgh, School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Maegan C. Ford
- Columbia University Medical Center, New York, New York, USA
| | | | - Lianna J. Marks
- Stanford University School of Medicine, Palo Alto, California, USA
| | | | - Clinton C. Mason
- Department of Pediatrics, University of Utah, Salt Lake City, Utah, USA
| |
Collapse
|
23
|
Koike H, Morikawa M, Ishimaru H, Ideguchi R, Uetani M, Hiu T, Matsuo T, Miyoshi M. Amide proton transfer MRI differentiates between progressive multifocal leukoencephalopathy and malignant brain tumors: a pilot study. BMC Med Imaging 2022; 22:227. [PMID: 36572873 PMCID: PMC9793649 DOI: 10.1186/s12880-022-00959-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 12/22/2022] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Progressive multifocal leukoencephalopathy (PML) is a demyelinating disease of the central nerve system caused by the John Cunningham virus. On MRI, PML may sometimes appear similar to primary central nervous system lymphoma (PCNSL) and glioblastoma multiforme (GBM). The purpose of this pilot study was to evaluate the potential of amide proton transfer (APT) imaging for differentiating PML from PCNSL and GBM. METHODS Patients with PML (n = 4; two men; mean age 52.3 ± 6.1 years), PCNSL (n = 7; four women; mean age 74.4 ± 5.8 years), or GBM (n = 11; 6 men; mean age 65.0 ± 15.2 years) who underwent APT-CEST MRI between January 2021 and September 2022 were retrospectively evaluated. Magnetization transfer ratio asymmetry (MTRasym) values were measured on APT imaging using a region of interest within the lesion. Receiver operating characteristics curve analysis was used to determine diagnostic cutoffs for MTRasym. RESULTS The mean MTRasym values were 0.005 ± 0.005 in the PML group, 0.025 ± 0.005 in the PCNSL group, and 0.025 ± 0.009 in the GBM group. There were significant differences in MTRasym between PML and PCNSL (P = 0.023), and between PML and GBM (P = 0.015). For differentiating PML from PCNSL, an MTRasym threshold of 0.0165 gave diagnostic sensitivity, specificity, positive predictive value, and negative predictive value of 100% (all). For differentiating PML from GBM, an MTRasym threshold of 0.015 gave diagnostic sensitivity, specificity, positive predictive value, and negative predictive value of 100%, 90.9%, 80.0%, and 100%, respectively. CONCLUSION MTRasym values obtained from APT imaging allowed patients with PML to be clearly discriminated from patients with PCNSL or GBM.
Collapse
Affiliation(s)
- Hirofumi Koike
- grid.174567.60000 0000 8902 2273Department of Radiology, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8501 Japan
| | - Minoru Morikawa
- grid.411873.80000 0004 0616 1585Department of Radiology, Nagasaki University Hospital, 1-7-1 Sakamoto, Nagasaki, 852-8501 Japan
| | - Hideki Ishimaru
- grid.411873.80000 0004 0616 1585Department of Radiology, Nagasaki University Hospital, 1-7-1 Sakamoto, Nagasaki, 852-8501 Japan
| | - Reiko Ideguchi
- grid.174567.60000 0000 8902 2273Department of Radioisotope Medicine, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8588 Japan
| | - Masataka Uetani
- grid.174567.60000 0000 8902 2273Department of Radiology, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8501 Japan
| | - Takeshi Hiu
- grid.174567.60000 0000 8902 2273Department of Neurosurgery, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8501 Japan
| | - Takayuki Matsuo
- grid.174567.60000 0000 8902 2273Department of Neurosurgery, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8501 Japan
| | - Mitsuharu Miyoshi
- grid.481637.f0000 0004 0377 9208MR Application and Workflow, GE Healthcare Japan, 4-7-127 Asahigaoka, Hino, Tokyo 191-8503 Japan
| |
Collapse
|
24
|
Vergote VKJ, Deroose CM, Fieuws S, Laleman W, Sprangers B, Uyttebroeck A, Van Cleemput J, Verhoef G, Vos R, Tousseyn T, Dierickx D. Characteristics and Outcome of Post-Transplant Lymphoproliferative Disorders After Solid Organ Transplantation: A Single Center Experience of 196 Patients Over 30 Years. Transpl Int 2022; 35:10707. [PMID: 36589262 PMCID: PMC9794588 DOI: 10.3389/ti.2022.10707] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 11/29/2022] [Indexed: 12/15/2022]
Abstract
Post-transplant lymphoproliferative disorder (PTLD) is a rare but life-threatening complication after transplantation. In this retrospective, monocentric study we aimed to collect real life data regarding PTLD and determine the role of Epstein Barr Virus (EBV) status and year of diagnosis on prognosis. We identified 196 biopsy-proven PTLD after solid organ transplantation (SOT) diagnosed at the University Hospitals Leuven (Belgium) from 1989 to 2019. EBV status was positive in 61% of PTLD. The median overall survival (OS) was 5.7 years (95% CI: 2.99-11.1). Although EBV positivity was not significantly correlated with OS in multivariate analyses (HR: 1.44 (95% CI: 0.93-2.24); p = 0.10), subgroup analysis showed a significantly better median OS for EBV negative post-transplant diffuse large B-cell lymphoma (DLBCL) compared to EBV positive post-transplant DLBCL (8.8 versus 2.5 years respectively; p = 0.0365). There was a significant relation between year of PTLD diagnosis and OS: the more recent the PTLD diagnosis, the lower the risk for death (adjusted HR: 0.962 (95% CI: 0.931-0.933); p = 0.017). In conclusion, the prognosis of PTLD after SOT has improved in the past decades. Our analysis shows a significant relation between EBV status and OS in post-transplant DLBCL.
Collapse
Affiliation(s)
- Vibeke K. J. Vergote
- Department of Hematology, University Hospitals Leuven, Leuven, Belgium,*Correspondence: Vibeke K. J. Vergote, , orcid.org/0000-0003-1100-5600
| | | | - Steffen Fieuws
- Interuniversity Institute for Biostatistics and Statistical Bioinformatics, KU Leuven—University of Leuven, Leuven, Belgium
| | - Wim Laleman
- Department of Liver and Biliopancreatic Disorders, University Hospitals Leuven, Leuven, Belgium
| | - Ben Sprangers
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Leuven, Belgium,Department of Nephrology, University Hospitals Leuven, Leuven, Belgium
| | - Anne Uyttebroeck
- Department of Pediatric Hemato-Oncology, Department of Oncology, University Hospitals Leuven, KU Leuven, Leuven, Belgium
| | | | - Gregor Verhoef
- Department of Hematology, University Hospitals Leuven, Leuven, Belgium
| | - Robin Vos
- Department of Respiratory Medicine, University Hospitals Leuven, Leuven, Belgium,BREATHE, KU Leuven, Leuven, Belgium
| | - Thomas Tousseyn
- Department of Pathology, University Hospitals Leuven, Leuven, Belgium
| | - Daan Dierickx
- Department of Hematology, University Hospitals Leuven, Leuven, Belgium
| |
Collapse
|
25
|
Abstract
PURPOSE OF REVIEW This review provides readers with examples of refractory infections due to inborn errors of immunity, highlighting how they may be successfully treated by deducing and targeting the underlying immunodeficiency. RECENT FINDINGS The use of host-directed immunotherapy to treat infectious disease in inborn errors of immunity is currently limited but growing. Different strategies include depleting the cellular reservoir for pathogens with restricted cell-tropism; augmenting the diminished effector response; and restoring molecular equipoise. The immunotherapies illustrated are existing drugs that have been re-purposed and rationally used, depending on the molecular or cellular impact of the mutation. As more biologic response modifiers and molecular targeted therapies are developed for other indications, they open the avenues for their use in inborn errors of immunity. Conversely, as more molecular pathways underlying defective immune responses and refractory infections are elucidated, they lend themselves to tractability with these emerging therapies. SUMMARY Infections that fail appropriate antimicrobial therapy are a harbinger of underlying inborn errors of immunity. Dissecting the mechanism by which the immune system fails provides opportunities to target the host response and make it succeed.
Collapse
|
26
|
Markouli M, Ullah F, Omar N, Apostolopoulou A, Dhillon P, Diamantopoulos P, Dower J, Gurnari C, Ahmed S, Dima D. Recent Advances in Adult Post-Transplant Lymphoproliferative Disorder. Cancers (Basel) 2022; 14:cancers14235949. [PMID: 36497432 PMCID: PMC9740763 DOI: 10.3390/cancers14235949] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 11/24/2022] [Accepted: 11/27/2022] [Indexed: 12/03/2022] Open
Abstract
PTLD is a rare but severe complication of hematopoietic or solid organ transplant recipients, with variable incidence and timing of occurrence depending on different patient-, therapy-, and transplant-related factors. The pathogenesis of PTLD is complex, with most cases of early PLTD having a strong association with Epstein-Barr virus (EBV) infection and the iatrogenic, immunosuppression-related decrease in T-cell immune surveillance. Without appropriate T-cell response, EBV-infected B cells persist and proliferate, resulting in malignant transformation. Classification is based on the histologic subtype and ranges from nondestructive hyperplasias to monoclonal aggressive lymphomas, with the most common subtype being diffuse large B-cell lymphoma-like PTLD. Management focuses on prevention of PTLD development, as well as therapy for active disease. Treatment is largely based on the histologic subtype. However, given lack of clinical trials providing evidence-based data on PLTD therapy-related outcomes, there are no specific management guidelines. In this review, we discuss the pathogenesis, histologic classification, and risk factors of PTLD. We further focus on common preventive and frontline treatment modalities, as well as describe the application of novel therapies for PLTD and elaborate on potential challenges in therapy.
Collapse
Affiliation(s)
- Mariam Markouli
- Department of Internal Medicine, Laikon General Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Fauzia Ullah
- Department of Translational Hematology and Oncology Research, Cleveland Clinic Foundation, Cleveland, OH 44195, USA
| | - Najiullah Omar
- Department of Translational Hematology and Oncology Research, Cleveland Clinic Foundation, Cleveland, OH 44195, USA
| | - Anna Apostolopoulou
- Division of Infectious Diseases, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Puneet Dhillon
- Department of Internal Medicine, Cleveland Clinic Foundation, Cleveland, OH 44195, USA
| | - Panagiotis Diamantopoulos
- Department of Internal Medicine, Laikon General Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Joshua Dower
- Department of Hematology and Medical Oncology, Tufts Medical Center, Boston, MA 02111, USA
| | - Carmelo Gurnari
- Department of Translational Hematology and Oncology Research, Cleveland Clinic Foundation, Cleveland, OH 44195, USA
| | - Sairah Ahmed
- Department of Lymphoma-Myeloma, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Danai Dima
- Department of Translational Hematology and Oncology Research, Cleveland Clinic Foundation, Cleveland, OH 44195, USA
- Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic Foundation, Cleveland Clinic, Cleveland, OH 44195, USA
- Correspondence:
| |
Collapse
|
27
|
Post-Transplant Lymphoproliferative Disorder: A Rare Case of CNS Involvement following Renal Transplant. J Clin Med 2022; 11:jcm11237058. [PMID: 36498633 PMCID: PMC9739215 DOI: 10.3390/jcm11237058] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 11/12/2022] [Accepted: 11/26/2022] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND Post-transplant lymphoproliferative disorders (PTLD) are rare immunosuppression complications affecting 5% of transplant patients. Isolated central nervous system (CNS)-PTLD without nodal or extra-nodal organ involvement is rarely reported and is difficult to diagnose due to the non-specific clinical manifestations and imaging features overlapping with other common CNS lesions. CASE PRESENTATION We present a case of a 72-year-old female subjected to a renal transplant 11 years ago with progressively worsening headaches and confusion. Imaging revealed vasogenic edema in the left frontal and bilateral temporal lobes. She was subjected to a craniotomy and excisional biopsy to obtain tissue for diagnostic and therapeutic interventions. Pathology examination showed atypical EBV-positive lymphoplasmacytic infiltrate, consistent with Polymorphic type PTLD. CONCLUSIONS Patients diagnosed with PTLD need to have close monitoring of immunosuppressive medications while in the hospital. Early diagnosis is essential for patient survival with PTLD, as their health can deteriorate fast.
Collapse
|
28
|
Matsumura M, Miyagi S, Tokodai K, Kashiwadate T, Fujio A, Miyazawa K, Sasaki K, Saito Y, Kanai N, Unno M, Kamei T. Probable posttransplant lymphoproliferative disorder after pediatric living donor liver transplantation: Is a biopsy still needed? Clin Case Rep 2022; 10:e6454. [PMID: 36348984 PMCID: PMC9634264 DOI: 10.1002/ccr3.6454] [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: 05/14/2022] [Revised: 07/22/2022] [Accepted: 08/25/2022] [Indexed: 11/06/2022] Open
Abstract
Posttransplant lymphoproliferative disorder (PTLD) is a complication of solid organ transplantation and is associated with Epstein-Barr virus (EBV). Recently, EBV-related PTLD was defined as probable PTLD or proven PTLD. Probable PTLD involves significant lymphadenopathy, hepatosplenomegaly, or other end-organ manifestations, without a histological diagnosis, together with significant EBV DNAemia. Proven PTLD is the detection of EBV-encoded proteins in a tissue specimen, together with symptoms and/or signs originating from the affected organ. Probable PTLD after pediatric liver transplantation has not been well documented. Therefore, here, we aimed to describe cases of five pediatric patients with probable PTLD after liver transplantation, who were successfully treated with preemptive immunosuppression reduction with or without rituximab. All five patients (age range, 1-4 years; two girls and three boys) had EBV DNAemia. Three patients developed probable PTLD within 12 months of transplantation. Further, three patients had a significantly high EBV viral load, but the other two patients with lymphadenopathy and end-organ manifestation had a relatively low EBV viral load. Early onset pediatric PTLD with significant EBV DNAemia is almost universally EBV-related. Biopsy was not performed in any patient due to the relative inaccessibility of the lesion and young age of the patients. If the patient's symptoms are too mild, if excisional biopsy is too difficult to perform, or if the patient is too sick to undergo an invasive procedure, initiating preemptive treatment without a histological diagnosis could be the treatment option.
Collapse
Affiliation(s)
- Muneyuki Matsumura
- Department of SurgeryTohoku University Graduate School of MedicineSendaiJapan
| | - Shigehito Miyagi
- Department of SurgeryTohoku University Graduate School of MedicineSendaiJapan
| | - Kazuaki Tokodai
- Department of SurgeryTohoku University Graduate School of MedicineSendaiJapan
| | | | - Atsushi Fujio
- Department of SurgeryTohoku University Graduate School of MedicineSendaiJapan
| | - Koji Miyazawa
- Department of SurgeryTohoku University Graduate School of MedicineSendaiJapan
| | - Kengo Sasaki
- Department of SurgeryTohoku University Graduate School of MedicineSendaiJapan
| | - Yoshikatsu Saito
- Department of SurgeryTohoku University Graduate School of MedicineSendaiJapan
| | - Norifumi Kanai
- Department of SurgeryTohoku University Graduate School of MedicineSendaiJapan
| | - Michiaki Unno
- Department of SurgeryTohoku University Graduate School of MedicineSendaiJapan
| | - Takashi Kamei
- Department of SurgeryTohoku University Graduate School of MedicineSendaiJapan
| |
Collapse
|
29
|
Incidence of Lymphoproliferative Disorders After Renal Transplantation is Down, but the Poor Prognosis Remains. Multicenter 32-Year Cohort Study. Transplant Proc 2022; 54:2462-2466. [DOI: 10.1016/j.transproceed.2022.10.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 10/01/2022] [Indexed: 11/15/2022]
|
30
|
Stubbins RJ, Lam R, Zhu J, Ghosh S, Mabilangan C, Kuruvilla J, Goswami RS, Lai R, Preiksaitis JK, Jain MD, Peters AC. Tumor Infiltrating Lymphocytes Predict Survival in Solid Organ Transplant Recipients With Monomorphic Post-transplant Lymphoproliferative Disorders. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2022; 22:744-752. [PMID: 35717340 DOI: 10.1016/j.clml.2022.05.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 05/19/2022] [Indexed: 06/15/2023]
Abstract
INTRODUCTION The tumor microenvironment (TME) in post-transplant lymphoproliferative disorders (PTLDs) remains unexplored. Tumor infiltrating lymphocytes (TILs) are prognostic in other lymphomas. We assessed the prognostic impact of TILs in monomorphic B-cell PTLD. METHODS TIL density (CD3+ cells/mm2) was determined by CD3 immunohistochemistry in archived diagnostic biopsies from patients diagnosed with monomorphic B-cell PTLD. RESULTS Amongst monomorphic PTLDs (N = 107), low TIL-count was associated with inferior 2-year progression-free survival (PFS) (41% versus 86%, P = .003) and 2-year overall survival (OS) (52% versus 93%, P = .003) by Kaplan-Meier analysis. Low TIL-count was significant on Cox univariate regression for inferior PFS (HR 4.5, 95% CI 2.0-9.9, P < .001) and OS (HR 4.6, 95% CI 1.8-11.8, P < .001). Multivariate analysis with clinical variables (age ≥60 years, high LDH, stage III/IV, CNS involvement) and TIL-count showed significance for PFS (HR 3.3, 95% CI 1.3-8.3, P = .010) and a non-significant trend for OS (HR 2.6, 95% CI 0.9-7.3, P = .064). A composite score including TILs and clinical variables (age ≥60 years, high LDH, stage III/IV, CNS involvement) effectively stratified monomorphic PTLD patients by PFS and OS (2-year OS: low-risk 93%, intermediate-risk 61%, high-risk 23%, P < .001). CONCLUSIONS The TME and TILs are prognostically relevant in monomorphic PTLD. Prognostic models including measures of the TME may improve risk stratification for patients with monomorphic PTLDs.
Collapse
Affiliation(s)
- Ryan J Stubbins
- Leukemia/BMT Program of BC, BC Cancer, Vancouver, BC, Canada
| | - Ryan Lam
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, ON, Canada
| | - James Zhu
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB, Canada
| | - Sunita Ghosh
- Department of Oncology, Cross Cancer Institute, University of Alberta, Edmonton, AB, Canada
| | - Curtis Mabilangan
- Division of Infectious Diseases, Department of Medicine, University of Alberta, AB, Edmonton, Canada
| | - John Kuruvilla
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, ON, Canada
| | - Rashmi S Goswami
- Department of Laboratory Medicine and Molecular Diagnostics, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Raymond Lai
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB, Canada
| | - Jutta K Preiksaitis
- Division of Infectious Diseases, Department of Medicine, University of Alberta, AB, Edmonton, Canada
| | - Michael D Jain
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, H. Lee Moffitt Cancer Center and Research Institute; Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Anthea C Peters
- Department of Oncology, Cross Cancer Institute, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
31
|
Enfermedad linfoproliferativa difusa postrasplante renal: estudio longitudinal de 21.546 receptores durante 2 décadas en España. Nefrologia 2022. [DOI: 10.1016/j.nefro.2022.02.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
32
|
Hybel TE, Vase MØ, Maksten EF, Enemark MB, Lauridsen KL, Hamilton-Dutoit S, Andersen C, Møller MB, Sørensen SS, Jespersen B, Kampmann J, d’Amore F, Ludvigsen M. Intratumoral expression of CD38 in patients with post-transplant lymphoproliferative disorder. Acta Oncol 2021; 60:1637-1642. [PMID: 34474636 DOI: 10.1080/0284186x.2021.1973093] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Affiliation(s)
- Trine Engelbrecht Hybel
- Department of Hematology, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Maja Ølholm Vase
- Department of Hematology, Aarhus University Hospital, Aarhus, Denmark
| | | | - Marie Beck Enemark
- Department of Hematology, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | | | | | - Claus Andersen
- Department of Pathology, Copenhagen University Hospital, Copenhagen, Denmark
| | | | | | - Bente Jespersen
- Department of Nephrology, Aarhus University Hospital, Aarhus, Denmark
| | - Jan Kampmann
- Department of Nephrology, Odense University Hospital, Odense, Denmark
| | - Francesco d’Amore
- Department of Hematology, Aarhus University Hospital, Aarhus, Denmark
| | - Maja Ludvigsen
- Department of Hematology, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
33
|
Epstein-Barr virus posttransplant lymphoproliferative disorder: update on management and outcomes. Curr Opin Infect Dis 2021; 34:635-645. [PMID: 34751183 PMCID: PMC8589110 DOI: 10.1097/qco.0000000000000787] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW Management of Epstein-Barr virus posttransplant lymphoproliferative disorder (EBV PTLD) is complex, involving risk stratification, prevention and/or preemptive measures involving monitoring EBV DNAemia and balancing treatment options, using a combination of reduction of immune suppression, anti-B cell therapy, and cytotoxic T lymphocytes (CTLs). RECENT FINDINGS The highest risk factor for the development of EBV PTLD in hematopoietic cell transplant (HCT) remains T cell depletion, with increasing use of antithymocyte globulin (ATG) or alemtuzumab in conditioning. In solid organ transplantation (SOT), the incidence of PTLD is highest among EBV seronegative recipients who are at risk for primary EBV infection following transplant in the first 12 months. Prevention is a critical component of the management of EBV PTLD. Although preemptive therapy remains standard of care, there continues to be heterogenicity and debate over the optimal choice of EBV DNA quantification and the threshold to use. Novel therapies such as donor-derived multipathogen and EBV specific CTLs for the prevention and third party CTLs for the treatment of EBV PTLD are promising, with rapidly expanding evidence, including large scale Phase III trials currently underway. SUMMARY With an increasing number of risk groups for developing EBV PTLD in HCT and SOT, management strategies using prophylaxis or preemptive therapy remain standard of care, however the use of prophylactic or preemptive EBV specific or multipathogen CTLs show promising results and safety profiles.
Collapse
|
34
|
Yokoyama N, Kanbayashi T, Kobayashi S, Ishida T, Sonoo M. [Subacute cognitive impairment and urinary retention due to primary central nervous system post-transplant lymphoproliferative disorder: a case report]. Rinsho Shinkeigaku 2021; 61:750-755. [PMID: 34657922 DOI: 10.5692/clinicalneurol.cn-001623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
We report a 66-year-old man with primary central nervous system post-transplant lymphoproliferative disorder (PCNS-PTLD). He had received a living-donor kidney transplantation at the age of 64 years. Although he had a good postoperative course by continuing to take oral immunosuppressive agents, he was admitted to our hospital for subacute cognitive impairment and urinary retention two years after the transplantation. Brain MRI revealed high-intensity lesions on FLAIR and T2-weighted images in the left parietal operculum, deep white matter around the anterior horn of the lateral ventricle, and the genu and splenium of the corpus callosum. A part of these lesions showed ring enhancement. The cerebrospinal fluid examination revealed lymphocytic pleocytosis, elevation of protein level, and mild hypoglycorrhachia. Blood tests showed no abnormalities except for positive serum VCA-IgG antibody of Epstein-Barr virus. A brain biopsy was performed and diagnosis of PCNS-PTLD was made. There was no evidence of systemic PTLD. We reduced the dose of immunosuppressive agents and started the initial treatment with methylprednisolone pulse therapy. The patient showed a partial response to the treatment and transferred to another hospital for subsequent chemotherapy. PTLD is an important post-transplant complication that can affect the patient's prognosis. The incidence of PTLD is increasing with the growing numbers of transplantations and older age of donors and recipients. Although CNS involvement is known to be rare, PCNS-PTLD is an important differential diagnosis when symptoms of CNS origin develop in post-transplant patients.
Collapse
Affiliation(s)
| | | | | | - Tsuyoshi Ishida
- Department of Pathology, Teikyo University School of Medicine
| | - Masahiro Sonoo
- Department of Neurology, Teikyo University School of Medicine
| |
Collapse
|
35
|
Tse E, Au-Yeung R, Chau D, Hwang YY, Loong F, Kwong YL. Epstein-Barr virus-positive diffuse large B-cell lymphoma after frontline brentuximab vedotin treatment of classical Hodgkin lymphoma. Ann Hematol 2021; 101:1149-1152. [PMID: 34757467 DOI: 10.1007/s00277-021-04709-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 10/25/2021] [Indexed: 11/27/2022]
Affiliation(s)
- Eric Tse
- Department of Medicine, Professorial Block, Queen Mary Hospital, Pok Fu Lam Rd, Hong Kong, China
| | - Rex Au-Yeung
- Department of Pathology, Queen Mary Hospital, Hong Kong, China
| | - David Chau
- Department of Medicine, Professorial Block, Queen Mary Hospital, Pok Fu Lam Rd, Hong Kong, China
| | - Yu-Yan Hwang
- Department of Medicine, Professorial Block, Queen Mary Hospital, Pok Fu Lam Rd, Hong Kong, China
| | - Florence Loong
- Department of Pathology, Queen Mary Hospital, Hong Kong, China
| | - Yok-Lam Kwong
- Department of Medicine, Professorial Block, Queen Mary Hospital, Pok Fu Lam Rd, Hong Kong, China.
| |
Collapse
|
36
|
Boyle S, Tobin JWD, Perram J, Hamad N, Gullapalli V, Barraclough A, Singaraveloo L, Han MH, Blennerhassett R, Nelson N, Johnston AM, Talaulikar D, Karpe K, Bhattacharyya A, Cheah CY, Subramoniapillai E, Bokhari W, Lee C, Hawkes EA, Jabbour A, Strasser SI, Chadban SJ, Brown C, Mollee P, Hapgood G. Management and Outcomes of Diffuse Large B-cell Lymphoma Post-transplant Lymphoproliferative Disorder in the Era of PET and Rituximab: A Multicenter Study From the Australasian Lymphoma Alliance. Hemasphere 2021; 5:e648. [PMID: 34651103 PMCID: PMC8505336 DOI: 10.1097/hs9.0000000000000648] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 09/08/2021] [Indexed: 11/26/2022] Open
Abstract
There are limited data on post-transplant lymphoproliferative disorder (PTLD) in the era of positron emission tomography (PET) and rituximab (R). Furthermore, there is limited data on the risk of graft rejection with modern practices in reduction in immunosuppression (RIS). We studied 91 patients with monomorphic diffuse large B-cell lymphoma PTLD at 11 Australian centers: median age 52 years, diagnosed between 2004 and 2017, median follow-up 4.7 years (range, 0.5-14.5 y). RIS occurred in 88% of patients. For patients initially treated with R-monotherapy, 45% achieved complete remission, rising to 71% with the addition of rituximab, cyclophosphamide, doxorubicin, vincristine, prednisolone (R-CHOP) for those not in complete remission. For patients initially treated with R-CHOP, the complete remission rate was 76%. There was no difference in overall survival (OS) between R-monotherapy and R-chemotherapy patients. There was no difference in OS for patients with systemic lymphoma (n = 68) versus central nervous system (CNS) involvement (n = 23) (3-y OS 72% versus 73%; P = 0.78). Treatment-related mortality was 7%. End of treatment PET was prognostic for patients with systemic lymphoma with longer OS in the PET negative group (3-y OS 91% versus 57%; P = 0.01). Graft rejection occurred in 9% (n = 4 biopsy-proven; n = 4 suspected) during the entire follow-up period with no cases of graft loss. RIS and R-based treatments are safe and effective with a low likelihood of graft rejection and high cure rate for patients achieving complete remission with CNS or systemic PTLD.
Collapse
Affiliation(s)
- Stephen Boyle
- Department of Haematology, Princess Alexandra Hospital, Brisbane, Queensland, Australia
- University of Queensland, Brisbane, Queensland, Australia
| | - Joshua W. D. Tobin
- University of Queensland, Brisbane, Queensland, Australia
- Department of Haematology, Gold Coast University Hospital, Gold Coast, Queensland, Australia
| | - Jacinta Perram
- Institute of Haematology, Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
| | - Nada Hamad
- Department of Haematology, St Vincent’s Hospital, Sydney, New South Wales, Australia
- Faculty of Medicine, University of New South Wales, Sydney, New South Wales, Australia
| | - Veena Gullapalli
- Department of Haematology, St Vincent’s Hospital, Sydney, New South Wales, Australia
| | - Allison Barraclough
- Department of Haematology and Olivia Newton John Cancer Research Institute, The Austin Hospital, Melbourne, Victoria, Australia
| | | | - Min-Hi Han
- Department of Haematology, Sir Charles Gairdner Hospital, Nedlands, Western Australia, Australia
| | - Richard Blennerhassett
- Department of Haematology, Westmead Hospital, Sydney, New South Wales, Australia
- University of Sydney, Sydney, New South Wales, Australia
| | - Niles Nelson
- Royal Hobart Hospital, Hobart, Tasmania, Australia
| | - Anna M. Johnston
- Royal Hobart Hospital, Hobart, Tasmania, Australia
- University of Tasmania, Hobart, Tasmania, Australia
| | - Dipti Talaulikar
- Department of Haematology, Canberra Hospital, Canberra, Australian Capital Territory, Australia
- Australian National University, Canberra, Australian Capital Territory, Australia
| | - Krishna Karpe
- Department of Renal Medicine, Canberra Hospital, Canberra, Australian Capital Territory, Australia
| | - Abir Bhattacharyya
- Department of Haematology, Westmead Hospital, Sydney, New South Wales, Australia
| | - Chan Yoon Cheah
- Department of Haematology, Sir Charles Gairdner Hospital, Nedlands, Western Australia, Australia
- University of Western Australia, Crawley, Western Australia, Australia
| | | | - Waqas Bokhari
- Royal Brisbane Hospital, Brisbane, Queensland, Australia
| | - Cindy Lee
- Royal Adelaide Hospital, Adelaide, South Australia, Australia
| | - Eliza A. Hawkes
- Department of Haematology and Olivia Newton John Cancer Research Institute, The Austin Hospital, Melbourne, Victoria, Australia
- Eastern Health, Box Hill, Victoria, Australia
- University of Melbourne, Melbourne, Victoria, Australia
| | - Andrew Jabbour
- Department of Cardiology, St Vincent’s Hospital, Sydney, New South Wales, Australia
| | - Simone I. Strasser
- University of Sydney, Sydney, New South Wales, Australia
- AW Morrow Gastroenterology and Liver Centre, Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
| | - Steven J. Chadban
- University of Sydney, Sydney, New South Wales, Australia
- Department of Renal Medicine, Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
- Kidney Node, Charles Perkins Centre, University of Sydney, Sydney, New South Wales, Australia
| | - Christina Brown
- Institute of Haematology, Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
- University of Sydney, Sydney, New South Wales, Australia
| | - Peter Mollee
- Department of Haematology, Princess Alexandra Hospital, Brisbane, Queensland, Australia
- University of Queensland, Brisbane, Queensland, Australia
| | - Greg Hapgood
- Department of Haematology, Princess Alexandra Hospital, Brisbane, Queensland, Australia
- University of Queensland, Brisbane, Queensland, Australia
| |
Collapse
|
37
|
Feng G, Li Q, Zhu H, Jiang Y, Yuan J, Fu Y, Deng Q. Safety and Efficacy of Anti-CD19-Chimeric Antigen Receptor T Cell Combined With Programmed Cell Death 1 Inhibitor Therapy in a Patient With Refractory Post-Transplant Lymphoproliferative Disease: Case Report and Literature Review. Front Oncol 2021; 11:726134. [PMID: 34604065 PMCID: PMC8481808 DOI: 10.3389/fonc.2021.726134] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 08/27/2021] [Indexed: 12/12/2022] Open
Abstract
Post-transplant lymphoproliferative disease (PTLD) often exhibits poor prognosis and high mortality, and there are no uniform guidelines for the treatment of this disease. Anti-CD19 chimeric antigen receptor (CAR) T cells show significant efficacy in treatment of relapse/refractory diffuse large B-cell lymphoma (DLBCL). Treatment using anti-CD19-CAR T-cell therapy in PTLD has been limited by immunosuppressants and has not been widely employed. In this study, a refractory post kidney transplant DLBCL patient with a high tumor burden was enrolled in a clinical trial of anti-CD19-CAR T-cell therapy. The tacrolimus dose was not decreased during combination chemotherapy, as the creatinine level of the patient increased. To improve the function of autologous T cells, combination therapy with anti-CD19-CAR T cells and programmed cell death 1 (PD-1) inhibitors was selected. After treatment with the combination therapy, the patient was diagnosed with grade 1 cytokine release syndrome and grade 3 immune effector cell-associated neurotoxicity syndrome. The amplification peak of anti-CD19-CAR T cells reached 9.01% on day 7. With PD-1 inhibitor maintenance therapy, his disease was maintained in partial remission for 18 weeks. However, his tumor suddenly increased in size, and he discontinued the treatment, including radiation therapy. The anti-CD19-CAR T cell and PD-1 inhibitors have a combined effect on PTLD, and this combination therapy needs to be further explored.
Collapse
Affiliation(s)
- Gang Feng
- Department of Kidney Transplantation, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin, China
| | - Qing Li
- Department of Hematology, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin, China
| | - Haibo Zhu
- Department of Hematology, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin, China
| | - Yanyu Jiang
- Department of Hematology, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin, China
| | - Jijun Yuan
- Shanghai Genbase Biotechnology Co. Ltd., Shanghai, China
| | - Yingxin Fu
- Department of Kidney Transplantation, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin, China
| | - Qi Deng
- Department of Kidney Transplantation, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin, China
| |
Collapse
|
38
|
Ashrafi F, Shahidi S, Mehrzad V, Mortazavi M, Hosseini SF. Survival of Post-Transplant Lymphoproliferative Disorder after Kidney Transplantation in Patients under Rapamycin Treatment. Int J Hematol Oncol Stem Cell Res 2021; 15:239-248. [PMID: 35291663 PMCID: PMC8888361 DOI: 10.18502/ijhoscr.v15i4.7479] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Accepted: 12/12/2020] [Indexed: 11/24/2022] Open
Abstract
Background: One of the important causes of mortality and morbidity in kidney transplanted patients is Post Transplant Lymphoproliferative Disease (PTLD), which is due to immunosuppression therapy and viral activity. It seems that Rapamycin, with dual antineoplastic and immunosuppressive effects, may have a pivotal role in the treatment of PTLD patients and preserving transplanted kidneys. Methods and Materials: Twenty patients with PTLD were enrolled. Immunosuppressive therapy was reduced or ceased, and Rapamycin was initiated at the time of PTLD diagnosis. We evaluated the effects of switching immunosuppressive drugs to Rapamycin on graft status, the response of tumor, and 6, 12 months, and 5-year survival in patients. Results: PTLD remission was achieved in 14 patients, while six patients died; no relapse was detected in recovered patients. The median of PTLD free time was 25 months, and the mean overall survival in patients with PTLD treated by Rapamycin was 84.8 (95% CI=61.3-108.23).The five-year survival rate was 67%, 12 months survival was 73.8%, and six months' survival was 80%. The response rate to Rapamycin and immunosuppression reduction alone was 46.6%. Four out of 13 Diffuse Large B-Cell Lymphoma patients achieved a complete response just only after the reduction of immunosuppressive drugs and the consumption of Rapamycin. Conclusion: The present study demonstrated the effectiveness of conversion from immunosuppressive medication, particularly of Calcineurin inhibitors to Rapamycin in PTLD patients. However, more research is needed to confirm the Rapamycin effect on patients with PTLD.
Collapse
Affiliation(s)
- Farzaneh Ashrafi
- Hematology Oncology Division, Department of Internal Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Shahrzad Shahidi
- Nephrology Division, Department of Internal Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Valiollah Mehrzad
- Department of Clinical Oncology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | | | - Sayyideh Forough Hosseini
- Department of Oncology, Hematology, Cancer Prevention Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
39
|
King RL, Khurana A, Mwangi R, Fama A, Ristow KM, Maurer MJ, Macon WR, Ansell SM, Bennani NN, Kudva YC, Walker RC, Watt KD, Schwab TR, Kushwaha SS, Cerhan JR, Habermann TM. Clinicopathologic Characteristics, Treatment, and Outcomes of Post-transplant Lymphoproliferative Disorders: A Single-institution Experience Using 2017 WHO Diagnostic Criteria. Hemasphere 2021; 5:e640. [PMID: 34514344 PMCID: PMC8423401 DOI: 10.1097/hs9.0000000000000640] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Accepted: 08/09/2021] [Indexed: 12/31/2022] Open
Abstract
The World Health Organization Classification of Tumours of Haematopoietic and Lymphoid Tissues (WHO 2017) included updated criteria for diagnosis and classification of post-transplant lymphoproliferative disorders (PTLDs). This study evaluated the clinicopathologic spectrum using WHO 2017 criteria and adult PTLD patients' outcomes over 30 years between 1987 and 2017 at Mayo Clinic (Rochester, MN). Patients were retrospectively reviewed for clinical features, outcomes, and diagnostic pathology material and classified based on WHO 2017 criteria. A total of 227 patients were diagnosed with PTLD, with a median time from transplant to PTLD of 45 months. PTLD occurred >1 year after transplant in 149 (66%) patients. Monomorphic PTLD was the most common subtype (173, 76%), with diffuse large B cell lymphoma as the commonest morphology (n = 137). Epstein-Barr virus was positive in 61% of total cases and 90% of PTLD that developed within 1 year from transplant. The median event-free survival (EFS) and overall survival for the entire cohort were 21 months (95% confidence interval [CI]: 9-35) and 82 months (95% CI: 39-115), respectively. The EFS or overall survival was not impacted by Epstein-Barr virus status but differed based on WHO subtypes and year of diagnosis. Management changed over time with increased use of rituximab or chemotherapy + immunosuppression reduction as initial therapy. When compared to the matched general population and de novo diffuse large B cell lymphoma, patients not achieving EFS 24 status (no progression/treatment or death within 24 mo of diagnosis) had a worse standardized mortality ratio 16.75 (95% CI: 13.91-20) versus SMR 1.72 (95% CI: 1.26-2.28) in those who achieved EFS24. Cause of death was mostly attributed to non-lymphoma-related causes in those achieving EFS 24.
Collapse
Affiliation(s)
- Rebecca L. King
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | - Arushi Khurana
- Division of Hematology, Mayo Clinic, Rochester, Minnesota, USA
| | - Raphael Mwangi
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota, USA
| | - Angelo Fama
- Hematology, Azienda Unità Sanitaria Locale di Reggio Emilia - IRCCS, Italy
| | - Kay M. Ristow
- Division of Hematology, Mayo Clinic, Rochester, Minnesota, USA
| | - Matthew J. Maurer
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota, USA
| | - William R. Macon
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | | | - N. Nora Bennani
- Division of Hematology, Mayo Clinic, Rochester, Minnesota, USA
| | - Yogish C. Kudva
- Department of Endocrinology, Diabetes and Metabolism, Mayo Clinic, Rochester, Minnesota, USA
| | - Randall C. Walker
- Department of Infectious Disease, Mayo Clinic, Rochester, Minnesota, USA
| | - Kymberly D. Watt
- Division of Gastroenterology/Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| | - Thomas R. Schwab
- Department of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Sudhir S. Kushwaha
- 9Division of Cardiovascular Diseases, Mayo Clinic, Rochester, Minnesota, USA
| | - James R. Cerhan
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota, USA
| | | |
Collapse
|
40
|
Zierhut H, Kanzelmeyer N, Buescher A, Höcker B, Mauz-Körholz C, Tönshoff B, Metzler M, Pohl M, Pape L, Maecker-Kolhoff B. Course of renal allograft function after diagnosis and treatment of post-transplant lymphoproliferative disorders in pediatric kidney transplant recipients. Pediatr Transplant 2021; 25:e14042. [PMID: 34021949 DOI: 10.1111/petr.14042] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 03/23/2021] [Accepted: 04/23/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND Post-transplant lymphoproliferative disease (PTLD) is a life-threatening complication in renal transplant recipients. Immunomodulatory and chemotherapeutic treatment potentially affect allograft function. The aim of this study was to evaluate graft function of pediatric kidney transplant recipients following diagnosis and standardized treatment of PTLD. METHODS Patients were identified from the German Ped-PTLD registry, and data on renal function were retrospectively retrieved from patient charts. For PTLD treatment, immunosuppressive therapy was reduced and all children received rituximab (375 mg/m2 ) for up to six doses. Two patients required additional low-dose chemotherapy. Renal allograft function was monitored by consecutive measurements of estimated glomerular filtration rate (eGFR) at defined time points. Follow-up was up to 60 months after PTLD. RESULTS Twenty patients were included in this cohort analysis. Median time from transplantation to PTLD was 2.4 years. Histopathology showed monomorphic lesions in 16 and polymorphic in 4 patients. Two patients experienced PTLD relapse after 2 and 14 months. Range-based analysis of variance showed stable allograft function in 17 of 20 patients (85%). Mean eGFR increased during early treatment phase. One patient experienced graft rejection 5.3 years after diagnosis of PTLD. Another patient developed recurrence of primary renal disease (focal-segmental glomerulosclerosis) and lost his renal allograft 3.8 years post-transplant (2.0 years after PTLD diagnosis). CONCLUSION Treatment of PTLD with rituximab with or without low-dose chemotherapy in combination with reduced immunosuppression, mostly comprising of an mTOR inhibitor-based, calcineurin inhibitor-free regimen, is associated with stable graft function and favorable graft survival in pediatric renal transplant patients.
Collapse
Affiliation(s)
- Henriette Zierhut
- Department of Pediatric Hematology and Oncology, Hannover Medical School, Hannover, Germany
| | - Nele Kanzelmeyer
- Department of Pediatric Kidney, Liver and Metabolic Diseases, Hannover Medical School, Hannover, Germany
| | - Anja Buescher
- Department of Pediatric Nephrology, University Hospital of Essen, Essen, Germany
| | - Britta Höcker
- Department of Pediatrics I, University Children's Hospital, Heidelberg, Germany
| | - Christine Mauz-Körholz
- Pediatric Hematology and Oncology, Gießen and Medical Faculty of the Martin-Luther University of Halle, Justus-Liebig-University, Giessen, Germany
| | - Burkhard Tönshoff
- Department of Pediatrics I, University Children's Hospital, Heidelberg, Germany
| | - Markus Metzler
- Department of Pediatrics, University Hospital Erlangen, Erlangen, Germany
| | - Martin Pohl
- Department of General Pediatrics, Adolescent Medicine and Neonatology, Faculty of Medicine, Medical Center - University of Freiburg, University of Freiburg, Freiburg, Germany
| | - Lars Pape
- Department of Pediatric Kidney, Liver and Metabolic Diseases, Hannover Medical School, Hannover, Germany
| | - Britta Maecker-Kolhoff
- Department of Pediatric Hematology and Oncology, Hannover Medical School, Hannover, Germany
| |
Collapse
|
41
|
EBV-associated primary CNS lymphoma occurring after immunosuppression is a distinct immunobiological entity. Blood 2021; 137:1468-1477. [PMID: 33202420 DOI: 10.1182/blood.2020008520] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 11/08/2020] [Indexed: 02/07/2023] Open
Abstract
Primary central nervous system lymphoma (PCNSL) is confined to the brain, eyes, and cerebrospinal fluid without evidence of systemic spread. Rarely, PCNSL occurs in the context of immunosuppression (eg, posttransplant lymphoproliferative disorders or HIV [AIDS-related PCNSL]). These cases are poorly characterized, have dismal outcome, and are typically Epstein-Barr virus (EBV)-associated (ie, tissue-positive). We used targeted sequencing and digital multiplex gene expression to compare the genetic landscape and tumor microenvironment (TME) of 91 PCNSL tissues all with diffuse large B-cell lymphoma histology. Forty-seven were EBV tissue-negative: 45 EBV- HIV- PCNSL and 2 EBV- HIV+ PCNSL; and 44 were EBV tissue-positive: 23 EBV+ HIV+ PCNSL and 21 EBV+ HIV- PCNSL. As with prior studies, EBV- HIV- PCNSL had frequent MYD88, CD79B, and PIM1 mutations, and enrichment for the activated B-cell (ABC) cell-of-origin subtype. In contrast, these mutations were absent in all EBV tissue-positive cases and ABC frequency was low. Furthermore, copy number loss in HLA class I/II and antigen-presenting/processing genes were rarely observed, indicating retained antigen presentation. To counter this, EBV+ HIV- PCNSL had a tolerogenic TME with elevated macrophage and immune-checkpoint gene expression, whereas AIDS-related PCNSL had low CD4 gene counts. EBV-associated PCNSL in the immunosuppressed is immunobiologically distinct from EBV- HIV- PCNSL, and, despite expressing an immunogenic virus, retains the ability to present EBV antigens. Results provide a framework for targeted treatment.
Collapse
|
42
|
Central nervous system infections after solid organ transplantation. Curr Opin Infect Dis 2021; 34:207-216. [PMID: 33741794 DOI: 10.1097/qco.0000000000000722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
PURPOSE OF REVIEW Significant advances to our understanding of several neuroinfectious complications after a solid organ transplant (SOT) have occurred in the last few years. Here, we review the central nervous system (CNS) infections that are relevant to SOT via a syndromic approach with a particular emphasis on recent updates in the field. RECENT FINDINGS A few key studies have advanced our understanding of the epidemiology and clinical characteristics of several CNS infections in SOT recipients. Risk factors for poor prognosis and protective effects of standard posttransplant prophylactic strategies have been better elucidated. Newer diagnostic modalities which have broad clinical applications like metagenomic next-generation sequencing, as well as those that help us better understand esoteric concepts of disease pathogenesis have been studied. Finally, several studies have provided newer insights into the treatment of these diseases. SUMMARY Recent findings reflect the steady progress in our understanding of CNS infections post SOT. They provide several avenues for improvement in the prevention, early recognition, and therapeutic outcomes of these diseases.
Collapse
|
43
|
Anastasiou M, Mamez AC, Masouridi S, Vargas MI, Hadaya K, Egervari K, Chalandon Y. Successful treatment of central nervous system lymphoproliferative disorder in a kidney-pancreas and stem cell transplanted patient using intrathecal rituximab. BMJ Case Rep 2021; 14:14/8/e238236. [PMID: 34353823 PMCID: PMC8344276 DOI: 10.1136/bcr-2020-238236] [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] [Indexed: 12/20/2022] Open
Abstract
Central nervous system lymphoproliferative disorder (CNS-PTLD) after organ transplant is a unique clinicopathological entity and is associated with poor survival rates. When the CNS is involved, intravenous rituximab might not be the treatment of choice, due to its poor CNS penetration. However, intrathecal (IT) administration of rituximab has shown to be safe and efficient in small studies and in case series. We report here the case of a patient with late development of CNS-PTLD after kidney-pancreas transplantation who achieved complete remission after surgical resection and four cycles of IT rituximab and we provide a review of the literature for this treatment option.
Collapse
Affiliation(s)
- Maria Anastasiou
- Oncology, Division Hematology, Hopitaux Universitaires de Geneve, Geneva, Switzerland
| | - Anne-Claire Mamez
- Oncology, Division Hematology, Hopitaux Universitaires de Geneve, Geneva, Switzerland
| | - Stavroula Masouridi
- Oncology, Division Hematology, Hopitaux Universitaires de Geneve, Geneva, Switzerland
| | | | - Karine Hadaya
- Nephrology, Hopitaux Universitaires de Geneve, Geneva, Switzerland
| | - Kristof Egervari
- Service of Clinical Pathology, Department of Genetic Medicine, Laboratory and Pathology, Hopitaux Universitaires de Geneve, Geneva, Switzerland
| | - Yves Chalandon
- Oncology, Division Hematology, Hopitaux Universitaires de Geneve, Geneva, Switzerland
| |
Collapse
|
44
|
Pearse WB, Petrich AM, Gordon LI, Karmali R, Winter JN, Ma S, Kaplan JB, Behdad A, Klein A, Jovanovic B, Helenowski I, Smith SM, Evens AM, Pro B. A phase I/II trial of brentuximab vedotin plus rituximab as frontline therapy for patients with immunosuppression-associated CD30+ and/or EBV + lymphomas. Leuk Lymphoma 2021; 62:3493-3500. [PMID: 34338127 DOI: 10.1080/10428194.2021.1957867] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Treatment strategies for post-transplant lymphoproliferative disorders (PTLD) consist of response-adapted risk-stratified methods using immunosuppression reduction, immunotherapy, and chemotherapy. We investigated the efficacy of Brentuximab vedotin given concurrently with Rituximab (BV + R) once weekly for four weeks, followed by optional consolidation, and up to one year of maintenance. Among 20 assessable patients, BV + R therapy resulted in an overall response rate of 75% (95% CI 51 to 91, p = 0.044) with 60% achieving a complete response. Median time to best response was 28 days. Two-year progression-free survival and overall survival rates were 75 and 90%, respectively. Most common severe grade 3/4 treatment-related toxicities included neutropenia (40%), hypertension (30%), infection (25%), and peripheral neuropathy (15%). BV + R is a novel and effective therapeutic strategy that achieved rapid and durable remissions in previously untreated PTLD patients; however, this treatment platform requires further modification due to the high rates of treatment-related toxicity.Key pointsBrentuximab vedotin + Rituximab showed ORR and CR rates of 75 and 60% in patients with immunosuppression-associated lymphoid malignanciesHigh rates of treatment delay were attributed to treatment-related toxicity; further dosing optimization of this regimen is required.
Collapse
Affiliation(s)
- William B Pearse
- Division of Hematology/Oncology, Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Adam M Petrich
- Division of Hematology/Oncology, Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Leo I Gordon
- Division of Hematology/Oncology, Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Reem Karmali
- Division of Hematology/Oncology, Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Jane N Winter
- Division of Hematology/Oncology, Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Shuo Ma
- Division of Hematology/Oncology, Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Jason B Kaplan
- Division of Hematology/Oncology, Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Amir Behdad
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Andreas Klein
- Department of Hematology/Oncology, Tufts Medical Center, Boston, MA, USA
| | - Borko Jovanovic
- Department of Preventive Medicine, Northwestern University, Chicago, IL, USA
| | - Irene Helenowski
- Department of Preventive Medicine, Northwestern University, Chicago, IL, USA
| | - Sonali M Smith
- Section of Hematology/Oncology, University of Chicago, Chicago, IL, USA
| | - Andrew M Evens
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA
| | - Barbara Pro
- Division of Hematology/Oncology, Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| |
Collapse
|
45
|
Tajima T, Hata K, Haga H, Nishikori M, Umeda K, Kusakabe J, Miyauchi H, Okamoto T, Ogawa E, Sonoda M, Hiramatsu H, Fujimoto M, Okajima H, Takita J, Takaori‐Kondo A, Uemoto S. Post-transplant Lymphoproliferative Disorders After Liver Transplantation: A Retrospective Cohort Study Including 1954 Transplants. Liver Transpl 2021; 27:1165-1180. [PMID: 33655645 PMCID: PMC8453854 DOI: 10.1002/lt.26034] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Revised: 01/29/2021] [Accepted: 02/17/2021] [Indexed: 12/11/2022]
Abstract
Post-transplant lymphoproliferative disorders (PTLDs) are life-threatening neoplasms after organ transplantation. Because of their rarity and multiple grades of malignancy, the incidence, outcomes, and clinicopathological features affecting patient survival after liver transplantation (LT) remain unclear. We reviewed 1954 LTs in 1849 recipients (1990-2020), including 886 pediatric (<18 years of age) and 963 adult recipients. The following clinicopathological factors were studied: age, sex, liver etiologies, malignancy grades, Epstein-Barr virus status, performance status (PS), Ann Arbor stage, international prognostic index, and histopathological diagnosis. Of 1849 recipients, 79 PTLD lesions (4.3%) were identified in 70 patients (3.8%). After excluding 3 autopsy cases incidentally found, 67 (45 pediatric [5.1%] and 22 adult [2.3%]) patients were finally enrolled. Comorbid PTLDs significantly worsened recipient survival compared with non-complicated cases (P < 0.001). The 3-year, 5-year, and 10-year overall survival rates after PTLD diagnosis were 74%, 66%, and 58%, respectively. The incidence of PTLDs after LT (LT-PTLDs) was significantly higher (P < 0.001) with earlier onset (P = 0.002) in children, whereas patient survival was significantly worse in adults (P = 0.002). Univariate and multivariate analyses identified the following 3 prognostic factors: age at PTLD diagnosis ≥18 years (hazard ratio [HR], 11.2; 95% confidence interval [CI], 2.63-47.4; P = 0.001), PS ≥2 at diagnosis (HR, 6.77; 95% CI, 1.56-29.3; P = 0.01), and monomorphic type (HR, 6.78; 95% CI, 1.40-32.9; P = 0.02). A prognostic index, the "LT-PTLD score," that consists of these 3 factors effectively stratified patient survival and progression-free survival (P = 0.003 and <0.001, respectively). In conclusion, comorbid PTLDs significantly worsened patient survival after LT. Age ≥18 years and PS ≥2 at PTLD diagnosis, and monomorphic type are independent prognostic factors, and the LT-PTLD score that consists of these 3 factors may distinguish high-risk cases and guide adequate interventions.
Collapse
Affiliation(s)
- Tetsuya Tajima
- Department of SurgeryDivision of Hepato‐Biliary‐Pancreatic Surgery and Transplantation/Pediatric SurgeryKyoto University Graduate School of MedicineKyotoJapan
| | - Koichiro Hata
- Department of SurgeryDivision of Hepato‐Biliary‐Pancreatic Surgery and Transplantation/Pediatric SurgeryKyoto University Graduate School of MedicineKyotoJapan
| | - Hironori Haga
- Department of Diagnostic PathologyKyoto University HospitalKyotoJapan
| | - Momoko Nishikori
- Department of Hematology and OncologyKyoto University Graduate School of MedicineKyotoJapan
| | - Katsutsugu Umeda
- Department of PediatricsKyoto University Graduate School of MedicineKyotoJapan
| | - Jiro Kusakabe
- Department of SurgeryDivision of Hepato‐Biliary‐Pancreatic Surgery and Transplantation/Pediatric SurgeryKyoto University Graduate School of MedicineKyotoJapan
| | - Hidetaka Miyauchi
- Department of SurgeryDivision of Hepato‐Biliary‐Pancreatic Surgery and Transplantation/Pediatric SurgeryKyoto University Graduate School of MedicineKyotoJapan
| | - Tatsuya Okamoto
- Department of SurgeryDivision of Hepato‐Biliary‐Pancreatic Surgery and Transplantation/Pediatric SurgeryKyoto University Graduate School of MedicineKyotoJapan
| | - Eri Ogawa
- Department of SurgeryDivision of Hepato‐Biliary‐Pancreatic Surgery and Transplantation/Pediatric SurgeryKyoto University Graduate School of MedicineKyotoJapan
| | - Mari Sonoda
- Department of SurgeryDivision of Hepato‐Biliary‐Pancreatic Surgery and Transplantation/Pediatric SurgeryKyoto University Graduate School of MedicineKyotoJapan
| | - Hidefumi Hiramatsu
- Department of PediatricsKyoto University Graduate School of MedicineKyotoJapan
| | - Masakazu Fujimoto
- Department of Diagnostic PathologyKyoto University HospitalKyotoJapan
| | - Hideaki Okajima
- Department of SurgeryDivision of Hepato‐Biliary‐Pancreatic Surgery and Transplantation/Pediatric SurgeryKyoto University Graduate School of MedicineKyotoJapan
| | - Junko Takita
- Department of PediatricsKyoto University Graduate School of MedicineKyotoJapan
| | - Akifumi Takaori‐Kondo
- Department of Hematology and OncologyKyoto University Graduate School of MedicineKyotoJapan
| | - Shinji Uemoto
- Department of SurgeryDivision of Hepato‐Biliary‐Pancreatic Surgery and Transplantation/Pediatric SurgeryKyoto University Graduate School of MedicineKyotoJapan
| |
Collapse
|
46
|
Mudigonda P, Berardi C, Chetram V, Barac A, Cheng R. Implications of cancer prior to and after heart transplantation. Heart 2021; 108:414-421. [PMID: 34210749 DOI: 10.1136/heartjnl-2020-318139] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Accepted: 06/02/2021] [Indexed: 12/11/2022] Open
Abstract
Cancer and cardiovascular disease share many risk factors. Due to improved survival of patients with cancer, the cohort of cancer survivors with heart failure referred for heart transplantation (HT) is growing. Specific considerations include time interval between cancer treatment and HT, risk for recurrence and risk for de novo malignancy (dnM). dnM is an important cause of post-HT morbidity and mortality, with nearly a third diagnosed with malignancy by 10 years post-HT. Compared with the age-matched general population, HT recipients have an approximately 2.5-fold to 4-fold increased risk of developing cancer. HT recipients with prior malignancy show variable cancer recurrence rates, depending on years in remission before HT: 5% recurrence if >5 years in remission, 26% recurrence if 1-5 years in remission and 63% recurrence if <1 year in remission. A myriad of mechanisms influence oncogenesis following HT, including reduced host immunosurveillance from chronic immunosuppression, influence of oncogenic viruses, and the cumulative intensity and duration of immunosuppression. Conversely, protective factors include acyclovir prophylaxis, use of proliferation signal inhibitors (PSI) and female gender. Management involves reducing immunosuppression, incorporating a PSI for immunosuppression and heightened surveillance for allograft rejection. Cancer treatment, including immunotherapy, may be cardiotoxic and lead to graft failure or rejection. Additionally, there exists a competing risk to reduce immunosuppression to improve cancer outcomes, which may increase risk for rejection. A multidisciplinary cardio-oncology team approach is recommended to optimise care and should include an oncologist, transplant cardiologist, transplant pharmacist, palliative care, transplant coordinator and cardio-oncologist.
Collapse
Affiliation(s)
- Parvathi Mudigonda
- Department of Medicine, Division of Cardiology, University of Washington, Seattle, Washington, USA
| | - Cecilia Berardi
- Department of Medicine, Division of Cardiology, University of Washington, Seattle, Washington, USA
| | - Vishaka Chetram
- Department of Medicine, MedStar Washington Hospital Center, Georgetown University, Washington, DC, USA
| | - Ana Barac
- Department of Cardiology, MedStar Heart and Vascular Institute, Georgetown University, Washington, DC, USA
| | - Richard Cheng
- Department of Medicine, Division of Cardiology, University of Washington, Seattle, Washington, USA
| |
Collapse
|
47
|
Patil R, Prashar R, Patel A. Heterogeneous Manifestations of Posttransplant Lymphoma in Renal Transplant Recipients: A Case Series. Transplant Proc 2021; 53:1519-1527. [PMID: 34134932 DOI: 10.1016/j.transproceed.2021.04.020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 04/05/2021] [Accepted: 04/19/2021] [Indexed: 01/07/2023]
Abstract
Posttransplant lymphoproliferative disorder (PTLD) occurs in 1% to 3% of adult renal transplant recipients (RTRs). PTLD has a heterogeneous presentation and is often associated with Epstein-Barr virus (EBV) and immunosuppression. We present a descriptive case series of 16 RTRs who demonstrate a variety of PTLD manifestations. Fifty-six percent received rabbit antithymocyte globulin induction, and 37.5% received basiliximab. Maintenance immunosuppression included glucocorticoids, tacrolimus, and mycophenolate mofetil. Median time from transplantation to PTLD diagnosis was 96.5 months. PTLD involved a single site in 44% of RTRs and multiple sites in 56%. PTLD was localized to the gastrointestinal tract in 9 RTRs, in lymph nodes in 9, central nervous system in 4, bone marrow in 3, skin in 3, lungs in 2, perinephric space in 2, mediastinum in 1, and native kidney in 1. PTLD was EBV positive in 8 RTRs, monomorphic/monoclonal in 14, and of B-cell lineage in 13. Three RTRs had T-cell PTLD. Immunosuppressive agents, except glucocorticoids, were discontinued at diagnosis. Treatment was chemotherapy either alone (in 14 RTRs) or in combination with radiation. Complete remission was achieved in 62.5% of RTRs. Renal dysfunction developed in 62.5% of RTRs, and 4 received dialysis. The overall mortality rate was 62.5%, with median time of death 6.5 months after diagnosis. PTLD that was EBV negative and had T-cell involvement presented with aggressive disease and a higher mortality. Clinicians should be aware of the various PTLD manifestations. Early diagnosis and a multidisciplinary approach to treatment is crucial for improved patient outcomes.
Collapse
Affiliation(s)
- Rujuta Patil
- Wayne State University School of Medicine, Detroit, Michigan
| | - Rohini Prashar
- Kidney and Pancreas Transplant Program, Henry Ford Transplant Institute, Detroit, Michigan
| | - Anita Patel
- Kidney and Pancreas Transplant Program, Henry Ford Transplant Institute, Detroit, Michigan.
| |
Collapse
|
48
|
Kinch A, Baecklund E, Molin D, Pauksens K, Sundström C, Tufveson G, Enblad G. Prior antithymocyte globulin therapy and survival in post-transplant lymphoproliferative disorders. Acta Oncol 2021; 60:771-778. [PMID: 33793378 DOI: 10.1080/0284186x.2021.1904520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Background: Treatment with antithymocyte globulin (ATG) is a well-recognized risk factor for the development of post-transplant lymphoproliferative disorders (PTLD) after solid organ transplantation, but it is unknown how its use affects overall survival after PTLD.Methods: A total of 114 patients with PTLD and available data on immunosuppressive regimen were included from a nation-wide case series of solid organ transplant recipients in Sweden. Prior use of ATG was correlated to clinical features, PTLD subtype, and survival.Results: A total of 47 (41%) patients had received ATG prior to the diagnosis of PTLD. The ATG-treated patients were more likely to be recipients of hearts or lungs, and less likely of kidneys (p < 0.01). They had experienced more acute rejections (p = 0.02). The PTLDs arose earlier, median 2.0 vs. 6.6 years post-transplant (p = 0.002) and were more often situated in the allograft (32% vs. 7%, p < 0.001) in patients with prior ATG vs. no ATG treatment. The PTLDs in the ATG group were more often Epstein-Barr virus-positive (80% vs. 40%, p < 0.001). There were more polymorphic PTLDs (17% vs. 1.5%, p = 0.004) and less T-cell PTLDs (4% vs. 19%, p = 0.02) in the ATG group than in the no ATG group. Diffuse large B-cell lymphoma was equally common in patients with and without prior ATG therapy, but the non-germinal center subtype was more frequent in the ATG group (p = 0.001). In an adjusted Cox proportional hazards regression model, prior ATG treatment and better performance status were associated with superior overall survival, whereas older age, T-cell subtype of PTLD, presence of B symptoms, and elevated lactate dehydrogenase were associated with inferior overall survival. Patients receiving ATG solely as rejection therapy had superior overall survival compared with those receiving ATG as induction therapy or both (p = 0.03).Conclusions: ATG therapy, especially rejection therapy, prior to PTLD development is an independent prognostic factor for superior overall survival after PTLD diagnosis.
Collapse
Affiliation(s)
- Amelie Kinch
- Department of Medical Sciences, Section of Infectious Diseases, Uppsala University, Uppsala, Sweden
| | - Eva Baecklund
- Department of Medical Sciences, Section of Rheumatology, Uppsala University, Uppsala, Sweden
| | - Daniel Molin
- Experimental and Clinical Oncology, Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Karlis Pauksens
- Department of Medical Sciences, Section of Infectious Diseases, Uppsala University, Uppsala, Sweden
| | - Christer Sundström
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Gunnar Tufveson
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Gunilla Enblad
- Experimental and Clinical Oncology, Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
49
|
McDonald L, O' Doherty R, Ryan E, Enright H, Dunlea E, Kelliher S, Fortune A, Fay M, Maung SW, Desmond R, Wall C, Kumar S, O' Shea D, Fadalla K, Connaghan DG, Smyth L. Posttransplant Lymphoproliferative Disorder After Solid Organ Transplant: A Heterogeneous, Aggressive Disorder. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2021; 21:694-700. [PMID: 34148849 DOI: 10.1016/j.clml.2021.05.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 05/07/2021] [Accepted: 05/16/2021] [Indexed: 11/17/2022]
Abstract
Posttransplant lymphoproliferative disorder (PTLD) is a rare complication of solid organ transplant. We identified 40 patients diagnosed with PTLD between 2009 and 2020 and analyzed their presentation, treatment strategies, and outcomes. Median age at diagnosis was 52.5 years (range 21.3 to 79). Median duration of immunosuppression was 95 months (range 4 to 292). Diffuse large B cell lymphoma (n = 16, 40%) and Burkitt lymphoma (n = 6, 15%) were the most common histological subtypes. First-line therapy varied. The median number of treatment lines was 1 (range 0 to 4). Sixteen patients (40%) achieved complete response after first-line therapy. Nineteen patients (47.5%) relapsed or progressed and received salvage therapy; 45% were alive at the end of the study period (median survival 52 months; range 1 to 266; 95% confidence interval 0 to 104). Causes of death included lymphoma-related (45.5%), therapy-related (27.3%), and other (27.3%). Five (22.7%) died within 3 months of diagnosis. Pearson's r test identified disease stage (P = .045) and proliferation index (P = .005) as negative predictors of response to frontline therapy. Bone marrow involvement (P = .033) and increased age (P = .018) were significant predictors of survival. Early mortality and poor response to frontline therapy are common, outlining the need for improved treatment strategies.
Collapse
Affiliation(s)
- Laura McDonald
- Department of Haematology, St Vincent's University Hospital, Elm Park, Dublin, Ireland.
| | - Roseann O' Doherty
- Department of Haematology, Mater Misericordiae University Hospital, Eccles St, Dublin, Ireland
| | - Eileen Ryan
- Department of Haematology, Cork University Hospital, Cork, Ireland
| | - Helen Enright
- Department of Haematology, Tallaght University Hospital, Dublin, Ireland
| | - Eoghan Dunlea
- Department of Haematology, Mater Misericordiae University Hospital, Eccles St, Dublin, Ireland
| | - Sarah Kelliher
- Department of Haematology, Mater Misericordiae University Hospital, Eccles St, Dublin, Ireland
| | - Anne Fortune
- Department of Haematology, Mater Misericordiae University Hospital, Eccles St, Dublin, Ireland
| | - Michael Fay
- Department of Haematology, Mater Misericordiae University Hospital, Eccles St, Dublin, Ireland
| | - S W Maung
- Department of Haematology, Mater Misericordiae University Hospital, Eccles St, Dublin, Ireland
| | - Ronan Desmond
- Department of Haematology, Tallaght University Hospital, Dublin, Ireland
| | - Catherine Wall
- Department of Haematology, Tallaght University Hospital, Dublin, Ireland
| | - Senthil Kumar
- Department of Haematology, University Hospital Waterford, Waterford, Ireland
| | - Derville O' Shea
- Department of Haematology, Cork University Hospital, Cork, Ireland
| | - Kamal Fadalla
- Department of Haematology, St Vincent's University Hospital, Elm Park, Dublin, Ireland
| | - D G Connaghan
- Department of Haematology, St Vincent's University Hospital, Elm Park, Dublin, Ireland
| | - Liam Smyth
- Department of Haematology, St Vincent's University Hospital, Elm Park, Dublin, Ireland
| |
Collapse
|
50
|
Abstract
OBJECTIVE We report a case of facial paresis and profound hearing loss from post-transplant lymphoproliferative disorder (PTLD) in a pediatric patient with neuroblastoma. PATIENT Three-year-old boy with rapidly progressive right facial paresis and sensorineural hearing loss. High-risk neuroblastoma had been diagnosed 1 year earlier, treated with chemotherapy and resection of the adrenal primary tumor. Two months after two autologous hematopoietic stem cell transplantations (HSCT), the patient developed facial paralysis. Magnetic resonance imaging (MRI) showed bilateral progressive internal auditory canal (IAC) enhancing lesions with a mass lesion on the right and wispy enhancement on the left and enhancement within the right cochlea. Lumbar puncture (LP) was positive for Epstein-Barr virus (EBV) making the diagnosis of PTLD most probable. Biopsy of the right IAC lesion was deferred because of potential procedural risks including intradural spread of tumor or fungus. The patient was treated with anti-fungal therapy and systemic rituximab without improvement. Subsequent intrathecal rituximab resulted in improvement of lesions on MRI and clearance of EBV from the cerebrospinal fluid (CSF). INTERVENTIONS Mastoidectomy for biopsies from the mastoid and middle ear. Intrathecal treatment with rituximab. MAIN OUTCOME MEASURES Imaging assessment of IAC lesion, CSF EBV titers, facial nerve function. RESULTS Gradual resolution of IAC mass lesions, remission of PTLD, and facial improvement from House-Brackmann score of 4 to 3. CONCLUSIONS PTLD causing facial paresis after autologous HSCT has not been previously reported and may be considered in the differential diagnosis of lesions causing facial paresis in patients who have received a stem cell or solid organ transplant.
Collapse
|