Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Blood ; 140(25): 2684-2696, 2022 12 22.
Article in English | MEDLINE | ID: mdl-35914226

ABSTRACT

Chimeric antigen receptor (CAR) T-cell therapy targeting T-cell acute lymphoblastic leukemia (T-ALL) faces limitations such as antigen selection and limited T-cell persistence. CD7 is an attractive antigen for targeting T-ALL, but overlapping expression on healthy T cells leads to fratricide of CD7-CAR T cells, requiring additional genetic modification. We took advantage of naturally occurring CD7- T cells to generate CD7-CAR (CD7-CARCD7-) T cells. CD7-CARCD7- T cells exhibited a predominantly CD4+ memory phenotype and had significant antitumor activity upon chronic antigen exposure in vitro and in xenograft mouse models. Based on these encouraging results, we next explored the utility of CD7- T cells for the immunotherapy of CD19+ hematological malignancies. Direct comparison of nonselected (bulk) CD19-CAR and CD19-CARCD7- T cells revealed that CD19-CARCD7- T cells had enhanced antitumor activity compared with their bulk counterparts in vitro and in vivo. Lastly, to gain insight into the behavior of CD19-CAR T cells with low levels of CD7 gene expression (CD7lo) in humans, we mined single-cell gene and T-cell receptor (TCR) expression data sets from our institutional CD19-CAR T-cell clinical study. CD19-CARCD7lo T cells were present in the initial CD19-CAR T-cell product and could be detected postinfusion. Intriguingly, the only functional CD4+ CD19-CAR T-cell cluster observed postinfusion exhibited CD7lo expression. Additionally, samples from patients responsive to therapy had a higher proportion of CD7lo T cells than nonresponders (NCT03573700). Thus, CARCD7- T cells have favorable biological characteristics and may present a promising T-cell subset for adoptive cell therapy of T-ALL and other hematological malignancies.


Subject(s)
Hematologic Neoplasms , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma , Humans , Mice , Animals , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Receptors, Antigen, T-Cell , Immunotherapy, Adoptive , Hematologic Neoplasms/therapy , Immunotherapy , Antigens, CD19
2.
Haematologica ; 109(6): 1656-1667, 2024 06 01.
Article in English | MEDLINE | ID: mdl-38832421

ABSTRACT

Recurrent and/or refractory (R/R) pediatric acute myeloid leukemia (AML) remains a recalcitrant disease with poor outcomes. Cell therapy with genetically modified immune effector cells holds the promise to improve outcomes for R/R AML since it relies on cytotoxic mechanisms that are distinct from chemotherapeutic agents. While T cells expressing chimeric antigen receptors (CAR T cells) showed significant anti-AML activity in preclinical models, early phase clinical studies have demonstrated limited activity, irrespective of the targeted AML antigen. Lack of efficacy is most likely multifactorial, including: (i) a limited array of AML-specific targets and target antigen heterogeneity; (ii) the aggressive nature of R/R AML and heavy pretreatment of patients; (iii) T-cell product manufacturing, and (iv) limited expansion and persistence of the CAR T cells, which is in part driven by the immunosuppressive AML microenvironment. Here we review the results of early phase clinical studies with AML-specific CAR T cells, and avenues investigators are exploring to improve their effector function.


Subject(s)
Immunotherapy, Adoptive , Leukemia, Myeloid, Acute , Receptors, Chimeric Antigen , Humans , Leukemia, Myeloid, Acute/therapy , Leukemia, Myeloid, Acute/immunology , Receptors, Chimeric Antigen/immunology , Immunotherapy, Adoptive/methods , Child , Clinical Trials as Topic , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Treatment Outcome , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/genetics , Tumor Microenvironment/immunology , Animals
3.
Gene Ther ; 30(3-4): 222-231, 2023 04.
Article in English | MEDLINE | ID: mdl-34997202

ABSTRACT

Autologous chimeric antigen receptor (CAR) T cells targeting the CD19 antigen have demonstrated a high complete response rate in relapsed/refractory B-cell malignancies. However, autologous CAR T cell therapy is not an option for all patients. Here we optimized conditions for clinical-grade manufacturing of allogeneic CD19-CAR T cells using CD45RA-depleted donor memory T cells (Tm) for a planned clinical trial. Tm were activated using the MACS GMP T Cell TransAct reagent and transduced in the presence of LentiBOOST with a clinical-grade lentiviral vector that encodes a 2nd generation CD19-CAR with a 41BB.zeta endodomain. Transduced T cells were transferred to a G-Rex cell culture device for expansion and harvested on day 7 or 8 for cryopreservation. The resulting CD19-CAR(Mem) T cells expanded on average 34.2-fold, and mean CAR expression was 45.5%. The majority of T cells were CD4+ and had a central memory or effector memory phenotype, and retained viral specificity. CD19-CAR(Mem) T cells recognized and killed CD19-positive target cells in vitro and had potent antitumor activity in an ALL xenograft model. Thus we have successfully developed a current good manufacturing practice-compliant process to manufacture donor-derived CD19-CAR(Mem) T cells. Our manufacturing process could be readily adapted for CAR(Mem) T cells targeting other antigens.


Subject(s)
Neoplasms , Receptors, Antigen, T-Cell , Humans , Antigens, CD19/genetics , Immunotherapy, Adoptive/methods , T-Lymphocytes , Cyclic GMP/metabolism
4.
Haematologica ; 108(4): 1039-1052, 2023 04 01.
Article in English | MEDLINE | ID: mdl-35899386

ABSTRACT

The outcome of patients with acute myeloid leukemia remains poor, and immunotherapy has the potential to improve this. T cells expressing chimeric antigen receptors or bispecific T-cell engagers targeting CD123 are actively being explored in preclinical and/or early phase clinical studies. We have shown that T cells expressing CD123-specific bispecific T-cell engagers (CD123.ENG T cells) have anti-acute myeloid leukemia activity. However, like chimeric antigen receptor T cells, their effector function diminishes rapidly once they are repeatedly exposed to antigen-positive target cells. Here we sought to improve the effector function of CD123.ENG T cells by expressing inducible co-stimulatory molecules consisting of MyD88 and CD40 (iMC), MyD88 (iM), or CD40 (iC), which are activated by a chemical inducer of dimerization. CD123.ENG T cells expressing iMC, iM, or iC maintained their antigen specificity in the presence of a chemical inducer of dimerization, as judged by cytokine production (interferon-γ, interleukin-2) and their cytolytic activity. In repeat stimulation assays, activating iMC and iM, in contrast to iC, enabled CD123.ENG T cells to secrete cytokines, expand, and kill CD123-positive target cells repeatedly. Activating iMC in CD123.ENG T cells consistently improved antitumor activity in an acute myeloid leukemia xenograft model. This translated into a significant survival advantage in comparison to that of mice that received CD123.ENG or CD123.ENG.iC T cells. In contrast, activation of only iM in CD123.ENG T cells resulted in donor-dependent antitumor activity. Our work highlights the need for both toll-like receptor pathway activation via MyD88 and provision of co-stimulation via CD40 to consistently enhance the antitumor activity of CD123.ENG T cells.


Subject(s)
Leukemia, Myeloid, Acute , T-Lymphocytes , Animals , Humans , Mice , Cell Line, Tumor , Interleukin-3 Receptor alpha Subunit/metabolism , Leukemia, Myeloid, Acute/metabolism , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , T-Lymphocytes/metabolism , CD40 Antigens/metabolism
6.
Cell Rep Med ; 5(2): 101422, 2024 Feb 20.
Article in English | MEDLINE | ID: mdl-38350450

ABSTRACT

The emergence of immune escape is a significant roadblock to developing effective chimeric antigen receptor (CAR) T cell therapies against hematological malignancies, including acute myeloid leukemia (AML). Here, we demonstrate feasibility of targeting two antigens simultaneously by combining a GRP78-specific peptide antigen recognition domain with a CD123-specific scFv to generate a peptide-scFv bispecific antigen recognition domain (78.123). To achieve this, we test linkers with varying length and flexibility and perform immunophenotypic and functional characterization. We demonstrate that bispecific CAR T cells successfully recognize and kill tumor cells that express GRP78, CD123, or both antigens and have improved antitumor activity compared to their monospecific counterparts when both antigens are expressed. Protein structure prediction suggests that linker length and compactness influence the functionality of the generated bispecific CARs. Thus, we present a bispecific CAR design strategy to prevent immune escape in AML that can be extended to other peptide-scFv combinations.


Subject(s)
Leukemia, Myeloid, Acute , Receptors, Chimeric Antigen , Humans , T-Lymphocytes , Interleukin-3 Receptor alpha Subunit/metabolism , Endoplasmic Reticulum Chaperone BiP , Receptors, Chimeric Antigen/metabolism , Leukemia, Myeloid, Acute/pathology
7.
J Hematol Oncol ; 17(1): 50, 2024 Jun 27.
Article in English | MEDLINE | ID: mdl-38937803

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).


Subject(s)
Hematologic Neoplasms , Hematopoietic Stem Cell Transplantation , Killer Cells, Natural , Memory T Cells , Transplantation Conditioning , Transplantation, Haploidentical , Humans , Female , Male , Killer Cells, Natural/transplantation , Killer Cells, Natural/immunology , Child , Adolescent , Transplantation, Haploidentical/methods , Child, Preschool , Hematopoietic Stem Cell Transplantation/methods , Hematopoietic Stem Cell Transplantation/adverse effects , Transplantation Conditioning/methods , Hematologic Neoplasms/therapy , Graft vs Host Disease/prevention & control , Graft vs Host Disease/etiology , Infant , Young Adult , Adult , Treatment Outcome , Graft vs Leukemia Effect
8.
Cell Rep Med ; 4(11): 101297, 2023 11 21.
Article in English | MEDLINE | ID: mdl-37992682

ABSTRACT

Lack of targetable antigens is a key limitation for developing successful T cell-based immunotherapies. Members of the unfolded protein response (UPR) represent ideal immunotherapy targets because the UPR regulates the ability of cancer cells to resist cell death, sustain proliferation, and metastasize. Glucose-regulated protein 78 (GRP78) is a key UPR regulator that is overexpressed and translocated to the cell surface of a wide variety of cancers in response to elevated endoplasmic reticulum (ER) stress. We show that GRP78 is highly expressed on the cell surface of multiple solid and brain tumors, making cell surface GRP78 a promising chimeric antigen receptor (CAR) T cell target. We demonstrate that GRP78-CAR T cells can recognize and kill GRP78+ brain and solid tumors in vitro and in vivo. Additionally, our findings demonstrate that GRP78 is upregulated on CAR T cells upon T cell activation; however, this expression is tumor-cell-line specific and results in heterogeneous GRP78-CAR T cell therapeutic response.


Subject(s)
Brain Neoplasms , Receptors, Chimeric Antigen , Humans , Endoplasmic Reticulum Chaperone BiP , Glucose , T-Lymphocytes , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Brain Neoplasms/therapy
9.
Cell Rep ; 42(7): 112804, 2023 07 25.
Article in English | MEDLINE | ID: mdl-37453060

ABSTRACT

The bone marrow microenvironment (BME) drives drug resistance in acute lymphoblastic leukemia (ALL) through leukemic cell interactions with bone marrow (BM) niches, but the underlying mechanisms remain unclear. Here, we show that the interaction between ALL and mesenchymal stem cells (MSCs) through integrin ß1 induces an epithelial-mesenchymal transition (EMT)-like program in MSC-adherent ALL cells, resulting in drug resistance and enhanced survival. Moreover, single-cell RNA sequencing analysis of ALL-MSC co-culture identifies a hybrid cluster of MSC-adherent ALL cells expressing both B-ALL and MSC signature genes, orchestrated by a WNT/ß-catenin-mediated EMT-like program. Blockade of interaction between ß-catenin and CREB binding protein impairs the survival and drug resistance of MSC-adherent ALL cells in vitro and results in a reduction in leukemic burden in vivo. Targeting of this WNT/ß-catenin-mediated EMT-like program is a potential therapeutic approach to overcome cell extrinsically acquired drug resistance in ALL.


Subject(s)
Epithelial-Mesenchymal Transition , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Humans , beta Catenin , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Coculture Techniques , Drug Resistance , Cell Proliferation , Tumor Microenvironment
10.
Front Immunol ; 13: 867103, 2022.
Article in English | MEDLINE | ID: mdl-35401520

ABSTRACT

In recent years, there has been an emphasis on harnessing the immune system for therapeutic interventions. Adoptive cell therapies (ACT) have emerged as an effective option for B-cell derived hematological malignancies. Despite remarkable successes with ACT, immune dysregulation and the leukemia microenvironment can critically alter clinical responses. Therefore, preclinical modeling can contribute to the advancement of ACT for leukemias. Human xenografts, the current mainstay of ACT in vivo models, cannot evaluate the impact of the immunosuppressive leukemia microenvironment on adoptively transferred cells. Syngeneic mouse models utilize murine tumor models and implant them into immunocompetent mice. This provides an alternative model, reducing the need for complicated breeding strategies while maintaining a matched immune system, stromal compartment, and leukemia burden. Syngeneic models that evaluate ACT have analyzed the complexity of cytotoxic T lymphocytes, T cell receptor transgenics, and chimeric antigen receptors. This review examines the immunosuppressive features of the leukemia microenvironment, discusses how preclinical modeling helps predict ACT associated toxicities and dysfunction, and explores publications that have employed syngeneic modeling in ACT studies for the improvement of therapy for leukemias.


Subject(s)
Leukemia , Receptors, Chimeric Antigen , Animals , Humans , Immunosuppressive Agents , Immunotherapy, Adoptive , Leukemia/therapy , Mice , T-Lymphocytes, Cytotoxic , Tumor Microenvironment
11.
Nat Commun ; 13(1): 587, 2022 01 31.
Article in English | MEDLINE | ID: mdl-35102167

ABSTRACT

Developing CAR T cells for acute myeloid leukemia (AML) has been hampered by a paucity of targets that are expressed on AML blasts and not on hematopoietic progenitor cells (HPCs). Here we demonstrate that GRP78 is expressed on the cell surface of primary AML blasts but not HPCs. To target GRP78, we generate T cell expressing a GRP78-specific peptide-based CAR, which show evidence of minimal fratricide post activation/transduction and antigen-dependent T cell differentiation. GRP78-CAR T cells recognize and kill GRP78-positive AML cells without toxicity to HPCs. In vivo, GRP78-CAR T cells have significant anti-AML activity. To prevent antigen-dependent T cell differentiation, we block CAR signaling and GRP78 cell surface expression post activation by using dasatinib during GRP78-CAR T cell manufacturing. This significantly improves their effector function in vitro and in vivo. Thus, targeting cell surface GRP78-positive AML with CAR T cells is feasible, and warrants further active exploration.


Subject(s)
Cell Membrane/metabolism , Endoplasmic Reticulum Chaperone BiP/immunology , Hematopoietic Stem Cells/immunology , Leukemia, Myeloid, Acute/immunology , Receptors, Chimeric Antigen/immunology , T-Lymphocytes/immunology , Animals , Cell Line, Tumor , Cell Membrane/drug effects , Cell Survival/drug effects , Cytokines/metabolism , Cytotoxicity, Immunologic/drug effects , Dasatinib/pharmacology , Gene Expression Regulation, Leukemic/drug effects , Humans , Leukemia, Myeloid, Acute/genetics , Mice, Inbred NOD , Mice, SCID , T-Lymphocytes/drug effects , Xenograft Model Antitumor Assays
12.
Cancer Discov ; 12(9): 2098-2119, 2022 09 02.
Article in English | MEDLINE | ID: mdl-35792801

ABSTRACT

Current chimeric antigen receptor-modified (CAR) T-cell products are evaluated in bulk, without assessing functional heterogeneity. We therefore generated a comprehensive single-cell gene expression and T-cell receptor (TCR) sequencing data set using pre- and postinfusion CD19-CAR T cells from blood and bone marrow samples of pediatric patients with B-cell acute lymphoblastic leukemia. We identified cytotoxic postinfusion cells with identical TCRs to a subset of preinfusion CAR T cells. These effector precursor cells exhibited a unique transcriptional profile compared with other preinfusion cells, corresponding to an unexpected surface phenotype (TIGIT+, CD62Llo, CD27-). Upon stimulation, these cells showed functional superiority and decreased expression of the exhaustion-associated transcription factor TOX. Collectively, these results demonstrate diverse effector potentials within preinfusion CAR T-cell products, which can be exploited for therapeutic applications. Furthermore, we provide an integrative experimental and analytic framework for elucidating the mechanisms underlying effector development in CAR T-cell products. SIGNIFICANCE: Utilizing clonal trajectories to define transcriptional potential, we find a unique signature of CAR T-cell effector precursors present in preinfusion cell products. Functional assessment of cells with this signature indicated early effector potential and resistance to exhaustion, consistent with postinfusion cellular patterns observed in patients. This article is highlighted in the In This Issue feature, p. 2007.


Subject(s)
Receptors, Chimeric Antigen , T-Lymphocytes , Antigens, CD19 , Humans , Immunotherapy, Adoptive/methods , Receptors, Antigen, T-Cell/genetics , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/metabolism
13.
Blood Adv ; 6(21): 5737-5749, 2022 11 08.
Article in English | MEDLINE | ID: mdl-35446934

ABSTRACT

T cells expressing CD19-specific chimeric antigen receptors (CD19-CARs) have potent antileukemia activity in pediatric and adult patients with relapsed and/or refractory B-cell acute lymphoblastic leukemia (B-ALL). However, not all patients achieve a complete response (CR), and a significant percentage relapse after CD19-CAR T-cell therapy due to T-cell intrinsic and/or extrinsic mechanisms. Thus, there is a need to evaluate new CD19-CAR T-cell products in patients to improve efficacy. We developed a phase 1/2 clinical study to evaluate an institutional autologous CD19-CAR T-cell product in pediatric patients with relapsed/refractory B-ALL. Here we report the outcome of the phase 1 study participants (n = 12). Treatment was well tolerated, with a low incidence of both cytokine release syndrome (any grade, n = 6) and neurotoxicity (any grade, n = 3). Nine out of 12 patients (75%) achieved a minimal residual disease-negative CR in the bone marrow (BM). High disease burden (≥40% morphologic blasts) before CAR T-cell infusion correlated with increased side effects and lower response rate, but not with CD19-CAR T-cell expansion. After infusion, CD8+ CAR T cells had a proliferative advantage over CD4+ CAR T cells and at peak expansion, had an effector memory phenotype with evidence of antigen-driven differentiation. Patients that proceeded to allogeneic hematopoietic cell transplantation (AlloHCT) had sustained, durable responses. In summary, the initial evaluation of our institutional CD19-CAR T-cell product demonstrates safety and efficacy while highlighting the impact of pre-infusion disease burden on outcomes. This trial was registered at www.clinicaltrials.gov as #NCT03573700.


Subject(s)
Burkitt Lymphoma , Lymphoma, B-Cell , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Receptors, Chimeric Antigen , Humans , Antigens, CD19 , CD8-Positive T-Lymphocytes , Cost of Illness , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , T-Lymphocytes
14.
Blood Cancer Discov ; 2(6): 559-561, 2021 Nov.
Article in English | MEDLINE | ID: mdl-35015677

ABSTRACT

Ideal targets for chimeric antigen receptor T-cell therapy for acute myeloid leukemia (AML) remain elusive. In this issue of Blood Cancer Discovery, Richards and colleagues explore CD93 as a potential AML target antigen, and devise an approach to mitigate "on-target/off-cancer toxicity."See related article by Richards et al., p. 648.

15.
Front Oncol ; 10: 262, 2020.
Article in English | MEDLINE | ID: mdl-32185132

ABSTRACT

Chimeric antigen receptor (CAR) T cells targeting CD19 have been successful treating patients with relapsed/refractory B cell acute lymphoblastic leukemia (ALL) and B cell lymphomas. However, relapse after CAR T cell therapy is still a challenge. In addition, preclinical and early clinical studies targeting acute myeloid leukemia (AML) have not been as successful. This can be attributed in part to the presence of an AML microenvironment that has a dampening effect on the antitumor activity of CAR T cells. The AML microenvironment includes cellular interactions, soluble environmental factors, and structural components. Suppressive immune cells including myeloid derived suppressor cells and regulatory T cells are known to inhibit T cell function. Environmental factors contributing to T cell exhaustion, including immune checkpoints, anti-inflammatory cytokines, chemokines, and metabolic alterations, impact T cell activity, persistence, and localization. Lastly, structural factors of the bone marrow niche, secondary lymphoid organs, and extramedullary sites provide opportunities for CAR T cell evasion by AML blasts, contributing to treatment resistance and relapse. In this review we discuss the effect of the AML microenvironment on CAR T cell function. We highlight opportunities to enhance CAR T cell efficacy for AML through manipulating, targeting, and evading the anti-inflammatory leukemic microenvironment.

16.
Pediatr Blood Cancer ; 53(6): 1060-3, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19637317

ABSTRACT

BACKGROUND: Red cell exchange (RCE) is part of the management of acute chest syndrome (ACS) in patients with sickle cell disease. The study describes the indications and outcome of the procedure. PROCEDURE: We retrospectively reviewed 53 episodes of ACS in 44 patients who received RCE from January 2003 to October 2006. Patients were aged between 18 months and 19 years. RESULTS: Sixty-two percent of the patients had at least one previous episode of ACS. ACS was diagnosed at presentation in 66%, in the remainder ACS was diagnosed after a median of 2 days. Clinical Respiratory Score (CRS) was assigned retrospectively to assess respiratory distress (0 = no distress, > 6 = severe). Median admission CRS of 2, progressed to 4 before RCE and declined to 2 within 24 hr afterwards. Median day of RCE was day 2 (IQR 1-3) and the main indication was worsening respiratory distress. No patient developed venous thrombosis, alloantibodies or other complications from RCE. Median length of hospitalization was 7 days (IQR: 5-9 days). Patients with a platelet count significantly lower than their baseline on admission had an increased risk of mechanical ventilation. CONCLUSIONS: RCE appears to be a safe and effective treatment for patients with sickle cell disease and ACS. One-third of patients who received RCE for ACS had no respiratory symptoms on admission.


Subject(s)
Anemia, Sickle Cell/therapy , Cytapheresis , Erythrocyte Transfusion , Adolescent , Anemia, Sickle Cell/complications , Chest Pain , Child , Female , Humans , Length of Stay , Male , Platelet Count , Respiration, Artificial , Respiratory Insufficiency , Retrospective Studies , Syndrome
17.
SELECTION OF CITATIONS
SEARCH DETAIL