Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 25
1.
Blood ; 143(11): 1049-1054, 2024 Mar 14.
Article En | MEDLINE | ID: mdl-38052031

ABSTRACT: We show that red cell exchange (RCE) treats hyperleukocytosis in acute leukemia. RCE provided similar leukoreduction to standard therapeutic leukoreduction and could be superior in patients with severe anemia or monocytic leukemias or when requiring rapid treatment.


Leukemia, Monocytic, Acute , Leukemia, Myeloid, Acute , Leukostasis , Adult , Humans , Leukostasis/therapy , Leukemia, Myeloid, Acute/therapy , Leukemia, Monocytic, Acute/therapy , Acute Disease , Leukapheresis , Leukocytosis/therapy
2.
Cell Rep Med ; 4(12): 101336, 2023 12 19.
Article En | MEDLINE | ID: mdl-38118406

Pre-existing anti-human leukocyte antigen (HLA) allo-antibodies constitute a major barrier to transplantation. Current desensitization approaches fail due to ineffective depletion of allo-specific memory B cells (Bmems) and long-lived plasma cells (LLPCs). We evaluate the efficacy of chimeric antigen receptor (CAR) T cells targeting CD19 and B cell maturation antigen (BCMA) to eliminate allo-antibodies in a skin pre-sensitized murine model of islet allo-transplantation. We find that treatment of allo-sensitized hosts with CAR T cells targeting Bmems and LLPCs eliminates donor-specific allo-antibodies (DSAs) and mitigates hyperacute rejection of subsequent islet allografts. We then assess the clinical efficacy of the CAR T therapy for desensitization in patients with multiple myeloma (MM) with pre-existing HLA allo-antibodies who were treated with the combination of CART-BCMA and CART-19 (ClinicalTrials.gov: NCT03549442) and observe clinically meaningful allo-antibody reduction. These findings provide logical rationale for clinical evaluation of CAR T-based immunotherapy in highly sensitized candidates to promote successful transplantation.


Receptors, Chimeric Antigen , Humans , Animals , Mice , Plasma Cells , B-Cell Maturation Antigen , T-Lymphocytes , Immunotherapy , Antibodies
3.
Camb Prism Precis Med ; 1: e25, 2023.
Article En | MEDLINE | ID: mdl-38550937

Precision Medicine is an emerging approach for disease treatment and prevention that takes into account individual variability in genes, environment, and lifestyle. Autoimmune diseases are those in which the body's natural defense system loses discriminating power between its own cells and foreign cells, causing the body to mistakenly attack healthy tissues. These conditions are very heterogeneous in their presentation and therefore difficult to diagnose and treat. Achieving precision medicine in autoimmune diseases has been challenging due to the complex etiologies of these conditions, involving an interplay between genetic, epigenetic, and environmental factors. However, recent technological and computational advances in molecular profiling have helped identify patient subtypes and molecular pathways which can be used to improve diagnostics and therapeutics. This review discusses the current understanding of the disease mechanisms, heterogeneity, and pathogenic autoantigens in autoimmune diseases gained from genomic and transcriptomic studies and highlights how these findings can be applied to better understand disease heterogeneity in the context of disease diagnostics and therapeutics.

4.
J Immunol ; 209(10): 1811-1816, 2022 11 15.
Article En | MEDLINE | ID: mdl-36344114

Achieving immunosuppression-free immune tolerance to an allograft is one of the central goals of transplantation. In this article, we review recent developments in the fields of T cell-based therapies and T cell engineering using chimeric Ag receptors and their potential for effective and targeted immune modulation of T and B cell activity in an effort to eliminate pre-existing alloantibodies (desensitization) and achieve long-term tolerance. Approaches that span preclinical to early clinical studies in transplantation will be reviewed, with specific emphasis on advances in T cell immunotherapy that have shown promise. Lastly, we conclude with a forward-looking discussion of how T cell-based therapies in other fields of medicine can be potentially applied to solid organ transplantation.


Organ Transplantation , Transplantation, Homologous , Immunotherapy , Immune Tolerance , Cell- and Tissue-Based Therapy
6.
Immunol Rev ; 303(1): 154-167, 2021 09.
Article En | MEDLINE | ID: mdl-34351644

Central to protective humoral immunity is the activation of B cells and their terminal differentiation into antibody-secreting plasma cells. Long-lived plasma cells (LLPC) may survive for years to decades. Such long-lived plasma cells are also responsible for producing pathogenic antibodies that cause a variety of challenges such as autoimmunity, allograft rejection, and drug neutralization. Up to now, various therapeutic strategies aimed at durably eliminating pathogenic antibodies have failed, in large part due to their inability to efficiently target LLPCs. Several antibody-based therapies have recently gained regulatory approval or are in clinical phases of development for the treatment of multiple myeloma, a malignancy of plasma cells. We discuss the exciting potential of using these emerging cancer immunotherapies to solve the antibody problem.


B-Lymphocytes , Plasma Cells , Antibody-Producing Cells , Autoimmunity , Immunity, Humoral
7.
J Clin Invest ; 131(16)2021 08 16.
Article En | MEDLINE | ID: mdl-34396987

Chimeric antigen receptor (CAR) T cells have induced remarkable antitumor responses in B cell malignancies. Some patients do not respond because of T cell deficiencies that hamper the expansion, persistence, and effector function of these cells. We used longitudinal immune profiling to identify phenotypic and pharmacodynamic changes in CD19-directed CAR T cells in patients with chronic lymphocytic leukemia (CLL). CAR expression maintenance was also investigated because this can affect response durability. CAR T cell failure was accompanied by preexisting T cell-intrinsic defects or dysfunction acquired after infusion. In a small subset of patients, CAR silencing was observed coincident with leukemia relapse. Using a small molecule inhibitor, we demonstrated that the bromodomain and extra-terminal (BET) family of chromatin adapters plays a role in downregulating CAR expression. BET protein blockade also ameliorated CAR T cell exhaustion as manifested by inhibitory receptor reduction, enhanced metabolic fitness, increased proliferative capacity, and enriched transcriptomic signatures of T cell reinvigoration. BET inhibition decreased levels of the TET2 methylcytosine dioxygenase, and forced expression of the TET2 catalytic domain eliminated the potency-enhancing effects of BET protein targeting in CAR T cells, providing a mechanism linking BET proteins and T cell dysfunction. Thus, modulating BET epigenetic readers may improve the efficacy of cell-based immunotherapies.


Immunotherapy, Adoptive , Leukemia, Lymphocytic, Chronic, B-Cell/immunology , Leukemia, Lymphocytic, Chronic, B-Cell/therapy , Proteins/antagonists & inhibitors , Proteins/immunology , Receptors, Chimeric Antigen/immunology , T-Lymphocytes/immunology , Antigens, CD19/immunology , Azepines/pharmacology , Epigenesis, Genetic , Glycolysis/drug effects , Humans , Immune Tolerance , Immunologic Memory , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Oxidative Phosphorylation/drug effects , Receptors, Chimeric Antigen/genetics , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , Triazoles/pharmacology
8.
Blood Adv ; 5(8): 2128-2136, 2021 04 27.
Article En | MEDLINE | ID: mdl-33881465

CD19-directed chimeric antigen receptor (CAR) T cells show characteristic proliferation kinetics after infusion that correlate with response. Clearance of circulating disease, B-cell aplasia (BCA), and cytokine release syndrome (CRS) are used to observe CAR T-cell function, given the lack of commercial CAR T-cell measurement assays. We investigated the utility of common hematology laboratory parameters in 166 patients with B-cell acute lymphoblastic leukemia (B-ALL) who were treated with CAR T-cell therapy targeting CD19. CAR T-cell infusion was followed by disappearance of circulating blasts in 86% of patients at a median of 6 days. After a lag phase, there was a rapid expansion in absolute lymphocyte count (ALC) in the second week that coincided with the appearance of atypical lymphocytes. The expansion phase was followed by a contraction phase with a concomitant decrease in atypical lymphocytes. In vitro CAR T-cell studies showed similar kinetics and morphological changes. Peak ALC and overall expansion was greater in sustained responders compared with that in nonresponders. Patients with early loss of BCA and those with eventual CD19+ minimal residual disease/relapse showed lower overall lymphocyte expansion compared with the controls. Pleomorphic lymphocytosis was noted in the cerebrospinal fluid at post-CAR time points. We conclude that lymphocyte counts and differential can also be used to evaluate CAR T-cell expansion after infusion, along with BCA and CRS. This is the first report to characterize the morphology of CAR T cells and determine the utility of lymphocyte kinetics.


Immunotherapy, Adoptive , Receptors, Antigen, T-Cell , Cell Proliferation , Humans , Kinetics , Lymphocyte Count , Recurrence , T-Lymphocytes
9.
Mol Ther Oncolytics ; 20: 387-398, 2021 Mar 26.
Article En | MEDLINE | ID: mdl-33614919

Metastatic medullary thyroid cancer (MTC) is a rare but often aggressive thyroid malignancy with a 5-year survival rate of less than 40% and few effective therapeutic options. Adoptive T cell immunotherapy using chimeric antigen receptor (CAR)-modified T cells (CAR Ts) is showing encouraging results in the treatment of cancer, but development is challenged by the availability of suitable target antigens. We identified glial-derived neurotrophic factor (GDNF) family receptor alpha 4 (GFRα4) as a putative antigen target for CAR-based therapy of MTC. We show that GFRα4 is highly expressed in MTC, in parafollicular cells within the thyroid from which MTC originates, and in normal thymus. We isolated two single-chain variable fragments (scFvs) targeting GFRα4 isoforms a and b by antibody phage display. CARs bearing the CD3ζ and the CD137 costimulatory domains were constructed using these GFRα4-specific scFvs. GFRα4-specific CAR Ts trigger antigen-dependent cytotoxicity and cytokine production in vitro, and they are able to eliminate tumors derived from the MTC TT cell line in an immunodeficient mouse xenograft model of MTC. These data demonstrate the feasibility of targeting GFRα4 by CAR T and support this antigen as a promising target for adoptive T cell immunotherapy and other antibody-based therapies for MTC.

10.
Cell Immunol ; 359: 104234, 2021 01.
Article En | MEDLINE | ID: mdl-33153708

Approaches to harness the immune system to alleviate disease have become remarkably sophisticated since the crude, yet impressively-effective, attempts using live bacteria in the late 1800s. Recent evidence that engineered T cell therapy can deliver durable results in patients with cancer has spurred frenzied development in the field of T cell therapy. The myriad approaches include an innumerable variety of synthetic transgenes, multiplex gene-editing, and broader application to diseases beyond cancer. In this article, we review the preclinical studies and over a decade of clinical experience with engineered conventional T cells that have paved the way for translating engineered regulatory T cell therapies.


Immunotherapy/methods , Immunotherapy/trends , T-Lymphocytes, Regulatory/transplantation , Humans , Immunotherapy, Adoptive/methods , Neoplasms/immunology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Transgenes
12.
Cell ; 183(1): 126-142.e17, 2020 10 01.
Article En | MEDLINE | ID: mdl-32961131

CD19-directed immunotherapies are clinically effective for treating B cell malignancies but also cause a high incidence of neurotoxicity. A subset of patients treated with chimeric antigen receptor (CAR) T cells or bispecific T cell engager (BiTE) antibodies display severe neurotoxicity, including fatal cerebral edema associated with T cell infiltration into the brain. Here, we report that mural cells, which surround the endothelium and are critical for blood-brain-barrier integrity, express CD19. We identify CD19 expression in brain mural cells using single-cell RNA sequencing data and confirm perivascular staining at the protein level. CD19 expression in the brain begins early in development alongside the emergence of mural cell lineages and persists throughout adulthood across brain regions. Mouse mural cells demonstrate lower levels of Cd19 expression, suggesting limitations in preclinical animal models of neurotoxicity. These data suggest an on-target mechanism for neurotoxicity in CD19-directed therapies and highlight the utility of human single-cell atlases for designing immunotherapies.


Blood-Brain Barrier/metabolism , Epithelial Cells/metabolism , Immunotherapy, Adoptive/adverse effects , Animals , Antibodies, Bispecific/immunology , Antigens, CD19/immunology , B-Lymphocytes/immunology , Blood-Brain Barrier/immunology , Brain/immunology , Brain/metabolism , Cell Line, Tumor , Cytotoxicity, Immunologic , Humans , Immunotherapy/adverse effects , Immunotherapy/methods , Immunotherapy, Adoptive/methods , Mice , Mice, Inbred NOD , Mice, SCID , Muscle, Smooth, Vascular/metabolism , Neoplasms , Receptors, Antigen, T-Cell/immunology , Receptors, Chimeric Antigen/immunology , Single-Cell Analysis/methods , T-Lymphocytes/immunology , Xenograft Model Antitumor Assays
13.
Blood Adv ; 3(22): 3539-3549, 2019 11 26.
Article En | MEDLINE | ID: mdl-31738832

Tisagenlecleucel, a chimeric antigen receptor (CAR) T-cell product targeting CD19 is approved for relapsed/refractory B-cell acute lymphoblastic leukemia (B-ALL). However, the impact of pretreatment variables, such as CD19 expression level, on leukemic blasts, the presence of CD19- subpopulations, and especially prior CD19-targeted therapy, on the response to CAR T-cell therapy has not been determined. We analyzed 166 patients treated with CAR T-cell therapy at our institution. Eleven patients did not achieve a minimal residual disease (MRD)- deep remission, whereas 67 patients had a recurrence after achieving a MRD- deep remission: 28 patients with CD19+ leukemia and 39 patients with CD19- leukemia. Return of CD19+ leukemia was associated with loss of CAR T-cell function, whereas CD19- leukemia was associated with continued CAR T-cell function. There were no significant differences in efficacy of CAR T cells in CD19-dim B-ALL, compared with CD19-normal or -bright B-ALL. Consistent with this, CAR T cells recognized and lysed cells with very low levels of CD19 expression in vitro. The presence of dim CD19 or rare CD19- events by flow cytometry did not predict nonresponse or recurrence after CAR T-cell therapy. However, prior therapy with the CD19-directed, bispecific T-cell engager blinatumomab was associated with a significantly higher rate of failure to achieve MRD- remission or subsequent loss of remission with antigen escape. Finally, immunophenotypic heterogeneity and lineage plasticity were independent of underlying clonotype and cytogenetic abnormalities.


Antigens, CD19/immunology , Immunotherapy, Adoptive , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/immunology , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/therapy , Receptors, Antigen, T-Cell/metabolism , Adolescent , Adult , Antibodies, Bispecific/administration & dosage , Antibodies, Bispecific/adverse effects , Antibodies, Bispecific/therapeutic use , Antineoplastic Agents, Immunological/administration & dosage , Antineoplastic Agents, Immunological/adverse effects , Antineoplastic Agents, Immunological/therapeutic use , Child , Child, Preschool , Combined Modality Therapy , Cytotoxicity, Immunologic , Female , Humans , Immunophenotyping , Immunotherapy, Adoptive/adverse effects , Immunotherapy, Adoptive/methods , Infant , Male , Neoplasm, Residual/diagnosis , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/diagnosis , Receptors, Antigen, T-Cell/genetics , Recurrence , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Treatment Outcome , Young Adult
15.
Mol Ther Methods Clin Dev ; 8: 210-221, 2018 Mar 16.
Article En | MEDLINE | ID: mdl-29552577

Adoptive cellular therapy using T cells with tumor specificity derived from either natural T cell receptors (TCRs) or an artificial chimeric antigen receptor (CAR) has reached late phase clinical testing, with two CAR T cell therapies achieving regulatory approval within the United States in 2017. The effective use of these therapies depends upon an understanding of their pharmacology, which is quite divergent from traditional small molecule or biologic drugs. We review the different types of T cell therapy under clinical development, the factors affecting cellular kinetics following infusion, and the relationship between these cellular kinetics and anti-cancer activity. We also discuss the toxicity associated with T cell therapies, with an emphasis on cytokine release syndrome and neurotoxicity, and the gaps in knowledge regarding these frequent and unique adverse effects.

16.
Science ; 353(6295): 179-84, 2016 Jul 08.
Article En | MEDLINE | ID: mdl-27365313

Ideally, therapy for autoimmune diseases should eliminate pathogenic autoimmune cells while sparing protective immunity, but feasible strategies for such an approach have been elusive. Here, we show that in the antibody-mediated autoimmune disease pemphigus vulgaris (PV), autoantigen-based chimeric immunoreceptors can direct T cells to kill autoreactive B lymphocytes through the specificity of the B cell receptor (BCR). We engineered human T cells to express a chimeric autoantibody receptor (CAAR), consisting of the PV autoantigen, desmoglein (Dsg) 3, fused to CD137-CD3ζ signaling domains. Dsg3 CAAR-T cells exhibit specific cytotoxicity against cells expressing anti-Dsg3 BCRs in vitro and expand, persist, and specifically eliminate Dsg3-specific B cells in vivo. CAAR-T cells may provide an effective and universal strategy for specific targeting of autoreactive B cells in antibody-mediated autoimmune disease.


Autoantibodies/immunology , Desmoglein 3/immunology , Immunosuppression Therapy/methods , Pemphigus/therapy , Receptors, Antigen, T-Cell/immunology , Recombinant Fusion Proteins/immunology , T-Lymphocytes/immunology , Animals , Autoantibodies/blood , Autoimmunity , B-Lymphocytes/immunology , CD3 Complex/chemistry , CD3 Complex/immunology , Cell Engineering , Cytotoxicity, Immunologic , Disease Models, Animal , Humans , Immunologic Memory , Mice , Pemphigus/immunology , Protein Structure, Tertiary , Receptors, Antigen, T-Cell/genetics , Recombinant Fusion Proteins/genetics , Tumor Necrosis Factor Receptor Superfamily, Member 9/chemistry , Tumor Necrosis Factor Receptor Superfamily, Member 9/immunology
17.
Blood ; 128(3): 360-70, 2016 07 21.
Article En | MEDLINE | ID: mdl-27166358

The mechanisms underlying the maintenance of long-lasting humoral immunity are not well understood. Studies in mice indicate that plasma cells (PCs) can survive up to a lifetime, even in the absence of regeneration by B cells, implying the presence of long-lived PCs as a mechanism for long-lasting immunity. Evidence from humans treated with anti-CD20, which depletes circulating B cells, also suggests B-cell-independent long-term survival of some PCs. On the other hand, antibody responses may be sustained solely by short-lived PCs with repopulation from clonally related memory B cells. To explore PC longevity and humoral immunity in humans, we investigated the fate of PCs and their antibodies in adult and pediatric patients who received chimeric antigen receptor-based adoptive T-cell immunotherapy targeting CD19 to treat B-cell lineage malignancies (CTL019). Treatment with CTL019 is frequently associated with B-cell aplasia that can persist for years. Serum antibody titers to vaccine-related antigens were measured, and quantitative assessment of B cells and PCs in blood and bone marrow was performed at various time points before and after CTL019 therapy. While total serum immunoglobulin concentrations decline following CTL019-induced B-cell aplasia, several vaccine/pathogen-specific serum immunoglobulin G and A (IgG and IgA) titers remain relatively stable for at least 6 and 12 months posttreatment, respectively. Analysis of bone marrow biopsies after CTL019 revealed 8 patients with persistence of antibody-secreting PCs at least 25 months post-CTL019 infusion despite absence of CD19(+)CD20(+) B cells. These results provide strong evidence for the existence of memory B-cell-independent, long-lived PCs in humans that contribute to long-lasting humoral immunity.


Adoptive Transfer , Antigens, CD19 , Lymphoma, B-Cell , Plasma Cells , T-Lymphocytes , Adolescent , Adult , Antigens, CD19/blood , Antigens, CD19/immunology , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Child , Female , Humans , Immunoglobulin A/blood , Immunoglobulin A/immunology , Immunoglobulin G/blood , Immunoglobulin G/immunology , Lymphoma, B-Cell/blood , Lymphoma, B-Cell/immunology , Lymphoma, B-Cell/pathology , Lymphoma, B-Cell/therapy , Male , Middle Aged , Plasma Cells/immunology , Plasma Cells/metabolism , Plasma Cells/pathology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes/pathology , T-Lymphocytes/transplantation
18.
Discov Med ; 21(116): 283-92, 2016 04.
Article En | MEDLINE | ID: mdl-27232514

Advances in HIV drug therapy have drastically decreased mortality and significantly improved quality of life for HIV infected patients since the early days of the epidemic. However, HIV drug-resistance, drug toxicities, therapy adherence, and the need for life-long treatment remain major challenges that continue to contribute to HIV-related global health concerns. Recent advances in cancer immunotherapy have proven the potency of cellular gene therapies. An ever-growing toolbox of methods of gene manipulation, cell modification, and clinical cell manufacturing, will enable moving beyond continuous drug therapy for more effective and durable treatments, including the possibility of inducing permanent resistance to HIV. These approaches, which target both host and viral factors, capitalize on points of vulnerability in the virus life cycle. Cellular and gene therapy has the potential to be an effective one-time therapy with less toxicity. Here, we review several promising strategies currently in pre-clinical development and clinical trials.


Cell- and Tissue-Based Therapy/methods , Genetic Therapy/methods , HIV Infections/therapy , HIV-1/physiology , T-Lymphocytes/transplantation , Anti-Retroviral Agents/adverse effects , Anti-Retroviral Agents/therapeutic use , CRISPR-Cas Systems/genetics , Cell- and Tissue-Based Therapy/adverse effects , DNA-Binding Proteins/genetics , Drug Resistance, Viral , Endoribonucleases/genetics , Escherichia coli Proteins/genetics , Gene Transfer Techniques , Genetic Therapy/adverse effects , HIV Infections/immunology , HIV-1/drug effects , Humans , Quality of Life , Receptors, CCR5/genetics , Transcription Activator-Like Effector Nucleases/genetics , Transplantation, Autologous/methods , rev Gene Products, Human Immunodeficiency Virus/genetics
19.
J Clin Apher ; 31(5): 448-53, 2016 Oct.
Article En | MEDLINE | ID: mdl-26575088

The thrombotic microangiopathy (TMA) Registry Network of North America (TRNA) is a collaborative network organized for the purpose of developing a multi-institutional registry and network to conduct clinical studies in a rare patient population. The TRNA was founded in 2013 by four academic medical centers (Columbia University Medical Center, Duke University Medical Center, University of Alabama at Birmingham, and University of Pennsylvania) to develop a national and demographically diverse dataset of patients with TMA. A clinical database was developed by network members using REDCap (Research Electronic Data Capture), a web-based database developed for clinical research. To facilitate rapid Institutional Review Board (IRB) approval at multiple sites, the TRNA utilized IRBshare, a streamlined IRB process to allow patient recruitment and enrollment into the TMA registry. This article reviews the process used to establish the TRNA network and discusses the significance of the first multi-institutional clinical apheresis network developed in the United States. J. Clin. Apheresis 31:448-453, 2016. © 2015 Wiley Periodicals, Inc.


Blood Component Removal/methods , Registries , Thrombotic Microangiopathies/therapy , Academic Medical Centers , Databases, Factual , Humans , North America , United States
20.
Curr Atheroscler Rep ; 17(7): 39, 2015 Jul.
Article En | MEDLINE | ID: mdl-26004920

Lipoprotein apheresis is an extracorporeal therapy whereby apo B-containing lipoproteins (LDL, vLDL, and Lp(a)) are selectively removed from the plasma of a patient. It is a variation of plasma exchange, which non-selectively removes all plasma proteins. Lipoprotein apheresis is most commonly used to treat patients with familial hypercholesterolemia, but is also often used for other hyperlipidemic patients, including those with elevated Lp(a), to limit and/or reverse the atherosclerotic complications of high cholesterol. Herein, we provide an overview of lipoprotein apheresis including indications, techniques, efficacy, and adverse events, as well as guidelines and other considerations important for selection of patients for this therapy.


Blood Component Removal , Lipoproteins/blood , Blood Component Removal/methods , Humans , Hyperlipoproteinemia Type II/blood , Hyperlipoproteinemia Type II/therapy , Kinetics , Patient Education as Topic
...