1
|
Dvorak CC, Long-Boyle JR, Holbrook-Brown L, Abdel-Azim H, Bertaina A, Vatsayan A, Talano JA, Bunin N, Anderson E, Flower A, Lalefar N, Higham CS, Kapoor N, Klein O, Odinakachukwu MC, Cho S, Jacobsohn DA, Collier W, Pulsipher MA. Effect of rabbit ATG PK on outcomes after TCR-αβ/CD19-depleted pediatric haploidentical HCT for hematologic malignancy. Blood Adv 2024; 8:6003-6014. [PMID: 39042892 PMCID: PMC11629297 DOI: 10.1182/bloodadvances.2024012670] [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: 01/16/2024] [Revised: 05/06/2024] [Accepted: 07/05/2024] [Indexed: 07/25/2024] Open
Abstract
ABSTRACT We hypothesized that the inferior disease-free survival (DFS) seen in older patients who underwent αβ-T-cell/CD19-depleted (AB-TCD) haploidentical hematopoietic cell transplantation (HCT) for hematologic malignancies is caused by excessive exposure to rabbit antithymocyte globulin (rATG; Thymoglobulin). Between 2015 and 2023, 163 patients with a median age of 13 years (range, 0.4-27.4) underwent AB-TCD haploidentical HCT for the treatment of acute lymphoblastic leukemia (n = 98), acute myeloid leukemia/myelodysplastic syndrome (n = 49), or other malignancies (n = 16) at 9 centers in 2 prospective trials. Exposures to rATG before and after HCT were predicted using a validated pharmacokinetic model. Receiver operating characteristic curves were used to identify the optimal target windows for rATG exposure in terms of outcomes. We identified 4 quadrants of rATG exposure, namely quadrant 1 (n = 52) with a high pre-HCT area under curve (AUC; ≥50 arbitrary units [AU] per day per milliliter) and a low post-HCT AUC (<12 AU per day per liter); quadrant 2 (n = 47) with a low pre- and post-HCT AUC; quadrant 3 (n = 13) with a low pre-HCT and a high post-HCT AUC; and quadrant 4 (n = 51) with a high pre- and post-HCT AUC. Quadrant 1 had a 3-year DFS of 86.5%, quadrant 2 had a DFS of 64.6%, quadrant 3 had a DFS of 32.9%, and for quadrant 4 it was 48.2%. An adjusted regression analysis demonstrated additional factors that were associated with an increased hazard for worse DFS, namely minimal residual disease (MRD) positivity and cytomegalovirus (CMV) R+/D- serostatus. Nonoptimal rATG exposure exhibited the strongest effect in unadjusted and adjusted (MRD status or CMV serostatus) analyses. High exposure to rATG after HCT was associated with inferior DFS following AB-TCD haploidentical HCT for pediatric patients with hematologic malignancies. Model-based dosing of rATG to achieve optimal exposure may improve DFS. These trials were registered at www.ClinicalTrials.gov as #NCT02646839 and #NCT04337515.
Collapse
Affiliation(s)
- Christopher C. Dvorak
- Division of Pediatric Allergy, Immunology, and Bone Marrow Transplantation, Department of Clinical Pharmacy, University of California San Francisco Benioff Children’s Hospitals, San Francisco, CA
| | - Janel R. Long-Boyle
- Division of Pediatric Allergy, Immunology, and Bone Marrow Transplantation, Department of Clinical Pharmacy, University of California San Francisco Benioff Children’s Hospitals, San Francisco, CA
| | - Lucia Holbrook-Brown
- Division of Hematology and Oncology, Section of Transplantation Intermountain Primary Children’s Hospital, Huntsman Cancer Institute, Spencer Fox Eccles School of Medicine at The University of Utah, Salt Lake City, UT
| | - Hisham Abdel-Azim
- Cancer Center, Children’s Hospital and Medical Center, Loma Linda University School of Medicine, Loma Linda, CA
| | - Alice Bertaina
- Division of Pediatric Hematology, Oncology, Stem Cell Transplantation & Regenerative Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA
| | - Anant Vatsayan
- Division of Blood and Marrow Transplantation, Children’s National Hospital, George Washington University School of Medicine and Health Sciences, Washington, DC
| | - Julie-An Talano
- Division of Pediatric Hematology, Oncology, and Blood and Marrow Transplant, Medical College of Wisconsin, Milwaukee, WI
| | - Nancy Bunin
- Division of Oncology, Children’s Hospital of Philadelphia, Philadelphia, PA
| | - Eric Anderson
- Division of Pediatric Hematology-Oncology, Rady Children’s Hospital, University of California San Diego, San Diego, CA
| | - Allyson Flower
- Division of Pediatric Hematology, Oncology, and Stem Cell Transplantation, New York Medical College, Valhalla, NY
| | - Nahal Lalefar
- Division of Hematology, University of California San Francisco Benioff Children’s Hospitals, Oakland, CA
| | - Christine S. Higham
- Division of Pediatric Allergy, Immunology, and Bone Marrow Transplantation, Department of Clinical Pharmacy, University of California San Francisco Benioff Children’s Hospitals, San Francisco, CA
| | - Neena Kapoor
- Division of Pediatric Hematology, Oncology, and Transplantation and Cellular Therapy, University of Southern California Children’s Hospital Los Angeles, Los Angeles, CA
| | - Orly Klein
- Division of Pediatric Hematology, Oncology, Stem Cell Transplantation & Regenerative Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA
| | - Maryanne C. Odinakachukwu
- Division of Blood and Marrow Transplantation, Children’s National Hospital, George Washington University School of Medicine and Health Sciences, Washington, DC
| | - Soohee Cho
- Division of Hematology and Oncology, Section of Transplantation Intermountain Primary Children’s Hospital, Huntsman Cancer Institute, Spencer Fox Eccles School of Medicine at The University of Utah, Salt Lake City, UT
| | - David A. Jacobsohn
- Division of Blood and Marrow Transplantation, Children’s National Hospital, George Washington University School of Medicine and Health Sciences, Washington, DC
| | - Willem Collier
- Division of Biostatistics, Department of Population and Public Health Sciences, University of Southern California, Los Angeles, CA
| | - Michael A. Pulsipher
- Division of Hematology and Oncology, Section of Transplantation Intermountain Primary Children’s Hospital, Huntsman Cancer Institute, Spencer Fox Eccles School of Medicine at The University of Utah, Salt Lake City, UT
| |
Collapse
|
2
|
Boelens JJ, Scordo M. Model-based dosing for better survival after transplantation. Blood Adv 2024; 8:6064-6066. [PMID: 39602148 PMCID: PMC11636124 DOI: 10.1182/bloodadvances.2024014236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2024] Open
Affiliation(s)
- Jaap Jan Boelens
- Department of Pediatrics, Transplantation and Cellular Therapies, MSK Kids, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Pediatrics, Weill Cornell Medical College, New York, NY
| | - Michael Scordo
- Department of Medicine, Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Medicine, Weill Cornell Medical College, New York, NY
| |
Collapse
|
3
|
Kelleher P, Greathead L, Whitby L, Brando B, Barnett D, Bloxham D, deTute R, Dunlop A, Farren T, Francis S, Payne D, Scott S, Snowden JA, Sorour Y, Stansfield E, Virgo P, Whitby A. European flow cytometry quality assurance guidelines for the diagnosis of primary immune deficiencies and assessment of immune reconstitution following B cell depletion therapies and transplantation. CYTOMETRY. PART B, CLINICAL CYTOMETRY 2024; 106:424-436. [PMID: 38940298 DOI: 10.1002/cyto.b.22195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 06/06/2024] [Accepted: 06/17/2024] [Indexed: 06/29/2024]
Abstract
Over the last 15 years activity of diagnostic flow cytometry services have evolved from monitoring of CD4 T cell subsets in HIV-1 infection to screening for primary and secondary immune deficiencies syndromes and assessment of immune constitution following B cell depleting therapy and transplantation. Changes in laboratory activity in high income countries have been driven by initiation of anti-retroviral therapy (ART) in HIV-1 regardless of CD4 T cell counts, increasing recognition of primary immune deficiency syndromes and the wider application of B cell depleting therapy and transplantation in clinical practice. Laboratories should use their experience in standardization and quality assurance of CD4 T cell counting in HIV-1 infection to provide immune monitoring services to patients with primary and secondary immune deficiencies. Assessment of immune reconstitution post B cell depleting agents and transplantation can also draw on the expertise acquired by flow cytometry laboratories for detection of CD34 stem cell and assessment of MRD in hematological malignancies. This guideline provides recommendations for clinical laboratories on providing flow cytometry services in screening for immune deficiencies and its emerging role immune reconstitution after B cell targeting therapies and transplantation.
Collapse
Affiliation(s)
- Peter Kelleher
- Immunology of Infection, Department of Infectious Disease, Imperial College London, London, UK
- Department of Infection and Immunity Sciences, North West London Pathology, London, UK
| | - Louise Greathead
- Department of Infection and Immunity Sciences, North West London Pathology, London, UK
| | - Liam Whitby
- UK NEQAS for Leucocyte Immunophenotyping, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
| | - Bruno Brando
- Hematology Laboratory and Transfusion Center, New Hospital of Legnano: Ospedale Nuovo di Legnano, Milan, Italy
| | - David Barnett
- UK NEQAS for Leucocyte Immunophenotyping, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
| | - David Bloxham
- Haematopathology and Oncology Diagnostic Service, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Ruth deTute
- Haematological Malignancy Diagnostic Service, St James's University Hospital, Leeds, UK
| | - Alan Dunlop
- Department of Haemato-Oncology, Royal Marsden Hospital, London, UK
| | - Timothy Farren
- Division of Haemato-Oncology, St Bartholomew's Hospital, Barts Health NHS Trust, London, UK
- Pathology Group, Blizard Institute, Queen Mary University of London, London, UK
| | - Sebastian Francis
- Department of Haematology, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
| | - Daniel Payne
- Tees Valley Pathology Service, James Cook University Hospital, Middlesbrough, UK
| | - Stuart Scott
- UK NEQAS for Leucocyte Immunophenotyping, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
| | - John A Snowden
- Department of Haematology, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
| | - Youssef Sorour
- Haematology, Doncaster and Bassetlaw Teaching Hospitals NHS Trust, Doncaster, UK
| | - Emma Stansfield
- Greater Manchester Immunology Service, Manchester University NHS Foundation Trust, Manchester, UK
| | - Paul Virgo
- Department of Immunology and Immunogenetics, North Bristol NHS Trust, Bristol, UK
| | - Alison Whitby
- UK NEQAS for Leucocyte Immunophenotyping, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
| |
Collapse
|
4
|
Barbarito G, Hiroshima L, Oppizzi L, Saini G, Kristovich K, Klein O, Hosszu K, Boehlke K, Gupta A, Mcavoy D, Shyr D, Boelens JJ, Bertaina A. Model-Based Antithymocyte Globulin in αβhaplo-Hematopoietic Stem Cell Transplantation Facilitates Engraftment, Expedites T Cell Recovery, and Mitigates the Risk of Acute Graft-versus-Host Disease. Transplant Cell Ther 2024; 30:810.e1-810.e16. [PMID: 38768907 DOI: 10.1016/j.jtct.2024.05.015] [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: 02/08/2024] [Revised: 05/06/2024] [Accepted: 05/13/2024] [Indexed: 05/22/2024]
Abstract
In αβ T-cell/CD19 B-cell depleted hematopoietic stem cell transplantation (αβhaplo-HSCT) recipients, antithymocyte globulin (ATG; Thymoglobulin) is used for preventing graft rejection and graft-versus-host disease (GVHD). The optimal dosing remains to be established, however. Here we present the first comparative analysis of 3 different ATG dosing strategies and their impact on immune reconstitution and GVHD. Our study aimed to evaluate the effects of 3 distinct dosing strategies of ATG on engraftment success, αβ+ and γδ+ T cell immune reconstitution, and the incidence and severity of acute GVHD in recipients of αβhaplo-HSCT. This comparative analysis included 3 cohorts of pediatric patients with malignant (n = 36) or nonmalignant (n = 8) disease. Cohorts 1 and 2 were given fixed ATG doses, whereas cohort 3 received doses via a new nomogram, based on absolute lymphocyte count (ALC) and body weight (BW). Cohort 3 showed a 0% incidence of day 100 grade II-IV acute GVHD, compared to 48% in cohort 1 and 27% in cohort 2. Furthermore, cohort 3 (the ALC/BW-based cohort) had a significant increase in CD4+ and CD8+ naïve T cells by day 90 (P = .04 and .03, respectively). Additionally, we found that the reconstitution and maturation of γδ+ T cells post-HSCT was not impacted across all 3 cohorts. Cumulative ATG exposure in all cohorts was lower than previously reported in T cell-replete settings, with a lower pre-HSCT exposure (<40 AU*day/mL) correlating with engraftment failure (P = .007). Conversely, a post-HSCT ATG exposure of 10 to 15 AU*day/mL was optimal for improving day 100 CD4+ (P = .058) and CD8+ (P = .03) immune reconstitution without increasing the risk of relapse or nonrelapse mortality. This study represents the first comparative analysis of ATG exposure in αβhaplo-HSCT recipients. Our findings indicate that (1) a 1- to 2-fold ATG to ATLG bioequivalence is more effective than previously established standards, and (2) ATG exposure post-HSCT does not adversely affect γδ+ T cell immune reconstitution. Furthermore, a model-based ATG dosing strategy effectively reduces graft rejection and day 100 acute GVHD while also promoting early CD4+/CD8+ immune reconstitution. These insights suggest that further optimization, including more distal administration of higher ATG doses within an ALC/BW-based strategy, will yield even greater improvements in outcomes.
Collapse
Affiliation(s)
- Giulia Barbarito
- Division of Hematology, Oncology, Stem Cell Transplantation, and Regenerative Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, California
| | - Lyndsie Hiroshima
- Division of Hematology, Oncology, Stem Cell Transplantation, and Regenerative Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, California
| | - Linda Oppizzi
- Division of Hematology, Oncology, Stem Cell Transplantation, and Regenerative Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, California
| | - Gopin Saini
- Division of Hematology, Oncology, Stem Cell Transplantation, and Regenerative Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, California
| | - Karen Kristovich
- Division of Hematology, Oncology, Stem Cell Transplantation, and Regenerative Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, California
| | - Orly Klein
- Division of Hematology, Oncology, Stem Cell Transplantation, and Regenerative Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, California
| | - Kinga Hosszu
- MSK Kids, Transplantation and Cellular Therapy Service, Memorial Sloan Kettering Cancer Center, New York, New York; Immune Discovery and Monitoring Service, Department of Pediatrics and Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Kylan Boehlke
- Division of Hematology, Oncology, Stem Cell Transplantation, and Regenerative Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, California
| | - Aditi Gupta
- Division of Hematology, Oncology, Stem Cell Transplantation, and Regenerative Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, California
| | - Devin Mcavoy
- MSK Kids, Transplantation and Cellular Therapy Service, Memorial Sloan Kettering Cancer Center, New York, New York; Immune Discovery and Monitoring Service, Department of Pediatrics and Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - David Shyr
- Division of Hematology, Oncology, Stem Cell Transplantation, and Regenerative Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, California
| | - Jaap Jan Boelens
- MSK Kids, Transplantation and Cellular Therapy Service, Memorial Sloan Kettering Cancer Center, New York, New York; Immune Discovery and Monitoring Service, Department of Pediatrics and Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Alice Bertaina
- Division of Hematology, Oncology, Stem Cell Transplantation, and Regenerative Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, California.
| |
Collapse
|
5
|
Naik S, Li Y, Talleur AC, Selukar S, Ashcraft E, Cheng C, Madden RM, Mamcarz E, Qudeimat A, Sharma A, Srinivasan A, Suliman AY, Epperly R, Obeng EA, Velasquez MP, Langfitt D, Schell S, Métais JY, Arnold PY, Hijano DR, Maron G, Merchant TE, Akel S, Leung W, Gottschalk S, Triplett BM. Memory T-cell enriched haploidentical transplantation with NK cell addback results in promising long-term outcomes: a phase II trial. J Hematol Oncol 2024; 17:50. [PMID: 38937803 PMCID: PMC11212178 DOI: 10.1186/s13045-024-01567-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 06/13/2024] [Indexed: 06/29/2024] Open
Abstract
BACKGROUND Relapse remains a challenge after transplantation in pediatric patients with hematological malignancies. Myeloablative regimens used for disease control are associated with acute and long-term adverse effects. We used a CD45RA-depleted haploidentical graft for adoptive transfer of memory T cells combined with NK-cell addback and hypothesized that maximizing the graft-versus-leukemia (GVL) effect might allow for reduction in intensity of conditioning regimen. METHODS In this phase II clinical trial (NCT01807611), 72 patients with hematological malignancies (complete remission (CR)1: 25, ≥ CR2: 28, refractory disease: 19) received haploidentical CD34 + enriched and CD45RA-depleted hematopoietic progenitor cell grafts followed by NK-cell infusion. Conditioning included fludarabine, thiotepa, melphalan, cyclophosphamide, total lymphoid irradiation, and graft-versus-host disease (GVHD) prophylaxis consisted of a short-course sirolimus or mycophenolate mofetil without serotherapy. RESULTS The 3-year overall survival (OS) and event-free-survival (EFS) for patients in CR1 were 92% (95% CI:72-98) and 88% (95% CI: 67-96); ≥ CR2 were 81% (95% CI: 61-92) and 68% (95% CI: 47-82) and refractory disease were 32% (95% CI: 11-54) and 20% (95% CI: 6-40). The 3-year EFS for all patients in morphological CR was 77% (95% CI: 64-87) with no difference amongst recipients with or without minimal residual disease (P = 0.2992). Immune reconstitution was rapid, with mean CD3 and CD4 T-cell counts of 410/μL and 140/μL at day + 30. Cumulative incidence of acute GVHD and chronic GVHD was 36% and 26% but most patients with acute GVHD recovered rapidly with therapy. Lower rates of grade III-IV acute GVHD were observed with NK-cell alloreactive donors (P = 0.004), and higher rates of moderate/severe chronic GVHD occurred with maternal donors (P = 0.035). CONCLUSION The combination of a CD45RA-depleted graft and NK-cell addback led to robust immune reconstitution maximizing the GVL effect and allowed for use of a submyeloablative, TBI-free conditioning regimen that was associated with excellent EFS resulting in promising long-term outcomes in this high-risk population. The trial is registered at ClinicalTrials.gov (NCT01807611).
Collapse
Affiliation(s)
- Swati Naik
- Department of Bone Marrow Transplantation & Cellular Therapy, St Jude Children's Research Hospital, Memphis, TN, USA.
| | - Ying Li
- Department of Bone Marrow Transplantation & Cellular Therapy, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Aimee C Talleur
- Department of Bone Marrow Transplantation & Cellular Therapy, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Subodh Selukar
- Department of Biostatistics, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Emily Ashcraft
- Department of Biostatistics, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Cheng Cheng
- Department of Biostatistics, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Renee M Madden
- Department of Bone Marrow Transplantation & Cellular Therapy, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Ewelina Mamcarz
- Department of Bone Marrow Transplantation & Cellular Therapy, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Amr Qudeimat
- Department of Bone Marrow Transplantation & Cellular Therapy, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Akshay Sharma
- Department of Bone Marrow Transplantation & Cellular Therapy, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Ashok Srinivasan
- Department of Bone Marrow Transplantation & Cellular Therapy, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Ali Y Suliman
- Department of Bone Marrow Transplantation & Cellular Therapy, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Rebecca Epperly
- Department of Bone Marrow Transplantation & Cellular Therapy, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Esther A Obeng
- Department of Bone Marrow Transplantation & Cellular Therapy, St Jude Children's Research Hospital, Memphis, TN, USA
| | - M Paulina Velasquez
- Department of Bone Marrow Transplantation & Cellular Therapy, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Deanna Langfitt
- Department of Bone Marrow Transplantation & Cellular Therapy, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Sarah Schell
- Department of Bone Marrow Transplantation & Cellular Therapy, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Jean-Yves Métais
- Department of Bone Marrow Transplantation & Cellular Therapy, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Paula Y Arnold
- Department of Pathology, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Diego R Hijano
- Department of Infectious Diseases, St Jude Children's Research Hospital, Memphis, TN, USA
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Gabriela Maron
- Department of Infectious Diseases, St Jude Children's Research Hospital, Memphis, TN, USA
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Thomas E Merchant
- Department of Radiation Oncology, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Salem Akel
- Department of Bone Marrow Transplantation & Cellular Therapy, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Wing Leung
- Department of Bone Marrow Transplantation & Cellular Therapy, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Stephen Gottschalk
- Department of Bone Marrow Transplantation & Cellular Therapy, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Brandon M Triplett
- Department of Bone Marrow Transplantation & Cellular Therapy, St Jude Children's Research Hospital, Memphis, TN, USA.
| |
Collapse
|
6
|
Admiraal R, Versluijs AB, Huitema ADR, Ebskamp L, Lacna A, de Kanter CTK, Bierings MB, Boelens JJ, Lindemans CA, Nierkens S. High-dose individualized antithymocyte globulin with therapeutic drug monitoring in high-risk cord blood transplant. Cytotherapy 2024; 26:599-605. [PMID: 38466262 DOI: 10.1016/j.jcyt.2024.02.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 01/25/2024] [Accepted: 02/13/2024] [Indexed: 03/12/2024]
Abstract
BACKGROUND Graft-versus-host disease (GvHD) and rejection are main limitations of cord blood transplantation (CBT), more so in patients with severe inflammation or previous rejections. While rigorous T-cell depletion with antithymocyte globulin (ATG) is needed to prevent GvHD and rejection, overexposure to ATG leads to slow T-cell recovery after transplantation, especially in CBT. OBJECTIVE To evaluate high-dose, upfront ATG with individualized dosing and therapeutic drug monitoring (TDM) in pediatric CBT for patients at high risk for GvHD and rejection. STUDY DESIGN Heavily inflamed patients and patients with a recent history of rejection were eligible for individualized high-dose ATG with real-time TDM. The ATG dosing scheme was adjusted to target a post-CBT exposure of <10 AU*day/mL, while achieving a pre-CBT exposure of 60-120 AU*day/mL; exposure levels previously defined for optimal efficacy and safety in terms of reduced GvHD and rejection, respectively. Main outcomes of interest included efficacy (target exposure attainment) and safety (incidence of GvHD and rejection). Other outcomes of interest included T-cell recovery and survival. RESULTS Twenty-one patients were included ranging from 2 months to 18 years old, receiving an actual median cumulative dose of ATG of 13.3 mg/kg (range 6-30 mg/kg) starting at a median 15 days (range 12-17) prior to CBT. Dosing was adjusted in 14 patients (increased in 3 and decreased in 11 patients). Eighteen (86%) and 19 (91%) patients reached the target pre-CBT and post-CBT exposure, respectively. Cumulative incidence for acute GvHD was 34% (95% CI 23-45) and 5% (95% CI 0-10%) for grade 2-4 and grade 3-4, respectively; cumulative incidence of rejection was 9% (95% CI 2-16%). Overall survival was 75% (95% CI 65-85%). CONCLUSION Individualized high-dose ATG with TDM is feasible and safe for patients with hyperinflammation in a CBT setting. We observe high target ATG exposure attainment, good immune reconstitution (despite very high doses of ATG) and acceptable rates of GvHD and rejection.
Collapse
Affiliation(s)
- Rick Admiraal
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands; Department of Pediatrics, University Medical Center Utrecht, Utrecht, the Netherlands.
| | - A Birgitta Versluijs
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands; Department of Pediatrics, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Alwin D R Huitema
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands; Department of Pharmacy & Pharmacology, Antoni van Leeuwenhoek Hospital, Netherlands Cancer Institute, Amsterdam, the Netherlands; Department of Clinical Pharmacy, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Lysette Ebskamp
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Amelia Lacna
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - C T Klaartje de Kanter
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands; Department of Pharmacy, Curacao Medical Center, Willemstad, Curacao
| | - Marc B Bierings
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands; Department of Pediatrics, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Jaap Jan Boelens
- Transplantation and Cellular Therapies, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Caroline A Lindemans
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands; Department of Pediatrics, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Stefan Nierkens
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands; Center for Translational Immunology, University Medical Center Utrecht, Utrecht, the Netherlands
| |
Collapse
|
7
|
Okamoto S, Perales MA, Sureda A, Urueta AK. Fostering next generation transplant physicians. BLOOD CELL THERAPY 2024; 7:56-63. [PMID: 38854400 PMCID: PMC11153206 DOI: 10.31547/bct-2024-004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 03/04/2024] [Indexed: 06/11/2024]
Abstract
As opposed to the rapid expansion of hematopoietic cell transplantation (HCT) and other cellular therapies (CT), we are now facing a global shortage of transplant physicians and other professionals to support the activity of HCT/CT. To overcome this obstacle, a variety of approaches are now being undertaken in four international HCT societies. This article aims to share their current attempts to foster the next generation of transplant physicians and allied professionals needed to secure the continued global growth of HCT/CT.
Collapse
Affiliation(s)
- Shinichiro Okamoto
- Division of Hematology, Department of Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Miguel-Angel Perales
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, USA
- Department of Medicine, Weill Cornell Medical College, New York, USA
| | - Anna Sureda
- Clinical Hematology Department, Institut Català d'Oncologia - L'Hospitalet, IDIBELL, Universitat de Barcelona, Barcelona, Spain
| | | |
Collapse
|
8
|
Lakkaraja M, Mauguen A, Boulad F, Cancio MI, Curran KJ, Harris AC, Kernan NA, Klein E, Kung AL, Oved J, Prockop S, Scaradavou A, Spitzer B, O'Reilly RJ, Boelens JJ. Impact of rabbit anti-thymocyte globulin (ATG) exposure on outcomes after ex vivo T-cell-depleted hematopoietic cell transplantation in pediatric and young adult patients. Cytotherapy 2024; 26:351-359. [PMID: 38349310 PMCID: PMC10997457 DOI: 10.1016/j.jcyt.2024.01.004] [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: 02/23/2023] [Revised: 11/03/2023] [Accepted: 01/24/2024] [Indexed: 04/07/2024]
Abstract
BACKGROUND AIMS Traditional weight-based dosing of rabbit anti-thymocyte globulin (rATG) used in allogeneic hematopoietic cell transplantation (HCT) to prevent graft-versus-host disease (GVHD) and graft rejection leads to variable exposures. High exposures induce delayed CD4+immune reconstitution (CD4+IR) and greater mortality. We sought to determine the impact of rATG exposure in children and young adults receiving various types of EX-VIVO T-cell-depleted (EX-VIVO-TCD) HCT. METHODS Patients receiving their first EX-VIVO-TCD HCT (CliniMACS CD34+, Isolex or soybean lectin agglutination), with removal of residual T cells by E-rosette depletion (E-) between 2008 and 2018 at Memorial Sloan Kettering Cancer Center were retrospectively analyzed. rATG exposure post-HCT was estimated (AU*d/L) using a validated population pharmacokinetic model. Previously defined rATG-exposures, <30, 30-55, ≥55 AU*d/L, were related with outcomes of interest. Cox proportional hazard and cause-specific models were used for analyses. RESULTS In total, 180 patients (median age 11 years; range 0.1-44 years) were included, malignant 124 (69%) and nonmalignant 56 (31%). Median post-HCT rATG exposure was 32 (0-104) AU*d/L. Exposure <30 AU*d/L was associated with a 3-fold greater probability of CD4+IR (P < 0.001); 2- to 4-fold lower risk of death (P = 0.002); and 3- to 4-fold lower risk of non-relapse mortality (NRM) (P = 0.02). Cumulative incidence of NRM was 8-fold lower in patients who attained CD4+IR compared with those who did not (P < 0.0001). There was no relation between rATG exposure and aGVHD (P = 0.33) or relapse (P = 0.23). Effect of rATG exposure on outcomes was similar in three EX-VIVO-TCD methods. CONCLUSIONS Individualizing rATG dosing to target a low rATG exposure post-HCT while maintaining total cumulative exposure may better predict CD4+IR, reduce NRM and increase overall survival, independent of the EX-VIVO-TCD method.
Collapse
Affiliation(s)
- Madhavi Lakkaraja
- Fred Hutchinson Cancer Center, Seattle, Washington, USA; Department of Pediatrics, University of Washington School of Medicine, Seattle, Washington, USA
| | - Audrey Mauguen
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Farid Boulad
- Department of Pediatrics, BMT Service, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Maria I Cancio
- Department of Pediatrics, BMT Service, Memorial Sloan Kettering Cancer Center, New York, New York, USA; Department of Pediatrics, Weill Cornell Medicine, New York, New York, USA
| | - Kevin J Curran
- Department of Pediatrics, BMT Service, Memorial Sloan Kettering Cancer Center, New York, New York, USA; Department of Pediatrics, Weill Cornell Medicine, New York, New York, USA
| | - Andrew C Harris
- Department of Pediatrics, BMT Service, Memorial Sloan Kettering Cancer Center, New York, New York, USA; Department of Pediatrics, Weill Cornell Medicine, New York, New York, USA
| | - Nancy A Kernan
- Department of Pediatrics, BMT Service, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Elizabeth Klein
- Department of Pediatrics, BMT Service, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Andrew L Kung
- Department of Pediatrics, BMT Service, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Joseph Oved
- Department of Pediatrics, BMT Service, Memorial Sloan Kettering Cancer Center, New York, New York, USA; Department of Pediatrics, Weill Cornell Medicine, New York, New York, USA
| | - Susan Prockop
- Dana Farber Cancer Institute, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Andromachi Scaradavou
- Department of Pediatrics, BMT Service, Memorial Sloan Kettering Cancer Center, New York, New York, USA; Department of Pediatrics, Weill Cornell Medicine, New York, New York, USA
| | - Barbara Spitzer
- Department of Pediatrics, BMT Service, Memorial Sloan Kettering Cancer Center, New York, New York, USA; Department of Pediatrics, Weill Cornell Medicine, New York, New York, USA
| | - Richard J O'Reilly
- Department of Pediatrics, BMT Service, Memorial Sloan Kettering Cancer Center, New York, New York, USA; Department of Pediatrics, Weill Cornell Medicine, New York, New York, USA
| | - Jaap Jan Boelens
- Department of Pediatrics, BMT Service, Memorial Sloan Kettering Cancer Center, New York, New York, USA; Department of Pediatrics, Weill Cornell Medicine, New York, New York, USA.
| |
Collapse
|
9
|
Barriga F, Wietstruck A, Schulze-Schiappacasse C, Catalán P, Sotomayor C, Zúñiga P, Aguirre N, Vizcaya C, Le Corre N, Villarroel L. Individualized dose of anti-thymocyte globulin based on weight and pre-transplantation lymphocyte counts in pediatric patients: a single center experience. Bone Marrow Transplant 2024; 59:473-478. [PMID: 38253868 DOI: 10.1038/s41409-024-02206-5] [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: 05/15/2023] [Revised: 12/28/2023] [Accepted: 01/10/2024] [Indexed: 01/24/2024]
Abstract
Anti-thymocyte globulin (ATG) has become a standard in preventing GVHD in related and unrelated donor transplantation, but there is no consensus on the best administration schedule. The PARACHUTE trial reported excellent CD4 immune reconstitution (CD4 IR) using a dosing schedule based on the patient's weight and pre-conditioning absolute lymphocyte count (ALC). In 2015 we introduced the PARACHUTE dosing schedule for pediatric patients at our center. One hundred one patients were transplanted for malignant and non-malignant diseases. In this non-concurrent cohort CD4 IR+, defined by a single CD4 count >50/µL on day 90, was seen in 81% of patients. The incidence of grade II-IV and III to IV aGvHD was 26.6% and 15.3% and 5% for cGvHD with no severe cases. We found no difference in aGvHD between donor type and stem cell sources. Five-year EFS and OS were 77.5% and 83.5%. Grade III-IV GFRS was 75.2%. CD4 IR+ patients had better EFS (93.1% vs. 77.7%, p = 0.04) and lower non-relapse mortality (2.7% vs. 22.2%, p = 0.002). The PARACHUTE ATG dosing schedule individualized by weight and ALC results in good early immune reconstitution, low incidence of cGvHD, and favorable survival for patients with different disease groups, donor types, and stem cell sources.
Collapse
Affiliation(s)
- Francisco Barriga
- Section of Hematology, Oncology and Stem Cell Transplantation. Division of Pediatrics, Pontificia Universidad Católica de Chile, Santiago, Chile.
| | - Angelica Wietstruck
- Section of Hematology, Oncology and Stem Cell Transplantation. Division of Pediatrics, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Clara Schulze-Schiappacasse
- Department of Pediatric Infectious Diseases, Division of Pediatrics, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Paula Catalán
- Section of Hematology, Oncology and Stem Cell Transplantation. Division of Pediatrics, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Cristian Sotomayor
- Section of Hematology, Oncology and Stem Cell Transplantation. Division of Pediatrics, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Pamela Zúñiga
- Section of Hematology, Oncology and Stem Cell Transplantation. Division of Pediatrics, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Noemi Aguirre
- Section of Hematology, Oncology and Stem Cell Transplantation. Division of Pediatrics, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Cecilia Vizcaya
- Department of Pediatric Infectious Diseases, Division of Pediatrics, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Nicole Le Corre
- Department of Pediatric Infectious Diseases, Division of Pediatrics, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Luis Villarroel
- Department of Public Health, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
10
|
Chiu YH, Drijver A, Admiraal R, van Rhenen A, Nierkens S, van Laar JM, Spierings J. Anti-thymocyte globulin exposure in patients with diffuse cutaneous systemic sclerosis undergoing autologous haematopoietic stem cell transplantation. JOURNAL OF SCLERODERMA AND RELATED DISORDERS 2023; 8:241-246. [PMID: 37744043 PMCID: PMC10515999 DOI: 10.1177/23971983231188232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 06/28/2023] [Indexed: 09/26/2023]
Abstract
Introduction Autologous haematopoietic stem cell transplantation improves event-free survival and lung function and reduces skin thickening in patients with progressive diffuse cutaneous systemic sclerosis. Anti-thymocyte globulin is a key lymphoablative constituent of conditioning protocols and is administered in a weight-based dosage. However, whether anti-thymocyte globulin exposure contributes to response to autologous haematopoietic stem cell transplantation and lymphocyte reconstitution in diffuse cutaneous systemic sclerosis patients is unknown. We aimed to explore the relationship between anti-thymocyte globulin exposure, lymphocyte reconstitution and treatment response in diffuse cutaneous systemic sclerosis patients undergoing autologous haematopoietic stem cell transplantation. Methods A retrospective cohort of 15 diffuse cutaneous systemic sclerosis patients undergoing autologous haematopoietic stem cell transplantation was performed. Clinical characteristics and routine laboratory results were retrieved from electronic medical records. Anti-thymocyte globulin concentrations were measured in cryopreserved plasma samples at four time points (day 1 and week 1, 2 and 4) after stem cell reinfusion. Anti-thymocyte globulin exposure was estimated using a validated population pharmacokinetic model. Results During a median follow-up of 45 months (interquartile range 19-66), 11 (73%) patients had a treatment response, and 4 (27%) were non-responders. Although all patients received the same weight-based anti-thymocyte globulin dosage, 7.5 mg/kg divided over 3 days, anti-thymocyte globulin exposure varied. Anti-thymocyte globulin exposure was higher in responders than in non-responders (163 AU*day/mL (interquartile range 153-183) and 137 AU*day/mL (interquartile range 101-149), respectively, p = .026). Anti-thymocyte globulin exposure was not correlated with lymphocyte reconstitution or infection rate. Conclusion Weight-based dosing of anti-thymocyte globulin results in variable anti-thymocyte globulin exposure and treatment response across individuals.
Collapse
Affiliation(s)
- Yu-Hsiang Chiu
- Department of Rheumatology and Clinical Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
- Division of Rheumatology/Immunology/Allergy, Department of Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei
| | - Anouk Drijver
- Department of Rheumatology and Clinical Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Rick Admiraal
- Princess Máxima Center for Paediatric Oncology, Utrecht, The Netherlands
| | - Anna van Rhenen
- Department of Haematology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Stefan Nierkens
- Princess Máxima Center for Paediatric Oncology, Utrecht, The Netherlands
- Centre for Translational Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Jacob M van Laar
- Department of Rheumatology and Clinical Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Julia Spierings
- Department of Rheumatology and Clinical Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
11
|
Tamari R, Scordo M, Kunvarjee BM, Proli A, Lin A, Flynn J, Cho C, Devlin S, Klein E, Boulad F, Cancio MI, Curran KJ, Jakubowski AA, Kernan NA, Kung AL, O’Reilly RJ, Papadopoulos EB, Prockop S, Scaradavou A, Shaffer BC, Shah G, Spitzer B, Gyurkocza B, Giralt SA, Perales MA, Boelens JJ. Association between busulfan exposure and survival in patients undergoing a CD34+ selected stem cell transplantation. Blood Adv 2023; 7:5225-5233. [PMID: 37379285 PMCID: PMC10500467 DOI: 10.1182/bloodadvances.2023009708] [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: 01/19/2023] [Revised: 06/12/2023] [Accepted: 06/12/2023] [Indexed: 06/30/2023] Open
Abstract
Busulfan is an alkylating drug routinely used in conditioning regimens for allogeneic hematopoietic cell transplantation (allo-HCT). A myeloablative conditioning regimen, including busulfan, is commonly used in patients undergoing T-cell depletion (TCD) and allo-HCT, but data on optimal busulfan pharmacokinetic (PK) exposure in this setting are limited. Between 2012 and 2019, busulfan PK was performed to target an area under the curve exposure between 55 and 66 mg × h/L over 3 days using a noncompartmental analysis model. We retrospectively re-estimated busulfan exposure following the published population PK (popPK) model (2021) and correlated it with outcomes. To define optimal exposure, univariable models were performed with P splines, wherein hazard ratio (HR) plots were drawn, and thresholds were found graphically as the points at which the confidence interval crossed 1. Cox proportional hazard and competing risk models were used for analyses. 176 patients were included, with a median age of 59 years (range, 2-71). Using the popPK model, the median cumulative busulfan exposure was 63.4 mg × h/L (range, 46.3-90.7). The optimal threshold was at the upper limit of the lowest quartile (59.5 mg × h/L). 5-year overall survival (OS) with busulfan exposure ≥59.5 vs <59.5 mg × h/L was 67% (95% CI, 59-76) vs 40% (95% CI, 53-68), respectively (P = .02), and this association remained in a multivariate analyses (HR, 0.5; 95% CI, 0.29; 0.88; P = .02). In patients undergoing TCD allo-HCT, busulfan exposure is significantly associated with OS. The use of a published popPK model to optimize exposure may significantly improve the OS.
Collapse
Affiliation(s)
- Roni Tamari
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Medicine, Weill Cornell Medical College, New York, NY
| | - Michael Scordo
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Medicine, Weill Cornell Medical College, New York, NY
| | - Binni M. Kunvarjee
- Department of Pharmacy, Memorial Sloan Kettering Cancer Center, New York, NY
| | | | - Andrew Lin
- Department of Pharmacy, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Jessica Flynn
- Department of Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Christina Cho
- Stem Cell Transplantion and Cellular Therapy Program, John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ
| | - Sean Devlin
- Department of Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Elizabeth Klein
- Department of Pediatrics, Stem Cell Transplantation and Cellular Therapies Service, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Farid Boulad
- Department of Pediatrics, Stem Cell Transplantation and Cellular Therapies Service, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Pediatrics, Weill Cornell Medicine, New York, NY
| | - Maria I. Cancio
- Department of Pediatrics, Stem Cell Transplantation and Cellular Therapies Service, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Pediatrics, Weill Cornell Medicine, New York, NY
| | - Kevin J. Curran
- Department of Pediatrics, Stem Cell Transplantation and Cellular Therapies Service, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Pediatrics, Weill Cornell Medicine, New York, NY
| | - Ann A. Jakubowski
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Medicine, Weill Cornell Medical College, New York, NY
| | - Nancy A. Kernan
- Department of Pediatrics, Stem Cell Transplantation and Cellular Therapies Service, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Pediatrics, Weill Cornell Medicine, New York, NY
| | - Andrew L. Kung
- Department of Pediatrics, Stem Cell Transplantation and Cellular Therapies Service, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Pediatrics, Weill Cornell Medicine, New York, NY
| | - Richard J. O’Reilly
- Department of Pediatrics, Stem Cell Transplantation and Cellular Therapies Service, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Pediatrics, Weill Cornell Medicine, New York, NY
| | - Esperanza B. Papadopoulos
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Medicine, Weill Cornell Medical College, New York, NY
| | - Susan Prockop
- Department of Pediatrics, Boston Children’s Hospital and Dana Farber Cancer Institute, Boston, MA
| | - Andromachi Scaradavou
- Department of Pediatrics, Stem Cell Transplantation and Cellular Therapies Service, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Pediatrics, Weill Cornell Medicine, New York, NY
| | - Brian C. Shaffer
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Medicine, Weill Cornell Medical College, New York, NY
| | - Gunjan Shah
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Medicine, Weill Cornell Medical College, New York, NY
| | - Barbara Spitzer
- Department of Pediatrics, Stem Cell Transplantation and Cellular Therapies Service, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Pediatrics, Weill Cornell Medicine, New York, NY
| | - Boglarka Gyurkocza
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Medicine, Weill Cornell Medical College, New York, NY
| | - Sergio A. Giralt
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Medicine, Weill Cornell Medical College, New York, NY
| | - Miguel-Angel Perales
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Medicine, Weill Cornell Medical College, New York, NY
| | - Jaap Jan Boelens
- Department of Pediatrics, Stem Cell Transplantation and Cellular Therapies Service, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Pediatrics, Weill Cornell Medicine, New York, NY
| |
Collapse
|
12
|
Troullioud Lucas AG, Lindemans CA, Bhoopalan SV, Dandis R, Prockop SE, Naik S, Keerthi D, de Koning C, Sharma A, Nierkens S, Boelens JJ. Early immune reconstitution as predictor for outcomes after allogeneic hematopoietic cell transplant; a tri-institutional analysis. Cytotherapy 2023; 25:977-985. [PMID: 37330731 PMCID: PMC10984694 DOI: 10.1016/j.jcyt.2023.05.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 04/19/2023] [Accepted: 05/26/2023] [Indexed: 06/19/2023]
Abstract
BACKGROUND AIMS CD4 immune reconstitution (IR) after allogeneic hematopoietic cell transplant (allo-HCT) correlates with lower non-relapse mortality (NRM), but its impact on leukemia relapse remains less clear, especially in children. We studied the correlation between IR of lymphocyte subsets and HCT outcomes in a large cohort of children/young adults with hematological malignancies. METHODS We retrospectively analyzed CD4, CD8, B-cell and natural killer (NK) cell reconstitution in patients after first allo-HCT for a hematological malignancy at three large academic institutions (n = 503; period 2008-2019). We used Cox proportional hazard and Fine-Gray competing risk models, martingale residual plots and maximally selected log-rank statistics to assess the impact of IR on outcomes. RESULTS Achieving CD4 >50 and/or B cells >25 cells/μL before day 100 after allo-HCT was a predictor of lower NRM (CD4 IR: hazard ratio [HR] 0.26, 95% confidence interval [CI] 0.11-0.62, P = 0.002; CD4 and B cell IR: HR 0.06, 95% CI 0.03-0.16, P < 0.001), acute graft-versus-host disease (GVHD) (CD4 and B cell IR: HR 0.02, 95% CI 0.01-0.04, P < 0.001) and chronic GVHD (CD4 and B cell IR: HR 0.16, 95% CI 0.05-0.49, P = 0.001) in the full cohort, and of lower risk of relapse (CD4 and B cell IR: HR 0.24, 95% CI 0.06-0.92, P = 0.038) in the acute myeloid leukemia subgroup. No correlation between CD8 and NK-cell IR and relapse or NRM was found. CONCLUSIONS CD4 and B-cell IR was associated with clinically significant lower NRM, GVHD and, in patients with acute myeloid leukemia, disease relapse. CD8 and NK-cell IR was neither associated with relapse nor NRM. If confirmed in other cohorts, these results can be easily implemented for risk stratification and clinical decision making.
Collapse
Affiliation(s)
- Alexandre G Troullioud Lucas
- Transplantation and Cellular Therapy, MSK Kids, Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA; Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Caroline A Lindemans
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands; Department of Pediatrics, University Medical Center Utrecht, Utrecht, the Netherlands
| | | | - Rana Dandis
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Susan E Prockop
- Pediatric Stem Cell Transplantation, Boston Children's Hospital/Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Swati Naik
- Department of Bone Marrow Transplantation and Cell Therapy, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Dinesh Keerthi
- Department of Bone Marrow Transplantation and Cell Therapy, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Coco de Koning
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Akshay Sharma
- Department of Bone Marrow Transplantation and Cell Therapy, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Stefan Nierkens
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands; Center for Translational Immunology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Jaap Jan Boelens
- Transplantation and Cellular Therapy, MSK Kids, Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA.
| |
Collapse
|
13
|
Zelikson V, Sabo R, Serrano M, Aqeel Y, Ward S, Al Juhaishi T, Aziz M, Krieger E, Simmons G, Roberts C, Reed J, Buck G, Toor A. Allogeneic haematopoietic cell transplants as dynamical systems: influence of early-term immune milieu on long-term T-cell recovery. Clin Transl Immunology 2023; 12:e1458. [PMID: 37457614 PMCID: PMC10345185 DOI: 10.1002/cti2.1458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 01/11/2023] [Accepted: 06/26/2023] [Indexed: 07/18/2023] Open
Abstract
Objectives Immune recovery following haematopoietic cell transplantation (HCT) functions as a dynamical system. Reducing the duration of intense immune suppression and augmenting antigen presentation has the potential to optimise T-cell reconstitution, potentially influencing long-term outcomes. Methods Based on donor-derived T-cell recovery, 26 patients were adaptively randomised between mycophenolate mofetil (MMF) administered for 30-day post-transplant with filgrastim for cytokine support (MMF30 arm, N = 11), or MMF given for 15 days with sargramostim (MMF15 arm, N = 15). All patients underwent in vivo T-cell depletion with 5.1 mg kg-1 antithymocyte globulin (administered over 3 days, Day -9 through to Day -7) and received reduced intensity 450 cGy total body irradiation (3 fractions on Day -1 and Day 0). Patients underwent HLA-matched related and unrelated donor haematopoietic cell transplantation (HCT). Results Clinical outcomes were equivalent between the two groups. The MMF15 arm demonstrated superior T-cell, as well as T-cell subset recovery and a trend towards superior T-cell receptor (TCR) diversity in the first month with this difference persisting through the first year. T-cell repertoire recovery was more rapid and sustained, as well as more diverse in the MMF15 arm. Conclusion The long-term superior immune recovery in the MMF15 arm, administered GMCSF, is consistent with a disproportionate impact of early interventions in HCT. Modifying the 'immune-milieu' following allogeneic HCT is feasible and may influence long-term T-cell recovery.
Collapse
Affiliation(s)
- Viktoriya Zelikson
- Department of Internal MedicineVirginia Commonwealth UniversityRichmondVAUSA
| | - Roy Sabo
- Department of BiostatisticsVirginia Commonwealth UniversityRichmondVAUSA
| | - Myrna Serrano
- Department of Microbiology and ImmunologyVirginia Commonwealth UniversityRichmondVAUSA
| | - Younus Aqeel
- Department of Internal MedicineVirginia Commonwealth UniversityRichmondVAUSA
| | - Savannah Ward
- Department of Internal MedicineVirginia Commonwealth UniversityRichmondVAUSA
| | - Taha Al Juhaishi
- Department of Internal MedicineVirginia Commonwealth UniversityRichmondVAUSA
| | - May Aziz
- Department of PharmacyVirginia Commonwealth UniversityRichmondVAUSA
| | - Elizabeth Krieger
- Department of PediatricsVirginia Commonwealth UniversityRichmondVAUSA
| | - Gary Simmons
- Department of Internal MedicineVirginia Commonwealth UniversityRichmondVAUSA
| | - Catherine Roberts
- Department of Internal MedicineVirginia Commonwealth UniversityRichmondVAUSA
| | - Jason Reed
- Department of PhysicsVirginia Commonwealth UniversityRichmondVAUSA
| | - Gregory Buck
- Department of BiostatisticsVirginia Commonwealth UniversityRichmondVAUSA
| | - Amir Toor
- Department of Internal MedicineVirginia Commonwealth UniversityRichmondVAUSA
- Lehigh Valley Topper Cancer InstituteAllentownPAUSA
| |
Collapse
|
14
|
Dadwal SS, Papanicolaou GA, Boeckh M. How I prevent viral reactivation in high-risk patients. Blood 2023; 141:2062-2074. [PMID: 36493341 PMCID: PMC10163320 DOI: 10.1182/blood.2021014676] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 11/10/2022] [Accepted: 11/22/2022] [Indexed: 12/14/2022] Open
Abstract
Preventing viral infections at an early stage is a key strategy for successfully improving transplant outcomes. Preemptive therapy and prophylaxis with antiviral agents have been successfully used to prevent clinically significant viral infections in hematopoietic cell transplant recipients. Major progress has been made over the past decades in preventing viral infections through a better understanding of the biology and risk factors, as well as the introduction of novel antiviral agents and advances in immunotherapy. High-quality evidence exists for the effective prevention of herpes simplex virus, varicella-zoster virus, and cytomegalovirus infection and disease. Few data are available on the effective prevention of human herpesvirus 6, Epstein-Barr virus, adenovirus, and BK virus infections. To highlight the spectrum of clinical practice, here we review high-risk situations that we handle with a high degree of uniformity and cases that feature differences in approaches, reflecting distinct hematopoietic cell transplant practices, such as ex vivo T-cell depletion.
Collapse
Affiliation(s)
- Sanjeet S. Dadwal
- Division of Infectious Disease, Department of Medicine, City of Hope National Medical Center, Duarte, CA
| | - Genovefa A. Papanicolaou
- Infectious Disease Service, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Medicine, Weill Cornell Medical College, Cornell University, New York, NY
| | - Michael Boeckh
- Vaccine and Infectious and Clinical Research Divisions, Fred Hutchinson Cancer Center, Seattle, WA
- Division of Allergy and Infectious Diseases, Department of Medicine, University of Washington, Seattle, WA
| |
Collapse
|
15
|
Wolfe AE, Markey KA. The contribution of the intestinal microbiome to immune recovery after HCT. Front Immunol 2022; 13:988121. [PMID: 36059482 PMCID: PMC9434312 DOI: 10.3389/fimmu.2022.988121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 07/27/2022] [Indexed: 11/13/2022] Open
Abstract
Allogenic hematopoietic stem-cell transplantation (allo-HCT) is a curative-intent immunotherapy for high-risk hematological malignancies and immune deficiencies. Allo-HCT carries a high risk of treatment-related mortality (TRM), largely due to infection or graft-versus-host disease (GVHD). Robust immune recovery is essential for optimal patient outcomes, given the immunologic graft-versus-leukemia effect prevents relapse, and functional innate and adaptive immunity are both needed for the prevention and control of infection. Most simply, we measure immune recovery by enumerating donor lymphocyte subsets in circulation. In functional terms, ideal immune recovery is more difficult to define, and current lab techniques are limited to the measurement of specific vaccine-responses or mitogens ex vivo. Clinically, poor immune function manifests as problematic infection with viral, bacterial and fungal organisms. Furthermore, the ideal recovering immune system is capable of exerting graft-versus-tumor effects to prevent relapse, and does not induce graft-versus-host disease. Large clinical observational studies have linked loss of diversity within the gut microbiome with adverse transplant outcomes including decreased overall survival and increased acute and chronic GVHD. Furthermore, the correlation between intestinal microbial communities and numeric lymphocyte recovery has now been reported using a number of approaches. Large sets of clinically available white blood cell count data, clinical flow cytometry of lymphocyte subsets and bespoke flow cytometry analyses designed to capture microbiota-specific T cells (e.g. Mucosal-associated invariant T cells, subsets of the gd T cells) have all been leveraged in an attempt to understand links between the microbiota and the recovering immune system in HCT patients. Additionally, preclinical studies suggest an immunomodulatory role for bacterial metabolites (including butyrate, secondary bile acids, and indole derivatives from tryptophan metabolism) in transplant outcomes, though further studies are needed to unravel mechanisms relevant to the post-HCT setting. An understanding of mechanistic relationships between the intestinal microbiome and post-transplant outcomes is necessary for reduction of risk associated with transplant, to inform prophylactic procedures, and ensure optimal immune reconstitution without alloreactivity. Here, we summarize the current understanding of the complex relationship between bacterial communities, their individual members, and the metabolites they produce with immune function in both the allo-HCT and steady-state setting.
Collapse
Affiliation(s)
- Alex E. Wolfe
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, United States
| | - Kate A. Markey
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, United States
- Division of Medical Oncology, University of Washington, Seattle, WA, United States
| |
Collapse
|
16
|
Takahashi T, Prockop SE. T-cell depleted haploidentical hematopoietic cell transplantation for pediatric malignancy. Front Pediatr 2022; 10:987220. [PMID: 36313879 PMCID: PMC9614427 DOI: 10.3389/fped.2022.987220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 09/20/2022] [Indexed: 11/13/2022] Open
Abstract
Access to allogenic hematopoietic cell transplantation (HCT), a potentially curative treatment for chemotherapy-resistant hematologic malignancies, can be limited if no human leukocyte antigen (HLA) identical related or unrelated donor is available. Alternative donors include Cord Blood as well as HLA-mismatched unrelated or related donors. If the goal is to minimize the number of HLA disparities, partially matched unrelated donors are more likely to share 8 or 9 of 10 HLA alleles with the recipient. However, over the last decade, there has been success with haploidentical HCT performed using the stem cells from HLA half-matched related donors. As the majority of patients have at least one eligible and motivated haploidentical donor, recruitment of haploidentical related donors is frequently more rapid than of unrelated donors. This advantage in the accessibility has historically been offset by the increased risks of graft rejection, graft-versus-host disease and delayed immune reconstitution. Various ex vivo T-cell depletion (TCD) methods have been investigated to overcome the immunological barrier and facilitate immune reconstitution after a haploidentical HCT. This review summarizes historical and contemporary clinical trials of haploidentical TCD-HCT, mainly in pediatric malignancy, and describes the evolution of these approaches with a focus on serial improvements in the kinetics of immune reconstitution. Methods of TCD discussed include in vivo as well as ex vivo positive and negative selection. In addition, haploidentical TCD as a platform for post-HCT cellular therapies is discussed. The present review highlights that, as a result of the remarkable progress over half a century, haploidentical TCD-HCT can now be considered as a preferred alternative donor option for children with hematological malignancy in need of allogeneic HCT.
Collapse
Affiliation(s)
- Takuto Takahashi
- Pediatric Stem Cell Transplantation, Boston Children's Hospital/Dana-Farber Cancer Institute, Boston, MA, United States.,Department of Experimental and Clinical Pharmacology, University of Minnesota College of Pharmacy, Minneapolis, MN, United States
| | - Susan E Prockop
- Pediatric Stem Cell Transplantation, Boston Children's Hospital/Dana-Farber Cancer Institute, Boston, MA, United States
| |
Collapse
|