1
|
Kikkawa K, Tamaki M, Maruno K, Hazama T, Takahashi T, Yamada Y, Nakashima M, Ito N. Kidney transplantation after ABO-incompatible hematopoietic stem cell transplantation: A case report and literature review. Transpl Immunol 2024; 87:102143. [PMID: 39491597 DOI: 10.1016/j.trim.2024.102143] [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: 10/04/2024] [Revised: 10/30/2024] [Accepted: 11/01/2024] [Indexed: 11/05/2024]
Abstract
Although some studies have reported kidney transplantation for end-stage kidney disease after hematopoietic stem cell transplantation, few have reported kidney transplantation after ABO-incompatible hematopoietic stem cell transplantation. In this report, we describe a case of kidney transplantation after major ABO-incompatible HSCT and reviewed previous reports of kidney transplantation after hematopoietic stem cell transplantation. A 21-year-old male patient received major ABO-incompatible hematopoietic stem cell transplantation from an unrelated donor for B-lymphoblastic lymphoma. He developed chronic kidney disease requiring kidney replacement therapy because of drug toxicity and underwent ABO-compatible living donor kidney transplantation from his mother with standard immunosuppression. He had no anti-donor blood type antibody before kidney transplantation. Ten months after kidney transplantation, he is in good clinical condition with good renal function. Eighty-four cases of kidney transplantation after hematopoietic stem cell transplantation have been reported in literature. Among them, 25 recipients were from the same donor as hematopoietic stem cell transplantation. Out of these 25 recipients, 15 did not undergo maintenance immunosuppressive therapy. The low rejection incidence (14 %) and high rate of infection (32 %) and malignancy (10 %) suggest that kidney transplant recipients after hematopoietic stem cell transplantation are over-immunosuppressed. There were only three reports of kidney transplantation after ABO-incompatible hematopoietic stem cell transplantation, including the present case. Kidney transplantation may be an effective renal replacement therapy for end-stage kidney disease after hematopoietic stem cell transplantation, even in ABO-incompatible hematopoietic stem cell transplantation cases.
Collapse
Affiliation(s)
- Kazuro Kikkawa
- Department of Urology, Japanese Red Cross Wakayama Medical Center, 4-20 Komatsubaradori, Wakayama 640-8558, Japan.
| | - Masahiro Tamaki
- Department of Urology, Japanese Red Cross Wakayama Medical Center, 4-20 Komatsubaradori, Wakayama 640-8558, Japan
| | - Kouhei Maruno
- Department of Urology, Japanese Red Cross Wakayama Medical Center, 4-20 Komatsubaradori, Wakayama 640-8558, Japan
| | - Tatsuya Hazama
- Department of Urology, Japanese Red Cross Wakayama Medical Center, 4-20 Komatsubaradori, Wakayama 640-8558, Japan
| | - Toshifumi Takahashi
- Department of Urology, Japanese Red Cross Wakayama Medical Center, 4-20 Komatsubaradori, Wakayama 640-8558, Japan
| | - Yuya Yamada
- Department of Urology, Japanese Red Cross Wakayama Medical Center, 4-20 Komatsubaradori, Wakayama 640-8558, Japan
| | - Masakazu Nakashima
- Department of Urology, Japanese Red Cross Wakayama Medical Center, 4-20 Komatsubaradori, Wakayama 640-8558, Japan
| | - Noriyuki Ito
- Department of Urology, Japanese Red Cross Wakayama Medical Center, 4-20 Komatsubaradori, Wakayama 640-8558, Japan
| |
Collapse
|
2
|
Rotz SJ, Bhatt NS, Hamilton BK, Duncan C, Aljurf M, Atsuta Y, Beebe K, Buchbinder D, Burkhard P, Carpenter PA, Chaudhri N, Elemary M, Elsawy M, Guilcher GMT, Hamad N, Karduss A, Peric Z, Purtill D, Rizzo D, Rodrigues M, Ostriz MBR, Salooja N, Schoemans H, Seber A, Sharma A, Srivastava A, Stewart SK, Baker KS, Majhail NS, Phelan R. International recommendations for screening and preventative practices for long-term survivors of transplantation and cellular therapy: a 2023 update. Bone Marrow Transplant 2024; 59:717-741. [PMID: 38413823 DOI: 10.1038/s41409-023-02190-2] [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: 12/05/2023] [Revised: 12/08/2023] [Accepted: 12/19/2023] [Indexed: 02/29/2024]
Abstract
As hematopoietic cell transplantation (HCT) and cellular therapy expand to new indications and international access improves, the volume of HCT performed annually continues to rise. Parallel improvements in HCT techniques and supportive care entails more patients surviving long-term, creating further emphasis on survivorship needs. Survivors are at risk for developing late complications secondary to pre-, peri- and post-transplant exposures and other underlying risk-factors. Guidelines for screening and preventive practices for HCT survivors were originally published in 2006 and updated in 2012. To review contemporary literature and update the recommendations while considering the changing practice of HCT and cellular therapy, an international group of experts was again convened. This review provides updated pediatric and adult survivorship guidelines for HCT and cellular therapy. The contributory role of chronic graft-versus-host disease (cGVHD) to the development of late effects is discussed but cGVHD management is not covered in detail. These guidelines emphasize special needs of patients with distinct underlying HCT indications or comorbidities (e.g., hemoglobinopathies, older adults) but do not replace more detailed group, disease, or condition specific guidelines. Although these recommendations should be applicable to the vast majority of HCT recipients, resource constraints may limit their implementation in some settings.
Collapse
Affiliation(s)
- Seth J Rotz
- Division of Pediatric Hematology, Oncology, and Blood and Marrow Transplantation, Pediatric Institute, Cleveland Clinic Foundation, Cleveland, OH, USA.
- Blood and Marrow Transplant Program, Department of Hematology and Oncology, Taussig Cancer Institute, Cleveland Clinic Foundation, Cleveland, OH, USA.
| | | | - Betty K Hamilton
- Blood and Marrow Transplant Program, Department of Hematology and Oncology, Taussig Cancer Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Christine Duncan
- Dana Farber/Boston Children's Cancer and Blood Disorders Center, Harvard University, Boston, MA, USA
| | - Mahmoud Aljurf
- King Faisal Specialist Hospital & Research Center, Riyadh, Saudi Arabia
| | - Yoshiko Atsuta
- Department of Registry Science for Transplant and Cellular Therapy, Aichi Medical University School of Medicine, Nagakute, Japan
- Japanese Data Center for Hematopoietic Cell Transplantation, Nagakute, Japan
| | - Kristen Beebe
- Phoenix Children's Hospital and Mayo Clinic Arizona, Phoenix, AZ, USA
| | - David Buchbinder
- Division of Hematology, Children's Hospital of Orange County, Orange, CA, USA
| | - Peggy Burkhard
- National Bone Marrow Transplant Link, Southfield, MI, USA
| | | | - Naeem Chaudhri
- King Faisal Specialist Hospital & Research Center, Riyadh, Saudi Arabia
| | - Mohamed Elemary
- Hematology and BMT, University of Saskatchewan, Saskatoon, SK, Canada
| | - Mahmoud Elsawy
- Division of Hematology, Dalhousie University, Halifax, NS, Canada
- QEII Health Sciences Center, Halifax, NS, Canada
| | - Gregory M T Guilcher
- Section of Pediatric Oncology/Transplant and Cellular Therapy, Alberta Children's Hospital, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Nada Hamad
- Department of Haematology, St Vincent's Hospital Sydney, Sydney, NSW, Australia
- St Vincent's Clinical School Sydney, University of New South Wales, Sydney, NSW, Australia
- School of Medicine Sydney, University of Notre Dame Australia, Sydney, WA, Australia
| | - Amado Karduss
- Bone Marrow Transplant Program, Clinica las Americas, Medellin, Colombia
| | - Zinaida Peric
- BMT Unit, Department of Hematology, University Hospital Centre Zagreb and School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Duncan Purtill
- Fiona Stanley Hospital, Murdoch, WA, Australia
- PathWest Laboratory Medicine, Nedlands, WA, Australia
| | - Douglas Rizzo
- Medical College of Wisconsin, Milwaukee, WI, USA
- Center for International Blood and Marrow Transplant Research, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | | | - Maria Belén Rosales Ostriz
- Division of hematology and bone marrow transplantation, Instituto de trasplante y alta complejidad (ITAC), Buenos Aires, Argentina
| | - Nina Salooja
- Centre for Haematology, Imperial College London, London, UK
| | - Helene Schoemans
- Department of Hematology, University Hospitals Leuven, Leuven, Belgium
- Department of Public Health and Primary Care, ACCENT VV, KU Leuven-University of Leuven, Leuven, Belgium
| | | | - Akshay Sharma
- Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Alok Srivastava
- Department of Haematology, Christian Medical College, Vellore, India
| | - Susan K Stewart
- Blood & Marrow Transplant Information Network, Highland Park, IL, 60035, USA
| | | | - Navneet S Majhail
- Sarah Cannon Transplant and Cellular Therapy Network, Nashville, TN, USA
| | - Rachel Phelan
- Center for International Blood and Marrow Transplant Research, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
- Division of Pediatric Hematology/Oncology/Blood and Marrow Transplant, Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
3
|
Winkler J, Tittlbach H, Schneider A, Vasova I, Strobel J, Herold S, Maas S, Spriewald BM, Repp R, Kordelas L, Mach M, Wolff D, Edinger M, Mackensen A, Winkler TH. Adoptive transfer of donor B lymphocytes: a phase 1/2a study for patients after allogeneic stem cell transplantation. Blood Adv 2024; 8:2373-2383. [PMID: 38467031 PMCID: PMC11127194 DOI: 10.1182/bloodadvances.2023012305] [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: 12/11/2023] [Revised: 02/20/2024] [Accepted: 03/03/2024] [Indexed: 03/13/2024] Open
Abstract
ABSTRACT Immune reconstitution after allogeneic hematopoietic stem cell transplantation (allo-HSCT) is slow and patients carry a high and prolonged risk of opportunistic infections. We hypothesized that the adoptive transfer of donor B cells can foster after HSCT immuno-reconstitution. Here, we report, to our knowledge, the results of a first-in-human phase 1/2a study aimed to evaluate the feasibility and safety of adoptively transferred donor B cells and to test their activity upon recall vaccination. Good manufactoring practice (GMP) B-cell products were generated from donor apheresis products using 2-step magnetic cell separation. Fifteen patients who had undergone allo-HSCT were enrolled and treated after taper of immunosuppression (median, day +148; range, 130-160). Patients received 4 different doses of B cells (0.5 × 106 to 4.0 × 106 B cells per kg body weight). To test the activity of infused donor memory B cells in vivo, patients were vaccinated with a pentavalent vaccine 7 days after B-cell transfer. We observed the mobilization of plasmablasts and an increase in serum titers against vaccine antigens, with a stronger response in patients receiving higher B-cell numbers. Analysis of immunoglobulin VH-sequences by next-generation sequencing revealed that plasmablasts responding to vaccination originated from memory B-cell clones from the donor. Donor B-cell transfer was safe, as no Epstein-Barr virus (EBV) reactivation was observed, and only low-grade graft-versus-host disease (GVHD) occurred in 4 out of 15 patients. This pilot trial may pave the way for further studies exploring the adoptive transfer of memory B cells to reduce the frequency of infections after allo-HSCT. This trial was registered at ClinicalTrial.gov as #NCT02007811.
Collapse
Affiliation(s)
- Julia Winkler
- Department of Internal Medicine 5 – Haematology and Oncology, University Hospital Erlangen, Erlangen, Germany
| | - Hannes Tittlbach
- Department of Internal Medicine 5 – Haematology and Oncology, University Hospital Erlangen, Erlangen, Germany
- Division of Genetics, Department for Biology, Nikolaus-Fiebiger-Center for Molecular Medicine, Friedrich-Alexander-University Erlangen-Nuremberg (FAU), Erlangen, Germany
| | - Andrea Schneider
- Division of Genetics, Department for Biology, Nikolaus-Fiebiger-Center for Molecular Medicine, Friedrich-Alexander-University Erlangen-Nuremberg (FAU), Erlangen, Germany
| | - Ingrid Vasova
- Department of Internal Medicine 5 – Haematology and Oncology, University Hospital Erlangen, Erlangen, Germany
| | - Julian Strobel
- Department of Transfusion Medicine and Haemostaseology, University Hospital Erlangen, Erlangen, Germany
| | - Susanne Herold
- Center for Clinical Studies Erlangen, University Hospital Erlangen, Erlangen, Germany
| | - Stefanie Maas
- Center for Clinical Studies Erlangen, University Hospital Erlangen, Erlangen, Germany
| | - Bernd M. Spriewald
- Department of Internal Medicine 5 – Haematology and Oncology, University Hospital Erlangen, Erlangen, Germany
| | - Roland Repp
- Medical Department 2, City Hospital Kiel, Kiel, Germany
| | - Lambros Kordelas
- Department of Bone Marrow Transplantation, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- DRK-Blutspendedienst West, Ratingen, Germany
| | - Michael Mach
- Institute for Clinical and Molecular Virology, University Hospital Erlangen, Erlangen, Germany
| | - Daniel Wolff
- Department of Hematology and Medical Oncology, University Hospital Regensburg, Regenburg, Germany
| | - Matthias Edinger
- Department of Hematology and Medical Oncology, University Hospital Regensburg, Regenburg, Germany
- Leibniz Institute for Immunotherapy, Regensburg, Germany
| | - Andreas Mackensen
- Department of Internal Medicine 5 – Haematology and Oncology, University Hospital Erlangen, Erlangen, Germany
| | - Thomas H. Winkler
- Division of Genetics, Department for Biology, Nikolaus-Fiebiger-Center for Molecular Medicine, Friedrich-Alexander-University Erlangen-Nuremberg (FAU), Erlangen, Germany
| |
Collapse
|
4
|
Rotz SJ, Bhatt NS, Hamilton BK, Duncan C, Aljurf M, Atsuta Y, Beebe K, Buchbinder D, Burkhard P, Carpenter PA, Chaudhri N, Elemary M, Elsawy M, Guilcher GM, Hamad N, Karduss A, Peric Z, Purtill D, Rizzo D, Rodrigues M, Ostriz MBR, Salooja N, Schoemans H, Seber A, Sharma A, Srivastava A, Stewart SK, Baker KS, Majhail NS, Phelan R. International Recommendations for Screening and Preventative Practices for Long-Term Survivors of Transplantation and Cellular Therapy: A 2023 Update. Transplant Cell Ther 2024; 30:349-385. [PMID: 38413247 PMCID: PMC11181337 DOI: 10.1016/j.jtct.2023.12.001] [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: 12/04/2023] [Accepted: 12/04/2023] [Indexed: 02/29/2024]
Abstract
As hematopoietic cell transplantation (HCT) and cellular therapy expand to new indications and international access improves, the number of HCTs performed annually continues to rise. Parallel improvements in HCT techniques and supportive care entails more patients surviving long term, creating further emphasis on survivorship needs. Survivors are at risk for developing late complications secondary to pretransplantation, peritransplantation, and post-transplantation exposures and other underlying risk factors. Guidelines for screening and preventive practices for HCT survivors were originally published in 2006 and then updated in 2012. An international group of experts was convened to review the contemporary literature and update the recommendations while considering the changing practices of HCT and cellular therapy. This review provides updated pediatric and adult survivorship guidelines for HCT and cellular therapy. The contributory role of chronic graft-versus-host disease (cGVHD) to the development of late effects is discussed, but cGVHD management is not covered in detail. These guidelines emphasize the special needs of patients with distinct underlying HCT indications or comorbidities (eg, hemoglobinopathies, older adults) but do not replace more detailed group-, disease-, or condition-specific guidelines. Although these recommendations should be applicable to the vast majority of HCT recipients, resource constraints may limit their implementation in some settings.
Collapse
Affiliation(s)
- Seth J Rotz
- Department of Pediatric Hematology, Oncology, and Blood and Marrow Transplantation, Pediatric Institute, Cleveland Clinic Foundation, Cleveland, Ohio; Blood and Marrow Transplant Program, Department of Hematology and Oncology, Taussig Cancer Institute, Cleveland Clinic Foundation, Cleveland, Ohio.
| | - Neel S Bhatt
- Fred Hutchinson Cancer Center, Seattle, Washington
| | - Betty K Hamilton
- Blood and Marrow Transplant Program, Department of Hematology and Oncology, Taussig Cancer Institute, Cleveland Clinic Foundation, Cleveland, Ohio
| | - Christine Duncan
- Dana Farber/Boston Children's Cancer and Blood Disorders Center, Harvard University, Boston, Massachusetts
| | - Mahmoud Aljurf
- King Faisal Specialist Hospital & Research Center, Riyadh, Saudi Arabia
| | - Yoshiko Atsuta
- Department of Registry Science for Transplant and Cellular Therapy, Aichi Medical University School of Medicine, Japanese Data Center for Hematopoietic Cell Transplantation, Nagakute, Japan
| | - Kristen Beebe
- Phoenix Children's Hospital and Mayo Clinic Arizona, Phoenix, Arizona
| | - David Buchbinder
- Division of Hematology, Children's Hospital of Orange County, Orange, California
| | | | | | - Naeem Chaudhri
- King Faisal Specialist Hospital & Research Center, Riyadh, Saudi Arabia
| | - Mohamed Elemary
- Hematology and BMT, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Mahmoud Elsawy
- Division of Hematology, Dalhousie University, QEII Health Sciences Center, Halifax, Nova Scotia, Canada
| | - Gregory Mt Guilcher
- Section of Pediatric Oncology/Transplant and Cellular Therapy, Alberta Children's Hospital, Cumming School of Medicine, University of Calgary, Calgary, Canada
| | - Nada Hamad
- Department of Haematology, St Vincent's Hospital Sydney, St Vincent's Clinical School Sydney, University of New South Wales, School of Medicine Sydney, University of Notre Dame Australia, Australia
| | - Amado Karduss
- Bone Marrow Transplant Program, Clinica las Americas, Medellin, Colombia
| | - Zinaida Peric
- BMT Unit, Department of Hematology, University Hospital Centre Zagreb and School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Duncan Purtill
- Fiona Stanley Hospital, Murdoch, PathWest Laboratory Medicine WA, Australia
| | - Douglas Rizzo
- Medical College of Wisconsin, Milwaukee, Wisconsin; Center for International Blood and Marrow Transplant Research, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
| | | | - Maria Belén Rosales Ostriz
- Division of hematology and bone marrow transplantation, Instituto de trasplante y alta complejidad (ITAC), Buenos Aires, Argentina
| | - Nina Salooja
- Centre for Haematology, Imperial College London, London, United Kingdom
| | - Helene Schoemans
- Department of Hematology, University Hospitals Leuven, Department of Public Health and Primary Care, ACCENT VV, KU Leuven, University of Leuven, Leuven, Belgium
| | | | - Akshay Sharma
- Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Alok Srivastava
- Department of Haematology, Christian Medical College, Vellore, India
| | | | | | - Navneet S Majhail
- Sarah Cannon Transplant and Cellular Therapy Network, Nashville, Tennessee
| | - Rachel Phelan
- Center for International Blood and Marrow Transplant Research, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin; Division of Pediatric Hematology/Oncology/Blood and Marrow Transplant, Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wisconsin
| |
Collapse
|
5
|
Lee CJ, Wang T, Chen K, Arora M, Brazauskas R, Spellman SR, Kitko C, MacMillan ML, Pidala JA, Badawy SM, Bhatt N, Bhatt VR, DeFilipp Z, Diaz MA, Farhadfar N, Gadalla S, Hashmi S, Hematti P, Hossain NM, Inamoto Y, Lekakis LJ, Sharma A, Solomon S, Lee SJ, Couriel DR. Severity of Chronic Graft-versus-Host Disease and Late Effects Following Allogeneic Hematopoietic Cell Transplantation for Adults with Hematologic Malignancy. Transplant Cell Ther 2024; 30:97.e1-97.e14. [PMID: 37844687 PMCID: PMC10842798 DOI: 10.1016/j.jtct.2023.10.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Revised: 09/06/2023] [Accepted: 10/09/2023] [Indexed: 10/18/2023]
Abstract
The study aimed to determine the association of chronic graft-versus-host disease (cGVHD) diagnosis and severity with the development of subsequent neoplasms (SN) and nonmalignant late effects (NM-LE) in 2-year disease-free adult survivors following hematopoietic cell transplantation (HCT) for a hematologic malignancy. To do so, we conducted a retrospective analysis of 3884 survivors of HCT for hematologic malignancy in the Center of International Blood and Marrow Transplant Research database. We conducted a landmark analysis at the 2-year post-transplantation date, comparing first SN and NM-LE in survivors with and without cGVHD. The cumulative incidence (CuI) of SN and NM-LE were estimated through 10 years post-HCT in both groups, with death or disease relapse as a competing risk. Cox proportional hazards models were used to evaluate the associations of cGVHD and its related characteristics with the development of SN and NM-LE. The estimated 10-year CuI of SN in patients with GVHD (n = 2669) and patients without cGVHD (n = 1215) was 15% (95% confidence interval [CI], 14% to 17%) versus 9% (7.2% to 11%) (P < .001). cGVHD by 2 years post-HCT was independently associated with SN (hazard ratio [HR], 1.94; 95% CI, 1.53 to 2.46; P < .0001) with a standardized incidence ratio of 3.2 (95% CI, 2.9 to 3.5; P < .0001). Increasing severity of cGVHD was associated with an increased risk of SN. The estimated 10-year CuI of first NM-LE in patients with and without cGVHD was 28 (95% CI, 26% to 30%) versus 13% (95% CI, 11% to 15%) (P < .001). cGVHD by 2 years post-HCT was independently associated with NM-LE (HR, 2.23; 95% CI, 1.81 to 2.76; P < .0001). Multivariate analysis of cGVHD-related factors showed that increasing severity of cGVHD, extensive grade, having both mucocutaneous and visceral involvement, and receiving cGVHD treatment for >12 months were associated with the greatest magnitude of risk for NM-LE. cGVHD was closely associated with both SN and NM-LE in adult survivors of HCT for hematologic malignancy. Patients identified as having more severe involvement and both mucocutaneous and visceral organ involvement may warrant enhanced monitoring and screening for SNs and NM-LEs. However, caution is warranted when interpreting these results, as patients with cGVHD may have more vigilant post-transplantation health care and surveillance for late effects.
Collapse
Affiliation(s)
- Catherine J Lee
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington.
| | - Tao Wang
- Division of Biostatistics, Institute for Heath and Equity, Medical College of Wisconsin, Milwaukee, Wisconsin; Center for International Blood and Marrow Transplant Research, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Karen Chen
- Division of Biostatistics, Institute for Heath and Equity, Medical College of Wisconsin, Milwaukee, Wisconsin; Center for International Blood and Marrow Transplant Research, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Mukta Arora
- Division of Hematology, Oncology and Transplant, University of Minnesota Medical Center, Minneapolis, Minnesota
| | - Ruta Brazauskas
- Division of Biostatistics, Institute for Heath and Equity, Medical College of Wisconsin, Milwaukee, Wisconsin; Center for International Blood and Marrow Transplant Research, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Stephen R Spellman
- Center for International Blood and Marrow Transplant Research, National Marrow Donor Program, Minneapolis, Minnesota
| | - Carrie Kitko
- Department of Pediatrics, Division of Hematology and Oncology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Margaret L MacMillan
- Blood and Marrow Transplant Program, University of Minnesota, Minneapolis, Minnesota; Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota
| | - Joseph A Pidala
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Sherif M Badawy
- Hematology, Oncology and Stem Cell Transplantation, Ann & Robert H. Lurie Children's Hospital of Chicago, Northwestern University, Evanston, Illinois
| | - Neel Bhatt
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Vijaya R Bhatt
- Section of Hematology, University of Nebraska, Omaha, Nebraska
| | - Zachariah DeFilipp
- Blood and Marrow Transplant Program, Massachusetts General Hospital, Boston, Massachusetts
| | - Miguel A Diaz
- Department of Pediatrics, Hospital Nino Jesus, Madrid, Spain
| | - Nosha Farhadfar
- Division of Hematology/Oncology, University of Florida College of Medicine, Gainesville, Florida
| | - Shahinaz Gadalla
- Clinical Genetics Branch, National Cancer Institute, Rockville, Maryland
| | - Shahrukh Hashmi
- Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota; Department of Medicine, Sheikh Shakhbout Medical City, Abu Dhabi, United Arab Emirates
| | - Peiman Hematti
- Section of Hematology/Oncology, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin
| | - Nasheed M Hossain
- Division of Hematology-Oncology, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvnaia
| | - Yoshihiro Inamoto
- Division of Hematopoietic Stem Cell Transplantation, National Cancer Center Hospital, Tokyo, Japan
| | | | - Akshay Sharma
- Department of Bone Marrow Transplantation and Cellular Therapy, St Jude Children's Research Hospital, Memphis, Tennessee
| | - Scott Solomon
- Northside Hospital Cancer Institute, Atlanta, Georgia
| | - Stephanie J Lee
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Daniel R Couriel
- Utah Transplant and Cellular Therapy Program, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah
| |
Collapse
|
6
|
van Yperen NC, Wauben B, van der Poel MW, Köhler S, van Greevenbroek MM, Schouten HC. Selection bias in follow-up studies of stem cell transplantation survivors: an experience within the Maastricht Observational study of late effects after Stem cell trAnsplantation (MOSA). Ann Hematol 2023; 102:641-649. [PMID: 36585483 DOI: 10.1007/s00277-022-05070-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 12/01/2022] [Indexed: 01/01/2023]
Abstract
Hematopoietic stem cell transplantation is an important treatment for many malignant hematological and non-hematological diseases. Survivors of hematopoietic stem cell transplantation (HCT) are at risk of long-term health problems and reduced quality of life related to previous treatments. Many studies about these long-term effects have been conducted over the last decades. However, selection bias is a concern in long-term follow-up studies and little is known about the non-participating group. As part of the Maastricht Observational study of late effects after Stem cell trAnsplantation (MOSA), investigating long-term health effects by extensively phenotyping HCT survivors, we conducted a survey to characterise the non-participating group. This survey mostly focused on quality of life and physical complaints. The survey responders were generally older than the MOSA group, had more history of relapsed disease, and described their general health as bad or mediocre significantly more often than the MOSA group. Also, more deaths occurred in the group of non-participants between the start of study inclusion in 2015 and analysis of the survey results in 2021. This study suggests that a selection of higher functioning HCT survivors with a relatively better quality of life participated in this long-term follow-up study of stem cell transplantation survivors. These results could also impact the results of other long-term follow-up studies in cancer survivors, knowing that possibly an unhealthier population is missed in these studies and some long-term negative effects of treatments might be underestimated.Trial registration number: NL-48599.
Collapse
Affiliation(s)
- Nicole C van Yperen
- Maastricht University Medical Center+, P. Debyelaan 25, 6229 HX, Maastricht, The Netherlands.
| | - Bianca Wauben
- Maastricht University Medical Center+, P. Debyelaan 25, 6229 HX, Maastricht, The Netherlands
| | | | - Sebastian Köhler
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Maastricht University, Universiteitssingel 40, 6229 ER, Maastricht, The Netherlands.,Alzheimer Center Limburg, Maastricht University Medical Center+, Dr. Tanslaan 12, 6229 ET, Maastricht, The Netherlands
| | - Marleen Mj van Greevenbroek
- Department of Internal Medicine, Maastricht University Medical Center+, Maastricht, The Netherlands.,School for Cardiovascular Diseases (CARIM), Maastricht University, Universiteitssingel 50, 6229 ER, Maastricht, The Netherlands
| | - Harry C Schouten
- Maastricht University Medical Center+, P. Debyelaan 25, 6229 HX, Maastricht, The Netherlands
| |
Collapse
|
7
|
Non- Aspergillus Hyaline Molds: A Host-Based Perspective of Emerging Pathogenic Fungi Causing Sinopulmonary Diseases. J Fungi (Basel) 2023; 9:jof9020212. [PMID: 36836326 PMCID: PMC9964096 DOI: 10.3390/jof9020212] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 01/31/2023] [Accepted: 02/01/2023] [Indexed: 02/09/2023] Open
Abstract
The incidence of invasive sino-pulmonary diseases due to non-Aspergillus hyaline molds is increasing due to an enlarging and evolving population of immunosuppressed hosts as well as improvements in the capabilities of molecular-based diagnostics. Herein, we review the following opportunistic pathogens known to cause sinopulmonary disease, the most common manifestation of hyalohyphomycosis: Fusarium spp., Scedosporium spp., Lomentospora prolificans, Scopulariopsis spp., Trichoderma spp., Acremonium spp., Paecilomyces variotii, Purpureocillium lilacinum, Rasamsonia argillacea species complex, Arthrographis kalrae, and Penicillium species. To facilitate an understanding of the epidemiology and clinical features of sino-pulmonary hyalohyphomycoses in the context of host immune impairment, we utilized a host-based approach encompassing the following underlying conditions: neutropenia, hematologic malignancy, hematopoietic and solid organ transplantation, chronic granulomatous disease, acquired immunodeficiency syndrome, cystic fibrosis, and healthy individuals who sustain burns, trauma, or iatrogenic exposures. We further summarize the pre-clinical and clinical data informing antifungal management for each pathogen and consider the role of adjunctive surgery and/or immunomodulatory treatments to optimize patient outcome.
Collapse
|
8
|
Linking the Center for International Blood and Marrow Transplant Research Registry to the California Cancer Registry and California Hospital Patient Discharge Data. Transplant Cell Ther 2022; 28:859.e1-859.e10. [PMID: 36174935 DOI: 10.1016/j.jtct.2022.09.016] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 09/02/2022] [Accepted: 09/22/2022] [Indexed: 12/24/2022]
Abstract
Advances in hematopoietic cell transplantation (HCT) have substantially improved patient survival, increasing the importance of studying outcomes and long-term adverse effects in the rapidly growing population of HCT survivors. Large-scale registry data from the Center for International Blood and Marrow Transplant Research (CIBMTR) are a valuable resource for studying mortality and late effects after HCT, providing detailed data reported by HCT centers on transplantation-related factors and key outcomes. This study was conducted to evaluate the robustness of CIBMTR outcome data and assess health-related outcomes and healthcare utilization among HCT recipients. We linked data from the CIBMTR for California residents with data from the population-based California Cancer Registry (CCR) and hospitalization information from the California Patient Discharge Database (PDD). In this retrospective cohort study, probabilistic and deterministic record linkage used key patient identifiers, such as Social Security number, ZIP code, sex, birth date, hematologic malignancy type and diagnosis date, and HCT type and date. Among 22,733 patients registered with the CIBMTR who underwent autologous or allogeneic HCT for hematologic malignancy between 1991 and 2016, 89.0% were matched to the CCR and/or PDD (n = 17,707 [77.9%] for both, n = 1179 [5.2%] for the CCR only, and n = 1342 [5.9%] for the PDD only). Unmatched patients were slightly more likely to have undergone a first autologous HCT than an allogeneic HCT (12.6% versus 9.0%), to have a larger number of missing linkage identifiers, and to have undergone HCT prior to 2010. Among the patients reported to the CIBMTR who matched to the CCR, 85.7% demonstrated concordance of both hematologic malignancy type and diagnosis date across data sources. This linkage presents unparalleled opportunities to advance our understanding of HCT practices and patient outcomes.
Collapse
|
9
|
Gordon O, Terpilowski M, Dulman R, Keller MD, Burbelo PD, Cohen JI, Bollard CM, Dave H. Robust immune responses to SARS-CoV-2 in a pediatric patient with B-Cell ALL receiving tisagenlecleucel. Pediatr Hematol Oncol 2022; 39:571-579. [PMID: 35135442 PMCID: PMC11524425 DOI: 10.1080/08880018.2022.2035864] [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: 08/05/2021] [Revised: 12/07/2021] [Accepted: 01/19/2022] [Indexed: 10/19/2022]
Abstract
Recipients of anti-CD19 targeted therapies such as chimeric antigen receptor (CAR)-T cell are considered at high risk for complicated Severe Acute Respiratory Syndrome Coronavirus (SARS-CoV-2) infection due to prolonged B cell aplasia and immunosuppression. These patients represent a unique cohort and so far, immune responses to SARS-CoV-2 have not been well characterized in this setting. We report a pediatric patient with B-cell acute lymphoblastic leukemia (B-ALL) who had asymptomatic SARS-CoV-2 infection while receiving blinatumomab, followed by lymphodepletion (LD) and tisagenlecleucel, a CD19 targeting CAR-T therapy. The patient had a complete response to tisagenlecleucel, did not develop cytokine release syndrome, or worsening of SARS-CoV-2 during therapy. The patient had evidence of ongoing persistence of IgG antibody responses to spike and nucleocapsid after LD followed by tisagenlecleucel despite the B-cell aplasia. Further we were able to detect SARS-CoV-2 specific T-cells recognizing multiple viral structural proteins for several months following CAR-T. The T-cell response was polyfunctional and predominantly CD4 restricted. This data has important implications for the understanding of SARS-CoV-2 immunity in patients with impaired immune systems and the potential application of SARS-CoV-2-specific T-cell therapeutics to treat patients with blood cancers who receive B cell depleting therapy.
Collapse
Affiliation(s)
- Oren Gordon
- Department of Pediatrics, Children’s National Hospital, Washington, DC, USA
| | - Madeline Terpilowski
- Center for Cancer and Immunology Research, Children’s Research Institute, Children’s National Hospital, Washington, DC, USA
| | - Robin Dulman
- Pediatric Specialists of Virginia, Department of Pediatric Hematology and Oncology, Fairfax, VA, USA
| | - Michael D. Keller
- Department of Pediatrics, Children’s National Hospital, Washington, DC, USA
- Center for Cancer and Immunology Research, Children’s Research Institute, Children’s National Hospital, Washington, DC, USA
| | - Peter D. Burbelo
- National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - Jeffrey I. Cohen
- Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Catherine M. Bollard
- Department of Pediatrics, Children’s National Hospital, Washington, DC, USA
- Center for Cancer and Immunology Research, Children’s Research Institute, Children’s National Hospital, Washington, DC, USA
| | - Hema Dave
- Department of Pediatrics, Children’s National Hospital, Washington, DC, USA
- Center for Cancer and Immunology Research, Children’s Research Institute, Children’s National Hospital, Washington, DC, USA
| |
Collapse
|
10
|
Ono K, Ishibashi Y, Kaname S. Successful Kidney and Hematopoietic Stem Cell Transplantation for Malignant Lymphoma from Different Donors: A Case Report and Literature Review. Transplant Proc 2022; 54:1589-1593. [PMID: 35840432 DOI: 10.1016/j.transproceed.2022.05.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 05/21/2022] [Indexed: 10/17/2022]
Abstract
There are particularly few reports on kidney transplantation after hematopoietic stem cell transplantation (HSCT) for malignant lymphoma, and none of the cases reported a favorable outcome in patients who received kidney transplantation from a different donor to HSCT. In this report, we describe the first case of kidney transplantation from a different donor to HSCT with a successful outcome. Furthermore, we reviewed the previously reported cases. A 59-year-old female patient received an HSCT from her younger brother after chemotherapy for malignant lymphoma. After HSCT, she did not have graft-versus-host disease (GVHD) requiring maintenance treatment. The patient developed chronic kidney disease requiring kidney replacement therapy, probably due to drug toxicity or cardio-renal syndrome. At age 65, she underwent an ABO-compatible, HLA-A, -B, -DR 5/6 mismatched kidney transplantation from her husband. Immunosuppressive therapy with tacrolimus, mycophenolate mofetil, methylprednisolone, and basiliximab was administered. The patient had urinary tract infections at 7 days, 9 weeks, and 4 months after kidney transplantation, and cytomegalovirus antigenemia at 9 weeks after kidney transplantation, which improved with antibiotic and valganciclovir, respectively. When each infection occurred, we weakened immunosuppressive therapy. Four years after kidney transplantation, the patient is in good clinical condition with a serum creatinine of 1.2 mg/dL, without critical infection or malignancy. In this case, we believe that it was important to optimize the immunosuppressive therapy. In addition, from a review of previous cases, it seemed important that there was no GVHD requiring maintenance therapy in order to prevent excessive immunosuppression.
Collapse
Affiliation(s)
- Keisuke Ono
- Department of Nephrology and Rheumatology, Kyorin University School of Medicine, Tokyo, Japan; Department of Nephrology, Japanese Red Cross Medical Center, Tokyo, Japan.
| | | | - Shinya Kaname
- Department of Nephrology and Rheumatology, Kyorin University School of Medicine, Tokyo, Japan
| |
Collapse
|
11
|
Assessing long-term effects after stem cell transplantation: design of the MOSA study. J Clin Epidemiol 2022; 148:10-16. [DOI: 10.1016/j.jclinepi.2022.03.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 02/14/2022] [Accepted: 03/21/2022] [Indexed: 11/22/2022]
|
12
|
Goeser F, Sifft B, Stein-Thoeringer C, Farowski F, Strassburg CP, Brossart P, Higgins PG, Scheid C, Wolf D, Holderried TAW, Vehreschild MJGT, Cruz Aguilar MR. Fecal microbiota transfer for refractory intestinal graft-versus-host disease - Experience from two German tertiary centers. Eur J Haematol 2021; 107:229-245. [PMID: 33934412 DOI: 10.1111/ejh.13642] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 04/28/2021] [Accepted: 04/29/2021] [Indexed: 12/17/2022]
Abstract
RATIONALE Steroid refractory graft-vs-host disease (sr-GvHD) represents a challenging complication after allogeneic hematopoietic cell transplantation (allo-HCT). Intestinal microbiota (IM) diversity and dysbiosis were identified as influencing factors for the development of acute GvHD. Fecal microbiota transfer (FMT) is hypothesized to restore IM dysbiosis, but there is limited knowledge about the significance of FMT in the treatment of sr-GvHD. OBJECTIVES We studied the effects of FMT on sr-GvHD in allo-HCT patients from two German tertiary clinical centers (n = 11 patients; period: March 2017 until July 2019). To assess safety and clinical efficacy, we analyzed clinical data pre- and post-FMT (day -14 to +30 relative to FMT). Moreover, IM were analyzed in donor samples and in a subset of patients pre- and post-FMT by 16S rRNA sequencing. RESULTS Post-FMT, we observed no intervention-associated, systemic inflammatory responses and only minor side effects (5/11 patients: abdominal pain and transformation of peristalsis-each 3/11 and vomiting-1/11). Stool frequencies and volumes were significantly reduced [pre- vs post-FMT (d14): P < .05, respectively] as well as clear attenuation regarding both grading and staging of sr-GvHD was present upon FMT. Moreover, IM analyses revealed an increase of alpha diversity as well as a compositional shifts toward the donor post-FMT. CONCLUSIONS In our study, we observed positive effects on sr-GVHD after FMT without the occurrence of major adverse events. Although these findings are in line with published data on beneficial effects of FMT in sr-GvHD, further randomized clinical studies are urgently needed to better define the clinical validity including mode of action.
Collapse
Affiliation(s)
- Felix Goeser
- Department of Internal Medicine I, University Hospital Bonn, Bonn, Germany.,German Center for Infection Research (DZIF), Partner Site Bonn-Cologne, Germany.,German Clinical Microbiome Study Group (GCMSG), Germany
| | - Barbara Sifft
- Department of Internal Medicine III, Oncology, Hematology, Rheumatology and Immune-Oncology, University Hospital Bonn, Bonn, Germany
| | | | - Fedja Farowski
- German Center for Infection Research (DZIF), Partner Site Bonn-Cologne, Germany.,German Clinical Microbiome Study Group (GCMSG), Germany.,Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, University of Cologne, Cologne, Germany.,Department of Internal Medicine, Infectious Diseases, Goethe University Frankfurt, Frankfurt am Main, Germany
| | | | - Peter Brossart
- Department of Internal Medicine III, Oncology, Hematology, Rheumatology and Immune-Oncology, University Hospital Bonn, Bonn, Germany
| | - Paul G Higgins
- German Center for Infection Research (DZIF), Partner Site Bonn-Cologne, Germany.,Institute for Medical Microbiology, Immunology and Hygiene, University of Cologne, Cologne, Germany
| | - Christoph Scheid
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, University of Cologne, Cologne, Germany
| | - Dominik Wolf
- Department of Internal Medicine III, Oncology, Hematology, Rheumatology and Immune-Oncology, University Hospital Bonn, Bonn, Germany.,UKIM 5, Hematology and Oncology, Medical University Innsbruck, Innsbruck, Austria
| | - Tobias A W Holderried
- Department of Internal Medicine III, Oncology, Hematology, Rheumatology and Immune-Oncology, University Hospital Bonn, Bonn, Germany
| | - Maria J G T Vehreschild
- German Center for Infection Research (DZIF), Partner Site Bonn-Cologne, Germany.,German Clinical Microbiome Study Group (GCMSG), Germany.,Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, University of Cologne, Cologne, Germany.,Department of Internal Medicine, Infectious Diseases, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Marta Rebeca Cruz Aguilar
- German Center for Infection Research (DZIF), Partner Site Bonn-Cologne, Germany.,German Clinical Microbiome Study Group (GCMSG), Germany.,Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, University of Cologne, Cologne, Germany
| |
Collapse
|
13
|
Late infectious complications in hematopoietic cell transplantation survivors: a population-based study. Blood Adv 2021; 4:1232-1241. [PMID: 32227211 DOI: 10.1182/bloodadvances.2020001470] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 02/26/2020] [Indexed: 02/08/2023] Open
Abstract
Few studies have compared the incidence of infections occurring ≥2 years after hematopoietic cell transplant (HCT) with other cancer patients and the general population. In this study, ≥2-year HCT survivors who were Washington residents treated from 1992 through 2009 (n = 1792; median age, 46 years; 52% allogeneic; 90% hematologic malignancies) were matched to individuals from the state cancer registry (n = 5455, non-HCT) and driver's license files (n = 16 340; Department of Licensing [DOL]). Based on hospital and death registry codes, incidence rate ratios (IRRs; 95% confidence interval [CI]) of infections by organism type and organ system were estimated using Poisson regression. With 7-year median follow-up, the incidence rate (per 1000 person-years) of all infections was 65.4 for HCT survivors vs 39.6 for the non-HCT group (IRR, 1.6; 95% CI, 1.3-1.9) and 7.2 for DOL (IRR, 10.0; 95% CI, 8.3-12.1). Bacterial and fungal infections were each 70% more common in HCT vs non-HCT cancer survivors (IRR, 1.7; P < .01), whereas the risk for viral infection was lower (IRR, 1.4; P = .07). Among potentially vaccine-preventable organisms, the IRR was 3.0 (95% CI, 2.1-4.3) vs the non-HCT group. Although the incidences of all infections decreased with time, the relative risk in almost all categories remained significantly increased in ≥5-year HCT survivors vs other groups. Risk factors for late infection included history of relapse and for some infections, history of chronic graft-versus-host disease. Providers caring for HCT survivors should maintain vigilance for infections and ensure adherence to antimicrobial prophylaxis and vaccination guidelines.
Collapse
|
14
|
Sy A, Chanson D, Berano Teh J, Wong FL, Nakamura R, Dadwal S, Armenian SH. Late-occurring infections in a contemporary cohort of hematopoietic cell transplantation survivors. Cancer Med 2021; 10:2956-2966. [PMID: 33835722 PMCID: PMC8086032 DOI: 10.1002/cam4.3896] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 02/28/2021] [Accepted: 03/22/2021] [Indexed: 11/12/2022] Open
Abstract
Background There is a paucity of studies describing the incidence and risk factors for late‐occurring (≥1 year) infectious complications in contemporary survivors of hematopoietic cell transplantation (HCT). Methods This was a retrospective cohort study of 641 1‐year survivors of HCT, transplanted between 2010 and 2013 as adults, and in remission from their primary disease. Standardized definitions were used to characterize viral, fungal, and bacterial infections. Cumulative incidence of infections was calculated, with relapse/progression considered as a competing risk event. Fine‐Gray subdistribution hazard ratio estimates and 95% confidence intervals (CI) were obtained, adjusted for relevant covariates. Results Median age at HCT was 55.2 years (range 18.1–78.1 years); 54.0% were survivors of allogeneic HCT. The 5‐year cumulative incidence of a late‐occurring infection for the entire cohort was 31.6%; the incidence of polymicrobial (≥2) infections was 10.1%. In survivors who developed at least one infection, the 5‐year incidence of a subsequent infection was 45.3%. Among allogeneic HCT survivors, patients with acute lymphoblastic (HR = 1.82 95% CI [1.12–2.96]) or myeloid (HR = 1.50 95% CI [1.02–2.20]) leukemia, and those with an elevated HCT‐Comorbidity index score (HR = 1.09 95% CI [1.01–1.17]) were more likely to develop late‐occurring infections; there was an incremental risk associated with severity of graft versus host disease (GVHD) at 1‐year post‐HCT (mild: HR = 2.17, 95% CI [1.09–4.33]; moderate/severe: HR = 3.78, 95% CI [1.90–7.53]; reference: no GVHD). Conclusions The burden of late‐occurring infections in HCT survivors is substantial, and there are important patient‐ and HCT‐related modifiers of risk over time. These findings may help guide personalized screening and prevention strategies to improve outcomes after HCT.
Collapse
Affiliation(s)
- Andrew Sy
- Department of Pediatrics, Lurie Children's Hospital of Chicago, Chicago, IL, USA
| | - Dayana Chanson
- Department of Population Sciences, City of Hope, Duarte, CA, USA
| | | | - Florence L Wong
- Department of Population Sciences, City of Hope, Duarte, CA, USA
| | - Ryotaro Nakamura
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope, Duarte, CA, USA
| | - Sanjeet Dadwal
- Department of Medicine, Division of Infectious Diseases, City of Hope, Duarte, CA, USA
| | - Saro H Armenian
- Department of Population Sciences, City of Hope, Duarte, CA, USA
| |
Collapse
|
15
|
Riddell P, Vasudevan-Nampoothiri R, Ma J, Singer LG, Lipton JH, Juvet SC. Lung transplantation for late-onset non-infectious chronic pulmonary complications of allogenic hematopoietic stem cell transplant. Respir Res 2021; 22:101. [PMID: 33827576 PMCID: PMC8025894 DOI: 10.1186/s12931-021-01699-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 03/29/2021] [Indexed: 11/12/2022] Open
Abstract
Background Late onset non-infectious pulmonary complications (LONIPCs) following allogenic hematopoietic stem cell transplantation (allo-HSCT) confer a significant mortality risk. Lung transplantation (LTx) has the potential to provide survival benefit but the impact of prior allo-HSCT on post-LTx outcomes is not well studied.
Methods This retrospective, single-centre cohort study assessed the post-LTx outcomes of adults with LONIPCs of allo-HSCT. Outcomes of LTx for LONIPCs were compared to propensity-score matched LTx controls (n = 38, non-HSCT) and recipients of re-LTx (n = 70) for chronic lung allograft dysfunction (CLAD).
Results Nineteen patients underwent DLTx for LONIPCs of allo-HSCT between 2003 and 2019. Post-LTx survival was 50% at 5-years. Survival to 1-year post-LTx was similar to matched controls (p = 0.473). Survival, conditional on 1-year survival, was lower in the allo-HSCT cohort (p = 0.034). An increased risk of death due to infection was identified in the allo-HSCT cohort compared to matched controls (p = 0.003). Compared to re-LTx recipients, the allo-HSCT cohort had superior survival to 1-year post-LTx (p = 0.034) but conditional 1-year survival was similar (p = 0.145).
Conclusion This study identifies an increased risk of post-LTx mortality in recipients with previous allo-HSCT, associated with infection. It supports the hypothesis that allo-HSCT LTx recipients are relatively more immunosuppressed than patients undergoing LTx for other indications. Optimisation of post-LTx immunosuppressive and antimicrobial strategies to account for this finding should be considered.
Supplementary Information The online version contains supplementary material available at 10.1186/s12931-021-01699-8.
Collapse
Affiliation(s)
- Peter Riddell
- Ajmera Transplant Centre and Toronto Lung Transplant Program, Toronto General Hospital, Toronto, Canada
| | - Ram Vasudevan-Nampoothiri
- Hans Messner Allogenic Blood and Marrow Transplant Program, Princess Margaret Cancer Centre, Toronto, Canada
| | - Jin Ma
- Biostatistics Research Unit, University Health Network, University of Toronto, Toronto, Canada
| | - Lianne G Singer
- Ajmera Transplant Centre and Toronto Lung Transplant Program, Toronto General Hospital, Toronto, Canada
| | - Jeff H Lipton
- Hans Messner Allogenic Blood and Marrow Transplant Program, Princess Margaret Cancer Centre, Toronto, Canada
| | - Stephen C Juvet
- Ajmera Transplant Centre and Toronto Lung Transplant Program, Toronto General Hospital, Toronto, Canada.
| |
Collapse
|
16
|
Carpenter PA, Englund JA. Commentary: Is Immune Recovery-Based Post-Transplantation Vaccination in Children Better Than Time-Based Revaccination? Transplant Cell Ther 2021; 27:281-283. [PMID: 33836865 DOI: 10.1016/j.jtct.2021.03.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 03/04/2021] [Indexed: 10/22/2022]
Affiliation(s)
- Paul A Carpenter
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington; University of Washington Department of Pediatrics, Seattle, Washington.
| | - Janet A Englund
- University of Washington Department of Pediatrics, Seattle, Washington; Pediatric Transplant Infectious Disease, Seattle Children's Hospital, Seattle, Washington; Affiliate, Fred Hutchinson Cancer Research Center, Seattle, Washington
| |
Collapse
|
17
|
Walsh LF, Sherbuk JE, Wispelwey B. Pneumococcal induced thrombotic thrombocytopenic purpura with features of purpura fulminans. BMJ Case Rep 2021; 14:14/1/e235580. [PMID: 33500295 PMCID: PMC7843298 DOI: 10.1136/bcr-2020-235580] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
A 42-year-old woman with a history of acute myeloid leukaemia status postallogeneic stem cell transplant presented with fevers, altered mental status, pulmonary infiltrates and septic shock that further progressed to thrombocytopenia and purpura fulminans. Laboratory studies were consistent with a diagnosis of thrombotic thrombocytopenic purpura (TTP). Blood cultures grew Streptococcus pneumoniae On chart review, our patient had a history of low immunoglobulin levels following stem cell transplant, which may have predisposed her to pneumococcal infection. The patient responded to therapy with ceftriaxone, plasma exchange, rituximab and caplacizumab. This is the fourth-documented case of pneumococcal induced TTP and, to the best of our knowledge, the first-describing pneumococcal induced TTP with purpura fulminans. We conclude that patients with TTP should be evaluated for infectious aetiologies and empiric antibiotics should be considered. Clinicians should be aware of the possibility for TTP to lead to purpura fulminans.
Collapse
Affiliation(s)
- Laura Frances Walsh
- Internal Medicine, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Jacqueline E Sherbuk
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, Virginia, USA
| | - Brian Wispelwey
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, Virginia, USA
| |
Collapse
|
18
|
Zinter MS, Hume JR. Effects of Hematopoietic Cell Transplantation on the Pulmonary Immune Response to Infection. Front Pediatr 2021; 9:634566. [PMID: 33575235 PMCID: PMC7871005 DOI: 10.3389/fped.2021.634566] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Accepted: 01/04/2021] [Indexed: 12/20/2022] Open
Abstract
Pulmonary infections are common in hematopoietic cell transplant (HCT) patients of all ages and are associated with high levels of morbidity and mortality. Bacterial, viral, fungal, and parasitic pathogens are all represented as causes of infection. The lung mounts a complex immune response to infection and this response is significantly affected by the pre-HCT conditioning regimen, graft characteristics, and ongoing immunomodulatory therapy. We review the published literature, including animal models as well as human data, to describe what is known about the pulmonary immune response to infection in HCT recipients. Studies have focused on the pulmonary immune response to Aspergillus fumigatus, gram-positive and gram-negative bacteria, and viruses, and show a range of defects associated with both the innate and adaptive immune responses after HCT. There are still many open areas for research, to delineate novel therapeutic targets for pulmonary infections as well as to explore linkages to non-infectious inflammatory lung conditions.
Collapse
Affiliation(s)
- Matt S. Zinter
- Department of Pediatrics, Divisions of Critical Care and Bone Marrow Transplantation, University of California, San Francisco, San Francisco, CA, United States
| | - Janet R. Hume
- Department of Pediatrics, Division of Critical Care Medicine, University of Minnesota Medical School, Minnesota, MN, United States
| |
Collapse
|
19
|
Stratton P, Battiwalla M, Tian X, Abdelazim S, Baird K, Barrett AJ, Cantilena CR, Childs RW, DeJesus J, Fitzhugh C, Fowler D, Gea-Banacloche J, Gress RE, Hickstein D, Hsieh M, Ito S, Kemp TJ, Khachikyan I, Merideth MA, Pavletic SZ, Quint W, Schiffman M, Scrivani C, Shanis D, Shenoy AG, Struijk L, Tisdale JF, Wagner S, Williams KM, Yu Q, Wood LV, Pinto LA. Immune Response Following Quadrivalent Human Papillomavirus Vaccination in Women After Hematopoietic Allogeneic Stem Cell Transplant: A Nonrandomized Clinical Trial. JAMA Oncol 2021; 6:696-705. [PMID: 32105293 DOI: 10.1001/jamaoncol.2019.6722] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Importance Human papillomavirus (HPV) infection is found in about 40% of women who survive allogeneic hematopoietic stem cell transplant and can induce subsequent neoplasms. Objective To determine the safety and immunogenicity of the quadrivalent HPV vaccine (HPV-6, -11, -16, and -18) in clinically stable women post-allogeneic transplant compared with female healthy volunteers. Interventions Participants received the quadrivalent HPV vaccine in intramuscular injections on days 1 and 2 and then 6 months later. Design, Setting, and Participants This prospective, open-label phase-1 study was conducted in a government clinical research hospital and included clinically stable women posttransplant who were or were not receiving immunosuppressive therapy compared with healthy female volunteers age 18 to 50 years who were followed up or a year after first receiving quadrivalent HPV vaccination. The study was conducted from June 2, 2010, until July 19, 2016. After all of the results of the study assays were completed and available in early 2018, the analysis took place from February 2018 to May 2019. Main Outcomes and Measures Anti-HPV-6, -11, -16, and -18-specific antibody responses using L1 virus-like particle enzyme-linked immunosorbent assay were measured in serum before (day 1) and at months 7 and 12 postvaccination. Anti-HPV-16 and -18 neutralization titers were determined using a pseudovirion-based neutralization assay. Results Of 64 vaccinated women, 23 (35.9%) were receiving immunosuppressive therapy (median age, 34 years [range, 18-48 years]; median 1.2 years posttransplant), 21 (32.8%) were not receiving immunosuppression (median age, 32 years [range, 18-49 years]; median 2.5 years posttransplant), and 20 (31.3%) were healthy volunteers (median age, 32 years [range, 23-45 years]). After vaccine series completion, 18 of 23 patients receiving immunosuppression (78.3%), 20 of 21 not receiving immunosuppression (95.2%), and all 20 volunteers developed antibody responses to all quadrivalent HPV vaccine types (P = .04, comparing the 3 groups). Geometric mean antibody levels for each HPV type were higher at months 7 and 12 than at baseline in each group (all geometric mean ratios >1; P < .001) but not significantly different across groups. Antibody and neutralization titers for anti-HPV-16 and anti-HPV-18 correlated at month 7 (Spearman ρ = 0.92; P < .001 for both). Adverse events were mild and not different across groups. Conclusions and Relevance Treatment with the HPV vaccination was followed by strong, functionally active antibody responses against vaccine-related HPV types and no serious adverse events. These findings suggest that HPV vaccination may be safely administered to women posttransplant to potentially reduce HPV infection and related neoplasia. Trial Registration ClinicalTrials.gov Identifier: NCT01092195.
Collapse
Affiliation(s)
- Pamela Stratton
- Office of the Clinical Director, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland.,Program in Reproductive and Adult Endocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland
| | - Minoo Battiwalla
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland.,Sarah Cannon Research Institute, Nashville, Tennessee
| | - Xin Tian
- Office of Biostatistics Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Suzanne Abdelazim
- Clinical Center, National Institutes of Health, Bethesda, Maryland.,Riverside Regional Medical Center, Newport News, Virginia
| | - Kristin Baird
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - A John Barrett
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland.,GW Cancer Center, The George Washington University Hospital, Washington, DC
| | - Caroline R Cantilena
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland.,University of Kansas School of Medicine, Kansas City
| | - Richard W Childs
- Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Jessica DeJesus
- Office of Biostatistics Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Courtney Fitzhugh
- Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Daniel Fowler
- Experimental Transplant and Immunology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland.,Rapa Therapeutics, Rockville, Maryland
| | - Juan Gea-Banacloche
- Experimental Transplant and Immunology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland.,Infectious Diseases Division, Mayo Clinic Arizona, Phoenix, Arizona
| | - Ronald E Gress
- Experimental Transplant and Immunology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Dennis Hickstein
- Experimental Transplant and Immunology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Matthew Hsieh
- Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Sawa Ito
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland.,Hematopoietic Stem Cell Transplant and Cell Therapy, Division of Hematology-Oncology, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Troy J Kemp
- HPV Immunology Laboratory, Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Izabella Khachikyan
- Program in Reproductive and Adult Endocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland.,Office of New Drugs, Center for Drug Evaluation and Research, Division of Anesthesia, Analgesia, and Addiction Products, US Food and Drug Administration, Silver Spring, Maryland
| | - Melissa A Merideth
- Office of the Clinical Director, National Human Genome Research Institute, Bethesda, Maryland
| | - Steven Z Pavletic
- Experimental Transplant and Immunology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Wim Quint
- DDL Diagnostic Laboratory, Rijswijk, the Netherlands
| | - Mark Schiffman
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, Maryland
| | - Claire Scrivani
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland.,University of Virginia School of Medicine, Charlottesville
| | - Dana Shanis
- Program in Reproductive and Adult Endocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland.,Rittenhouse Women's Wellness Center, Philadelphia, Pennsylvania
| | - Aarthi G Shenoy
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland.,Department of Hematology/Oncology, MedStar Washington Hospital Center, Washington, DC
| | - Linda Struijk
- DDL Diagnostic Laboratory, Rijswijk, the Netherlands
| | - John F Tisdale
- Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Sarah Wagner
- Cancer Genomics Research Laboratory, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research Inc, Frederick, Maryland
| | - Kirsten M Williams
- Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland.,Children's Research Institute, Children's National, Washington, DC
| | - Quan Yu
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Lauren V Wood
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland.,PDS Biotechnology, Berkeley Heights, New Jersey
| | - Ligia A Pinto
- HPV Immunology Laboratory, Frederick National Laboratory for Cancer Research, Frederick, Maryland
| |
Collapse
|
20
|
Winkler J, Tittlbach H, Schneider A, Buchstaller C, Mayr A, Vasova I, Roesler W, Mach M, Mackensen A, Winkler TH. Measuring the cellular memory B cell response after vaccination in patients after allogeneic stem cell transplantation. Ann Hematol 2020; 99:1895-1906. [PMID: 32519092 PMCID: PMC7340644 DOI: 10.1007/s00277-020-04072-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 04/28/2020] [Indexed: 02/04/2023]
Abstract
After allogeneic hematopoietic stem cell transplantation (HSCT), patients are repetitively vaccinated to reduce the risk of infection caused by the immune deficiency following allogeneic HSCT. By the vaccination of transplanted patients, the humoral memory function can be restored in the majority of cases. It is unknown, however, to what extent memory B cells derived from the donor contribute to the mobilization of antibody-secreting cells and long-term humoral memory in patients after allogeneic HSCT. We therefore analyzed patients after allogeneic HSCT for memory B cell responses 7 days after single vaccination against tetanus toxoid (TT), diphtheria toxoid (DT), pertussis toxoid (PT), Haemophilus influenzae type b (Hib), and poliovirus. Patients showed an insufficient mobilization of plasmablasts (PB) after vaccination, whereas healthy subjects (HD, n = 13) exhibited a significant increase of PB in the peripheral blood. Regarding vaccine-specific antibody-secreting PB, all HD responded against all vaccine antigens, as expected. However, only 65% of the patients responded with a measurable increase in IgG-secreting PB against TT, 65% against DT, 33% against PT, and 53% against poliovirus. Correspondingly, the antibody titers on day 7 after vaccination did not increase in patients. A significant increase of serum titers for the vaccine antigens was detectable in the majority of patients only after repetitive vaccinations. In contrast to the low mobilization of vaccine-specific PB after vaccination, a high number of PB before vaccination was detectable in patients following allogeneic HSCT. High frequencies of circulating PB correlated with the incidence of moderate/severe chronic GVHD. In summary, patients showed a weak mobilization of antigen-specific PB and an inadequate increase in antibody titers 7 days after the first vaccination. Patients with moderate or severe chronic GVHD in their history had a significantly higher percentage of IgG-secreting PB prior to vaccination. The antigen specificity of these IgG-secreting PB is currently unknown.
Collapse
Affiliation(s)
- Julia Winkler
- Department of Internal Medicine 5, Hematology/Oncology, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nuremberg, Glückstrasse 6, 91054, Erlangen, Germany.
| | - Hannes Tittlbach
- Department of Internal Medicine 5, Hematology/Oncology, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nuremberg, Glückstrasse 6, 91054, Erlangen, Germany.,Department of Biology, Division of Genetics, Nikolaus-Fiebiger-Center for Molecular Medicine, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Andrea Schneider
- Department of Biology, Division of Genetics, Nikolaus-Fiebiger-Center for Molecular Medicine, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Corinna Buchstaller
- Department of Medical Informatics, Biometry, and Epidemiology, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Andreas Mayr
- Department of Medical Biometry, Informatics and Epidemiology, University Hospital Bonn, Bonn, Germany
| | - Ingrid Vasova
- Department of Internal Medicine 5, Hematology/Oncology, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nuremberg, Glückstrasse 6, 91054, Erlangen, Germany
| | - Wolf Roesler
- Department of Internal Medicine 5, Hematology/Oncology, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nuremberg, Glückstrasse 6, 91054, Erlangen, Germany
| | - Michael Mach
- Institute for Clinical and Molecular Virology, University Hospital Erlangen, Erlangen, Germany
| | - Andreas Mackensen
- Department of Internal Medicine 5, Hematology/Oncology, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nuremberg, Glückstrasse 6, 91054, Erlangen, Germany
| | - Thomas H Winkler
- Department of Biology, Division of Genetics, Nikolaus-Fiebiger-Center for Molecular Medicine, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| |
Collapse
|
21
|
Ghesani N, Gavane S, Hafez A, Kostakoglu L. PET in Lymphoma. Clin Nucl Med 2020. [DOI: 10.1007/978-3-030-39457-8_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
22
|
Page KM, Stenger EO, Connelly JA, Shyr D, West T, Wood S, Case L, Kester M, Shim S, Hammond L, Hammond M, Webb C, Biffi A, Bambach B, Fatemi A, Kurtzberg J. Hematopoietic Stem Cell Transplantation to Treat Leukodystrophies: Clinical Practice Guidelines from the Hunter's Hope Leukodystrophy Care Network. Biol Blood Marrow Transplant 2019; 25:e363-e374. [PMID: 31499213 DOI: 10.1016/j.bbmt.2019.09.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 08/09/2019] [Accepted: 09/02/2019] [Indexed: 12/22/2022]
Abstract
The leukodystrophies are a heterogeneous group of inherited diseases characterized by progressive demyelination of the central nervous system leading to devastating neurologic symptoms and premature death. Hematopoietic stem cell transplantation (HSCT) has been successfully used to treat certain leukodystrophies, including adrenoleukodystrophy, globoid leukodystrophy (Krabbe disease), and metachromatic leukodystrophy, over the past 30 years. To date, these complex patients have primarily been transplanted at a limited number of pediatric centers. As the number of cases identified through pregnancy and newborn screening is increasing, additional centers will be required to treat these children. Hunter's Hope created the Leukodystrophy Care Network in part to create and standardize high-quality clinical practice guidelines to guide the care of affected patients. In this report the clinical guidelines for the care of pediatric patients with leukodystrophies undergoing treatment with HSCT are presented. The initial transplant evaluation, determination of patient eligibility, donor selection, conditioning, supportive care, and post-transplant follow-up are discussed. Throughout these guidelines the need for early detection and treatment and the role of the partnership between families and multidisciplinary providers are emphasized.
Collapse
Affiliation(s)
- Kristin M Page
- Pediatric Transplant and Cellular Therapy, Duke University, Durham, North Carolina.
| | - Elizabeth O Stenger
- Aflac Cancer & Blood Disorders Center, Children's Hospital of Atlanta/Emory University
| | - James A Connelly
- Monroe Carell Jr. Children's Hospital at Vanderbilt University, Nashville, Tennessee
| | - David Shyr
- Division of Pediatric Hematology/Oncology, University of Utah School of Medicine
| | - Tara West
- Pediatric Transplant and Cellular Therapy, Duke University, Durham, North Carolina
| | - Susan Wood
- Pediatric Transplant and Cellular Therapy, Duke University, Durham, North Carolina
| | - Laura Case
- Pediatric Transplant and Cellular Therapy, Duke University, Durham, North Carolina
| | - Maureen Kester
- Pediatric Transplant and Cellular Therapy, Duke University, Durham, North Carolina
| | - Soo Shim
- Ann & Robert H. Lurie Children's Hospital, Chichago, Illinois
| | - Lauren Hammond
- Leukodystrophy Care Network Steering Committee, Orchard Park, New York
| | - Matthew Hammond
- Leukodystrophy Care Network Steering Committee, Orchard Park, New York
| | - Christin Webb
- Leukodystrophy Care Network Steering Committee, Orchard Park, New York
| | - Alessandra Biffi
- Dana Farber/Boston Children's Cancer and Blood Disorders Center, Boston, Massachusetts
| | | | - Ali Fatemi
- Moser Center for Leukodystrophies, Kennedy Krieger Institute, Johns Hopkins University, Baltimore, Maryland
| | - Joanne Kurtzberg
- Pediatric Transplant and Cellular Therapy, Duke University, Durham, North Carolina
| |
Collapse
|
23
|
Safety and immunogenicity of conjugate quadrivalent meningococcal vaccination after hematopoietic cell transplantation. Blood Adv 2019; 2:1272-1276. [PMID: 29871892 DOI: 10.1182/bloodadvances.2018018911] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Accepted: 05/11/2018] [Indexed: 12/11/2022] Open
Abstract
Immunization with the conjugated quadrivalent (serogroups A, C, Y, and W-135) meningococcal vaccine (MCV4) after hematopoietic cell transplantation (HCT) is recommended. However, immune responses to MCV4 have not been prospectively studied after HCT. We conducted a vaccine response study among 67 adults who received 1 MCV4 dose a year after autologous or allogeneic HCT from January to September 2014. Pre- and postvaccination serogroup serum bactericidal antibody (SBA) titers were measured a median of 57 days after vaccination. Serogroup-specific responses were defined as a fourfold increase in SBA titer with postvaccination titers ≥1:8. Prior to vaccination, 44 (65.7%) patients had no protective titers (<1:8) to any meningococcal serogroup, and 3 (4.5%) patients had protective titers to all 4 serogroups. The median serogroup-specific postvaccination SBA titers were 1:2048 for A, 1:64 for C, 1:128 for W-135, and 1:128 for Y (P < .001 for all pre- and postvaccination pairwise comparisons; similar among serogroups, Spearman ρ 0.5-0.6, P < .0001). Among serogroup-specific nonimmune patients prior to vaccination, serogroup-specific response rates were 76.9%, 65.5%, 51.7%, and 65% to serogroups A, C, W-135, and Y, respectively. One dose of MCV4 elicited protective titers in the majority of patients. These data suggest that a second vaccine dose may be beneficial.
Collapse
|
24
|
Dyer G, Gilroy N, Brice L, Kabir M, Gottlieb D, Huang G, Hogg M, Brown L, Greenwood M, Larsen SR, Moore J, Hertzberg M, Tan J, Ward C, Kerridge I. A survey of infectious diseases and vaccination uptake in long-term hematopoietic stem cell transplant survivors in Australia. Transpl Infect Dis 2019; 21:e13043. [PMID: 30585673 DOI: 10.1111/tid.13043] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 11/17/2018] [Accepted: 12/09/2018] [Indexed: 01/05/2023]
Abstract
BACKGROUND This cross-sectional survey aimed to establish the prevalence of infectious diseases and vaccination uptake in long-term allogeneic hematopoietic stem cell transplants (HSCT) survivors in New South Wales, in order to reduce long-term post-HSCT morbidity and mortality and enhance long-term care. PATIENTS AND METHODS Hematopoietic stem cell transplants survivors aged over 18 years and transplanted between 2000-2012 in New South Wales (NSW) were eligible to participate. Survivors self-completed the Sydney Post BMT Study survey, FACT-BMT (V4), Chronic Graft versus Host Disease (cGVHD) Activity Assessment Self Report, Lee Chronic GvHD Symptom Scale, DASS21, Post Traumatic Growth Inventory, and the Fear of Recurrence Scale. RESULTS Of the 583 HSCT survivors contacted, 441 (78%) completed the survey. Respondents included 250 (57%) males and median age was 54 years (range 19-79 years). The median age at the time of transplant was 49 years (Range: 17-71), the median time since HSCT was 5 years (Range: 1-14) and 69% had cGVHD. Collectively, 41.7% of survivors reported a vaccine preventable disease (VPD) with the most common being influenza-like-illness (38.4%), varicella zoster/shingles (27.9%), pap smear abnormalities (9.8%), pneumococcal disease (5.1%), and varicella zoster (chicken pox) (4.6%). Only 31.8% had received the full post-HSCT vaccination schedule, and the majority (69.8%) of these had received the vaccines via their General Practitioner. cGVHD was not found to be a significant factor on multivariate analysis for those who were vaccinated. There was a trend toward lower vaccination rates in patients in a lower income strata. CONCLUSIONS Vaccinating post-HSCT survivors to prevent infections and their consequences have an established role in post-HSCT care. Improving rates of post-HSCT vaccination should be a major priority for BMT units.
Collapse
Affiliation(s)
- Gemma Dyer
- Blood and Marrow Transplant Network, New South Wales Agency for Clinical Innovation, Sydney, New South Wales, Australia.,Faculty of Medicine, Northern Clinical School, University of Sydney, Sydney, New South Wales, Australia
| | - Nicole Gilroy
- Blood and Marrow Transplant Network, New South Wales Agency for Clinical Innovation, Sydney, New South Wales, Australia
| | - Lisa Brice
- Department of Haematology, Royal North Shore Hospital, Sydney, New South Wales, Australia
| | - Masura Kabir
- Westmead Breast Cancer Institute, Sydney, New South Wales, Australia
| | - David Gottlieb
- Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia.,Department of Haematology, Westmead Hospital, Sydney, New South Wales, Australia
| | - Gillian Huang
- Department of Haematology, Westmead Hospital, Sydney, New South Wales, Australia
| | - Megan Hogg
- Department of Haematology, Westmead Hospital, Sydney, New South Wales, Australia
| | - Louisa Brown
- Department of Haematology, Calvary Mater Newcastle, Newcastle, New South Wales, Australia
| | - Matt Greenwood
- Faculty of Medicine, Northern Clinical School, University of Sydney, Sydney, New South Wales, Australia.,Department of Haematology, Royal North Shore Hospital, Sydney, New South Wales, Australia.,Northern Blood Research Centre, Kolling Institute, University of Sydney, New South Wales, Australia
| | - Stephen R Larsen
- Institute of Haematology, Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
| | - John Moore
- Department of Haematology, St Vincents Hospital, Sydney, New South Wales, Australia
| | - Mark Hertzberg
- Department of Haematology, Prince of Wales Hospital, Sydney, New South Wales, Australia
| | - Jeff Tan
- Department of Haematology, St Vincents Hospital, Sydney, New South Wales, Australia
| | - Christopher Ward
- Faculty of Medicine, Northern Clinical School, University of Sydney, Sydney, New South Wales, Australia.,Department of Haematology, Royal North Shore Hospital, Sydney, New South Wales, Australia.,Northern Blood Research Centre, Kolling Institute, University of Sydney, New South Wales, Australia
| | - Ian Kerridge
- Faculty of Medicine, Northern Clinical School, University of Sydney, Sydney, New South Wales, Australia.,Department of Haematology, Royal North Shore Hospital, Sydney, New South Wales, Australia.,Northern Blood Research Centre, Kolling Institute, University of Sydney, New South Wales, Australia
| |
Collapse
|
25
|
Shah GL, Majhail N, Khera N, Giralt S. Value-Based Care in Hematopoietic Cell Transplantation and Cellular Therapy: Challenges and Opportunities. Curr Hematol Malig Rep 2018; 13:125-134. [PMID: 29484578 DOI: 10.1007/s11899-018-0444-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
PURPOSE OF REVIEW Improved tolerability and outcomes after hematopoietic cell transplantation (HCT), along with the availability of alternative donors, have expanded its use. With this growth, and the development of additional cellular therapies, we also aim to increase effectiveness, efficiency, and the quality of the care provided. Fundamentally, the goal of value-based care is to have better health outcomes with streamlined processes, improved patient experience, and lower costs for both the patients and the health care system. HCT and cellular therapy treatments are multiphase treatments which allow for interventions at each juncture. RECENT FINDINGS We present a summary of the current literature with focus on program structure and overall system capacity, coordination of therapy across providers, standardization across institutions, diversity and disparities in care, patient quality of life, and cost implications. Each of these topics provides challenges and opportunities to improve value-based care for HCT and cellular therapy patients.
Collapse
Affiliation(s)
- Gunjan L Shah
- Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, Box 298, New York, NY, 10065, USA.
| | - Navneet Majhail
- Blood and Marrow Transplant Program, Cleveland Clinic, Cleveland, OH, USA
| | - Nandita Khera
- Division of Hematology/Oncology, Mayo Clinic, Phoenix, AZ, USA
| | - Sergio Giralt
- Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, Box 298, New York, NY, 10065, USA
| |
Collapse
|
26
|
Swimm A, Giver CR, DeFilipp Z, Rangaraju S, Sharma A, Ulezko Antonova A, Sonowal R, Capaldo C, Powell D, Qayed M, Kalman D, Waller EK. Indoles derived from intestinal microbiota act via type I interferon signaling to limit graft-versus-host disease. Blood 2018; 132:2506-2519. [PMID: 30257880 PMCID: PMC6284212 DOI: 10.1182/blood-2018-03-838193] [Citation(s) in RCA: 114] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 09/19/2018] [Indexed: 01/05/2023] Open
Abstract
The intestinal microbiota in allogeneic bone marrow transplant (allo-BMT) recipients modulates graft-versus-host disease (GVHD), a systemic inflammatory state initiated by donor T cells that leads to colitis, a key determinant of GVHD severity. Indole or indole derivatives produced by tryptophan metabolism in the intestinal microbiota limit intestinal inflammation caused by diverse stressors, so we tested their capacity to protect against GVHD in murine major histocompatibility complex-mismatched models of allo-BMT. Indole effects were assessed by colonization of allo-BMT recipient mice with tryptophanase positive or negative strains of Escherichia coli, or, alternatively, by exogenous administration of indole-3-carboxaldehyde (ICA), an indole derivative. Treatment with ICA limited gut epithelial damage, reduced transepithelial bacterial translocation, and decreased inflammatory cytokine production, reducing GVHD pathology and GVHD mortality, but did not compromise donor T-cell-mediated graft-versus-leukemia responses. ICA treatment also led to recipient-strain-specific tolerance of engrafted T cells. Transcriptional profiling and gene ontology analysis indicated that ICA administration upregulated genes associated with the type I interferon (IFN1) response, which has been shown to protect against radiation-induced intestinal damage and reduce subsequent GVHD pathology. Accordingly, protective effects of ICA following radiation exposure were abrogated in mice lacking IFN1 signaling. Taken together, these data indicate that indole metabolites produced by the intestinal microbiota act via type I IFNs to limit intestinal inflammation and damage associated with myeloablative chemotherapy or radiation exposure and acute GVHD, but preserve antitumor responses, and may provide a therapeutic option for BMT patients at risk for GVHD.
Collapse
Affiliation(s)
- Alyson Swimm
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA
| | - Cynthia R Giver
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, GA
| | - Zachariah DeFilipp
- Blood and Marrow Transplant Program, Massachusetts General Hospital, Boston, MA
| | - Sravanti Rangaraju
- Department of Hematology and Oncology, Indiana University, Indianapolis, IN
| | - Akshay Sharma
- Pediatric Hematology and Oncology, St. Jude Children's Research Hospital, Memphis, TN
| | - Alina Ulezko Antonova
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, GA
| | - Robert Sonowal
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA
| | - Christopher Capaldo
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA
| | - Domonica Powell
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA
- Immunology and Molecular Pathogenesis Graduate Program, Emory University School of Medicine, Atlanta, GA; and
| | - Muna Qayed
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Department of Pediatrics, Emory University, Atlanta, GA
| | - Daniel Kalman
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA
| | - Edmund K Waller
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, GA
| |
Collapse
|
27
|
Wolf D, Bader CS, Barreras H, Copsel S, Pfeiffer BJ, Lightbourn CO, Altman NH, Komanduri KV, Levy RB. Superior immune reconstitution using Treg-expanded donor cells versus PTCy treatment in preclinical HSCT models. JCI Insight 2018; 3:121717. [PMID: 30333311 DOI: 10.1172/jci.insight.121717] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 08/30/2018] [Indexed: 12/20/2022] Open
Abstract
Posttransplant cyclophosphamide (PTCy) has been found to be effective in ameliorating acute graft-versus-host disease (GVHD) in patients following allogeneic hematopoietic stem cell transplantation (aHSCT). Adoptive transfer of high numbers of donor Tregs in experimental aHSCT has shown promise as a therapeutic modality for GVHD regulation. We recently described a strategy for in vivo Treg expansion targeting two receptors: TNFRSF25 and CD25. To date, there have been no direct comparisons between the use of PTCy and Tregs regarding outcome and immune reconstitution within identical groups of transplanted mice. Here, we assessed these two strategies and found both decreased clinical GVHD and improved survival long term. However, recipients transplanted with Treg-expanded donor cells (TrED) exhibited less weight loss early after HSCT. Additionally, TrED recipients demonstrated less thymic damage, significantly more recent thymic emigrants, and more rapid lymphoid engraftment. Three months after HSCT, PTCy-treated and TrED recipients showed tolerance to F1 skin allografts and comparable immune function. Overall, TrED was found superior to PTCy with regard to weight loss early after transplant and initial lymphoid engraftment. Based on these findings, we speculate that morbidity and mortality after transplant could be diminished following TrED transplant into aHSCT recipients, and, therefore, that TrED could provide a promising clinical strategy for GVHD prophylaxis.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Krishna V Komanduri
- Sylvester Comprehensive Cancer Center.,Department of Microbiology & Immunology.,Department of Medicine, Miller School of Medicine, University of Miami, Miami, Florida, USA
| | - Robert B Levy
- Sylvester Comprehensive Cancer Center.,Department of Microbiology & Immunology.,Department of Medicine, Miller School of Medicine, University of Miami, Miami, Florida, USA
| |
Collapse
|
28
|
de Silva HD, Ffrench RA, Korem M, Orlowski E, Curtis DJ, Spencer A, Avery S, Patil S, Morrissey CO. Contemporary analysis of functional immune recovery to opportunistic and vaccine-preventable infections after allogeneic haemopoietic stem cell transplantation. Clin Transl Immunology 2018; 7:e1040. [PMID: 30323928 PMCID: PMC6173278 DOI: 10.1002/cti2.1040] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2018] [Revised: 09/02/2018] [Accepted: 09/03/2018] [Indexed: 01/23/2023] Open
Abstract
Objectives Infections are a major cause of mortality after allogeneic haemopoietic stem cell transplantation (alloHSCT), and immune recovery is necessary for prevention. Novel transplant procedures have changed the epidemiology of infections but contemporary data on functional immune recovery are limited. In this pilot study, we aimed to measure immune recovery in the current era of alloHSCT. Methods Twenty, 13, 11, 9 and 9 alloHSCT recipients had blood collected at baseline (time of conditioning) and 3‐, 6‐, 9‐, and 12‐months post‐alloHSCT, respectively. Clinical data were collected, and immune recovery was measured using immunophenotyping, lymphocyte proliferation, cytokine analysis and antibody isotyping. Results Median absolute T‐ and B‐cell counts were below normal from baseline until 9‐ to 12‐months post‐alloHSCT. Median absolute CD4+ T‐cell counts recovered at 12‐months post‐alloHSCT. Positive proliferative responses to Aspergillus, cytomegalovirus (CMV), Epstein‐Barr virus (EBV), influenza and tetanus antigens were detected from 9 months. IL‐6 was the most abundant cytokine in cell cultures. In cultures stimulated with CMV, EBV, influenza and tetanus peptides, the CD4+ T‐cell count correlated with IL‐1β (P = 0.045) and CD8+ T‐cell count with IFNγ (P = 0.013) and IL‐1β (P = 0.012). The NK‐cell count correlated with IL‐1β (P = 0.02) and IL‐17a (P = 0.03). Median serum levels of IgG1, IgG2 and IgG3 were normal while IgG4 and IgA were below normal range throughout follow‐up. Conclusions This pilot study demonstrates that immune recovery can be measured using CD4+ T‐cell counts, in vitro antigen stimulation and selected cytokines (IFNγ, IL‐1β, IL‐4, IL‐6, IL‐17, IL‐21, IL‐31) in alloHSCT recipients. While larger studies are required, monitoring immune recovery may have utility in predicting infection risk post‐alloHSCT.
Collapse
Affiliation(s)
- Harini D de Silva
- Burnet Institute Life Sciences Discipline Melbourne VIC Australia.,Department of Infectious Diseases Alfred Health and Monash University Melbourne VIC Australia.,Present address: Peter MacCallum Cancer Centre Melbourne VIC Australia
| | - Rosemary A Ffrench
- Burnet Institute Life Sciences Discipline Melbourne VIC Australia.,Department of Immunology Central Clinical School Monash University Melbourne VIC Australia
| | - Maya Korem
- Department of Infectious Diseases Alfred Health and Monash University Melbourne VIC Australia.,Present address: Hadassah University Medical Centre Jerusalem Israel
| | - Eva Orlowski
- Burnet Institute Life Sciences Discipline Melbourne VIC Australia
| | - David J Curtis
- Australian Centre for Blood Diseases Monash University Melbourne VIC Australia.,Malignant Haematology and Stem Cell Transplantation Service Alfred Health Melbourne VIC Australia
| | - Andrew Spencer
- Australian Centre for Blood Diseases Monash University Melbourne VIC Australia.,Malignant Haematology and Stem Cell Transplantation Service Alfred Health Melbourne VIC Australia
| | - Sharon Avery
- Malignant Haematology and Stem Cell Transplantation Service Alfred Health Melbourne VIC Australia
| | - Sushrut Patil
- Malignant Haematology and Stem Cell Transplantation Service Alfred Health Melbourne VIC Australia
| | - Catherine Orla Morrissey
- Department of Infectious Diseases Alfred Health and Monash University Melbourne VIC Australia.,Malignant Haematology and Stem Cell Transplantation Service Alfred Health Melbourne VIC Australia
| |
Collapse
|
29
|
Nonmalignant late cutaneous changes after allogeneic hematopoietic stem cell transplant in children. J Am Acad Dermatol 2018; 79:230-237. [DOI: 10.1016/j.jaad.2018.03.029] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2017] [Revised: 03/13/2018] [Accepted: 03/18/2018] [Indexed: 12/24/2022]
|
30
|
Long term survival among patients who are disease free at 1-year post allogeneic hematopoietic cell transplantation: a single center analysis of 389 consecutive patients. Bone Marrow Transplant 2018; 53:576-583. [DOI: 10.1038/s41409-017-0076-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 11/20/2017] [Accepted: 12/03/2017] [Indexed: 11/08/2022]
|
31
|
An aberrant NOTCH2-BCR signaling axis in B cells from patients with chronic GVHD. Blood 2017; 130:2131-2145. [PMID: 28851699 DOI: 10.1182/blood-2017-05-782466] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 08/24/2017] [Indexed: 12/16/2022] Open
Abstract
B-cell receptor (BCR)-activated B cells contribute to pathogenesis in chronic graft-versus-host disease (cGVHD), a condition manifested by both B-cell autoreactivity and immune deficiency. We hypothesized that constitutive BCR activation precluded functional B-cell maturation in cGVHD. To address this, we examined BCR-NOTCH2 synergy because NOTCH has been shown to increase BCR responsiveness in normal mouse B cells. We conducted ex vivo activation and signaling assays of 30 primary samples from hematopoietic stem cell transplantation patients with and without cGVHD. Consistent with a molecular link between pathways, we found that BCR-NOTCH activation significantly increased the proximal BCR adapter protein BLNK. BCR-NOTCH activation also enabled persistent NOTCH2 surface expression, suggesting a positive feedback loop. Specific NOTCH2 blockade eliminated NOTCH-BCR activation and significantly altered NOTCH downstream targets and B-cell maturation/effector molecules. Examination of the molecular underpinnings of this "NOTCH2-BCR axis" in cGVHD revealed imbalanced expression of the transcription factors IRF4 and IRF8, each critical to B-cell differentiation and fate. All-trans retinoic acid (ATRA) increased IRF4 expression, restored the IRF4-to-IRF8 ratio, abrogated BCR-NOTCH hyperactivation, and reduced NOTCH2 expression in cGVHD B cells without compromising viability. ATRA-treated cGVHD B cells had elevated TLR9 and PAX5, but not BLIMP1 (a gene-expression pattern associated with mature follicular B cells) and also attained increased cytosine guanine dinucleotide responsiveness. Together, we reveal a mechanistic link between NOTCH2 activation and robust BCR responses to otherwise suboptimal amounts of surrogate antigen. Our findings suggest that peripheral B cells in cGVHD patients can be pharmacologically directed from hyperactivation toward maturity.
Collapse
|
32
|
Abstract
The prevalence of autologous and allogeneic hematopoietic cell transplantation (HCT) survivors continues to increase. Among patients whose disease remains in remission for the first 2-5years after transplantation, it is estimated that approximately 80-90% will be alive over the subsequent 10years. However, the relative mortality rates of such patients continue to remain higher than those of their general population peers, with late complications contributing to significant long-term morbidity and mortality. Late effects in HCT survivors include secondary cancers, organ specific complications, late infections, quality of life impairments, psychosocial issues, sexual and fertility concerns, financial toxicity, and issues around return to work/school. A patient-centric and multidisciplinary approach to HCT survivorship care with collaborative and coordinated care from transplant centers and community healthcare providers is necessary to ensure their long-term health. Lifelong follow-up of HCT survivors is recommended, with established guidelines serving as the template for providing screening and preventive care based on patient-specific exposures. This review discussed common late complications, models for care delivery, and gaps and priorities for future research in the field of HCT survivorship.
Collapse
Affiliation(s)
- Navneet S Majhail
- Blood and Marrow Transplant Program, Cleveland Clinic, Cleveland, OH, USA.
| |
Collapse
|
33
|
Inamoto Y, Lee SJ. Late effects of blood and marrow transplantation. Haematologica 2017; 102:614-625. [PMID: 28232372 PMCID: PMC5395102 DOI: 10.3324/haematol.2016.150250] [Citation(s) in RCA: 103] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Accepted: 01/20/2017] [Indexed: 12/22/2022] Open
Abstract
Hematopoietic cell transplantation is a curative treatment for a variety of hematologic diseases. Advances in transplantation technology have reduced early transplant-related mortality and expanded application of transplantation to older patients and to a wider variety of diseases. Management of late effects after transplantation is increasingly important for a growing number of long-term survivors that is estimated to be half a million worldwide. Many studies have shown that transplant survivors suffer from significant late effects that adversely affect morbidity, mortality, working status and quality of life. Late effects include diseases of the cardiovascular, pulmonary, and endocrine systems, dysfunction of the thyroid gland, gonads, liver and kidneys, infertility, iron overload, bone diseases, infection, solid cancer, and neuropsychological effects. The leading causes of late mortality include recurrent malignancy, lung diseases, infection, secondary cancers and chronic graft-versus-host disease. The aim of this review is to facilitate better care of adult transplant survivors by summarizing accumulated evidence, new insights, and practical information about individual late effects. Further research is needed to understand the biology of late effects allowing better prevention and treatment strategies to be developed.
Collapse
Affiliation(s)
- Yoshihiro Inamoto
- Department of Hematopoietic Stem Cell Transplantation, National Cancer Center Hospital, Tokyo, Japan
| | - Stephanie J Lee
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| |
Collapse
|
34
|
Battiwalla M, Hashmi S, Majhail N, Pavletic S, Savani BN, Shelburne N. National Institutes of Health Hematopoietic Cell Transplantation Late Effects Initiative: Developing Recommendations to Improve Survivorship and Long-Term Outcomes. Biol Blood Marrow Transplant 2016; 23:6-9. [PMID: 27989931 DOI: 10.1016/j.bbmt.2016.10.020] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 10/27/2016] [Indexed: 01/02/2023]
Abstract
Continual advances in hematopoietic cell transplantation (HCT) have greatly improved early transplantation-related mortality and broadened the applicability of this intense but curative therapy. With growing success there is increasing awareness of late complications, occurring ≥1 year after treatment, and their associated morbidity and mortality in HCT survivors. These late effects occur with a wide spectrum in terms of latency, intensity, reversibility, and lethality. There is a need to understand the biology, surveillance, management, and patient experience of HCT-related effects, as well as the health care and research infrastructure to manage this growing population. To address these needs, the National Cancer Institute and National Heart, Lung and Blood Institute cosponsored a 12-month initiative to identify barriers and knowledge gaps and to formulate research and practice recommendations. Six major areas of interest were identified: research methodology and study design, subsequent neoplasms, patient-centered outcomes, immune dysregulation and pathobiology, cardiovascular disease and associated risk factors, and health care delivery. These findings were presented during the 2016 workshop and revised based on public response. This report provides an overview of the National Institutes of Health HCT Late Effects Initiative process and recommendations.
Collapse
Affiliation(s)
- Minoo Battiwalla
- Hematology Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland.
| | - Shahrukh Hashmi
- Mayo Clinic Transplant Center, Mayo Clinic, Rochester, Minnesota
| | - Navneet Majhail
- Blood and Marrow Transplant Program, Cleveland Clinic, Cleveland, Ohio
| | - Steven Pavletic
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Bipin N Savani
- Hematology & Stem Cell Transplantation Section, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Nonniekaye Shelburne
- Division of Cancer Control and Population Sciences, National Cancer Institute, National Institutes of Health, Rockville, Maryland
| |
Collapse
|