Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Blood Adv ; 8(7): 1806-1816, 2024 Apr 09.
Article in English | MEDLINE | ID: mdl-38181784

ABSTRACT

ABSTRACT: Stable, mixed-donor-recipient chimerism after allogeneic hematopoietic stem cell transplantation (HSCT) for patients with sickle cell disease (SCD) is sufficient for phenotypic disease reversal, and results from differences in donor/recipient-red blood cell (RBC) survival. Understanding variability and predictors of RBC survival among patients with SCD before and after HSCT is critical for gene therapy research which seeks to generate sufficient corrected hemoglobin to reduce polymerization thereby overcoming the red cell pathology of SCD. This study used biotin labeling of RBCs to determine the lifespan of RBCs in patients with SCD compared with patients who have successfully undergone curative HSCT, participants with sickle cell trait (HbAS), and healthy (HbAA) donors. Twenty participants were included in the analysis (SCD pre-HSCT: N = 6, SCD post-HSCT: N = 5, HbAS: N = 6, and HbAA: N = 3). The average RBC lifespan was significantly shorter for participants with SCD pre-HSCT (64.1 days; range, 35-91) compared with those with SCD post-HSCT (113.4 days; range, 105-119), HbAS (126.0 days; range, 119-147), and HbAA (123.7 days; range, 91-147) (P<.001). RBC lifespan correlated with various hematologic parameters and strongly correlated with the average final fraction of sickled RBCs after deoxygenation (P<.001). No adverse events were attributable to the use of biotin and related procedures. Biotin labeling of RBCs is a safe and feasible methodology to evaluate RBC survival in patients with SCD before and after HSCT. Understanding differences in RBC survival may ultimately guide gene therapy protocols to determine hemoglobin composition required to reverse the SCD phenotype as it relates directly to RBC survival. This trial was registered at www.clinicaltrials.gov as #NCT04476277.


Subject(s)
Anemia, Sickle Cell , Hematopoietic Stem Cell Transplantation , Humans , Anemia, Sickle Cell/pathology , Biotin , Erythrocytes/pathology , Hematopoietic Stem Cell Transplantation/adverse effects , Hematopoietic Stem Cell Transplantation/methods , Hemoglobins
2.
Mol Ther Methods Clin Dev ; 31: 101155, 2023 Dec 14.
Article in English | MEDLINE | ID: mdl-38074412

ABSTRACT

Ex vivo autologous hematopoietic stem cell lentiviral-based gene therapy with betibeglogene autotemcel has been studied in patients with transfusion-dependent ß-thalassemia in Phase III clinical trials (HGB-207 and HGB-212), with 90% of patients reaching transfusion independence (TI). Here, we explore manufacturing parameters, drug product quality attributes, and limited patient characteristics that had an impact on clinical efficacy in HGB-207 and HGB-212. Retrospective analysis revealed that the peripheral blood vector copy number (VCN) was related to TI, with a strong correlation between peripheral blood VCN at 6 months and gene therapy-derived therapeutic protein (HbAT87Q) expression at 6 months (correlation coefficient, 0.8681; p < 0.0001; R2 = 0.7536). A peripheral blood VCN threshold of ≥0.75 copies per diploid genome at 6 months post betibeglogene autotemcel infusion provided a stringent surrogate biomarker for TI and was used as the outcome variable for multivariate analysis using a random forest classifier. The top predictive feature of clinical efficacy was found to be the percentage of lentiviral vector-positive cells in the drug product. This retrospective analysis is critical to understanding the key product quality attributes that can predict clinical efficacy in lentiviral vector gene therapy within this clinical trial population.

3.
Am J Hematol ; 98(1): 11-22, 2023 01.
Article in English | MEDLINE | ID: mdl-36161320

ABSTRACT

lovo-cel (bb1111; LentiGlobin for sickle cell disease [SCD]) gene therapy (GT) comprises autologous transplantation of hematopoietic stem and progenitor cells transduced with the BB305 lentiviral vector encoding a modified ß-globin gene (ßA-T87Q ) to produce anti-sickling hemoglobin (HbAT87Q ). The efficacy and safety of lovo-cel for SCD are being evaluated in the ongoing phase 1/2 HGB-206 study (ClinicalTrials.gov: NCT02140554). The treatment process evolved over time, using learnings from outcomes in the initial patients to optimize lovo-cel's benefit-risk profile. Following modest expression of HbAT87Q in the initial patients (Group A, n = 7), alterations were made to the treatment process for patients subsequently enrolled in Group B (n = 2, patients B1 and B2), including improvements to cell collection and lovo-cel manufacturing. After 6 months, median Group A peripheral blood vector copy number (≥0.08 c/dg) and HbAT87Q levels (≥0.46 g/dL) were inadequate for substantial clinical effect but stable and sustained over 5.5 years; both markedly improved in Group B (patient B1: ≥0.53 c/dg and ≥2.69 g/dL; patient B2: ≥2.14 c/dg and ≥6.40 g/dL, respectively) and generated improved biologic and clinical efficacy in Group B, including higher total hemoglobin and decreased hemolysis. The safety of the lovo-cel for SCD treatment regimen largely reflected the known side effects of HSPC collection, busulfan conditioning regimen, and underlying SCD; acute myeloid leukemia was observed in two patients in Group A and deemed unlikely related to insertional oncogenesis. Changes made during development of the lovo-cel treatment process were associated with improved outcomes and provide lessons for future SCD GT studies.


Subject(s)
Anemia, Sickle Cell , Hematopoietic Stem Cell Transplantation , Humans , Lentivirus/genetics , Anemia, Sickle Cell/genetics , Anemia, Sickle Cell/therapy , Genetic Therapy/adverse effects , Hemoglobins/genetics
4.
N Engl J Med ; 386(7): 617-628, 2022 02 17.
Article in English | MEDLINE | ID: mdl-34898139

ABSTRACT

BACKGROUND: Sickle cell disease is characterized by the painful recurrence of vaso-occlusive events. Gene therapy with the use of LentiGlobin for sickle cell disease (bb1111; lovotibeglogene autotemcel) consists of autologous transplantation of hematopoietic stem and progenitor cells transduced with the BB305 lentiviral vector encoding a modified ß-globin gene, which produces an antisickling hemoglobin, HbAT87Q. METHODS: In this ongoing phase 1-2 study, we optimized the treatment process in the initial 7 patients in Group A and 2 patients in Group B with sickle cell disease. Group C was established for the pivotal evaluation of LentiGlobin for sickle cell disease, and we adopted a more stringent inclusion criterion that required a minimum of four severe vaso-occlusive events in the 24 months before enrollment. In this unprespecified interim analysis, we evaluated the safety and efficacy of LentiGlobin in 35 patients enrolled in Group C. Included in this analysis was the number of severe vaso-occlusive events after LentiGlobin infusion among patients with at least four vaso-occlusive events in the 24 months before enrollment and with at least 6 months of follow-up. RESULTS: As of February 2021, cell collection had been initiated in 43 patients in Group C; 35 received a LentiGlobin infusion, with a median follow-up of 17.3 months (range, 3.7 to 37.6). Engraftment occurred in all 35 patients. The median total hemoglobin level increased from 8.5 g per deciliter at baseline to 11 g or more per deciliter from 6 months through 36 months after infusion. HbAT87Q contributed at least 40% of total hemoglobin and was distributed across a mean (±SD) of 85±8% of red cells. Hemolysis markers were reduced. Among the 25 patients who could be evaluated, all had resolution of severe vaso-occlusive events, as compared with a median of 3.5 events per year (range, 2.0 to 13.5) in the 24 months before enrollment. Three patients had a nonserious adverse event related or possibly related to LentiGlobin that resolved within 1 week after onset. No cases of hematologic cancer were observed during up to 37.6 months of follow-up. CONCLUSIONS: One-time treatment with LentiGlobin resulted in sustained production of HbAT87Q in most red cells, leading to reduced hemolysis and complete resolution of severe vaso-occlusive events. (Funded by Bluebird Bio; HGB-206 ClinicalTrials.gov number, NCT02140554.).


Subject(s)
Anemia, Sickle Cell/therapy , Genetic Therapy , Genetic Vectors , Hematopoietic Stem Cell Transplantation , Hemoglobins/genetics , Lentivirus , Stem Cell Transplantation , beta-Globins/genetics , Adolescent , Adult , Anemia, Sickle Cell/blood , Anemia, Sickle Cell/complications , Child , Female , Fetal Hemoglobin , Hemoglobins/analysis , Hemoglobins/metabolism , Humans , Male , Middle Aged , Vascular Patency , Young Adult
6.
Br J Haematol ; 186(2): 286-299, 2019 07.
Article in English | MEDLINE | ID: mdl-30972754

ABSTRACT

Stress erythropoiesis and chronic inflammation in subjects with sickle cell disease (SCD) may have an impact on the bone marrow (BM) haematopoietic stem and progenitor cell (HSPC) quality and yield necessary for effective autologous, ex vivo HSPC gene therapy. BM from 19 subjects with SCD and five volunteers without SCD (non-SCD) was collected in different anticoagulants and processed immediately (day 0) or the following day (day 1). Inflammatory, contamination and aggregation markers within the mononuclear layer, and CD34, CD45 and Glycophorin-A (GPA) expression on HSPCs after CD34+ selection were analysed by conventional and imaging flow cytometry. Compared to non-SCD BM, multiple markers of inflammation, contamination (red cells, P < 0·01; platelets, P < 0·01) and aggregates (platelet/granulocytes, P < 0·01; mononuclear/red cells, P < 0·01) were higher in SCD BM. Total CD34+ cell count was lower in SCD BM (P < 0·05), however CD34+ count was higher in SCD BM when collected in acid citrate dextrose-A (ACDA) versus heparin (P < 0·05). Greater than 50% of CD34+ HSPCs from SCD BM are CD34dim due to higher erythroid lineage expression (P < 0·01) as single cell CD34+ CD45+ GPA+ (P < 0·01) and CD34+ CD45- GPA+ (P < 0·01) HSPCs. SCD BM is characterized by increased inflammation, aggregation and contamination contributing to significant differences in HSPC quality and yield compared to non-SCD BM.


Subject(s)
Anemia, Sickle Cell , Antigens, CD34/metabolism , Erythropoiesis , Hematopoietic Stem Cells , Stress, Physiological , Adult , Anemia, Sickle Cell/metabolism , Anemia, Sickle Cell/pathology , Female , Flow Cytometry , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/pathology , Humans , Inflammation/metabolism , Inflammation/pathology , Male
7.
Blood Adv ; 2(19): 2505-2512, 2018 10 09.
Article in English | MEDLINE | ID: mdl-30282642

ABSTRACT

Novel therapies for sickle cell disease (SCD) based on genetically engineered autologous hematopoietic stem and progenitor cells (HSPCs) are critically dependent on a safe and effective strategy for cell procurement. We sought to assess the safety and efficacy of plerixafor when used in transfused patients with SCD for HSC mobilization. Six adult patients with SCD were recruited to receive a single dose of plerixafor, tested at lower than standard (180 µg/kg) and standard (240 µg/kg) doses, followed by CD34+ cell monitoring in peripheral blood and apheresis collection. The procedures were safe and well-tolerated. Mobilization was successful, with higher peripheral CD34+ cell counts in the standard vs the low-dose group. Among our 6 donors, we improved apheresis cell collection results by using a deep collection interface and starting apheresis within 4 hours after plerixafor administration. In the subjects who received a single standard dose of plerixafor and followed the optimized collection protocol, yields of up to 24.5 × 106 CD34+ cells/kg were achieved. Interestingly, the collected CD34+ cells were enriched in immunophenotypically defined long-term HSCs and early progenitors. Thus, we demonstrate that plerixafor can be employed safely in patients with SCD to obtain sufficient HSCs for potential use in gene therapy.


Subject(s)
Anemia, Sickle Cell/therapy , Blood Component Removal , Hematopoietic Stem Cell Mobilization , Hematopoietic Stem Cells/metabolism , Heterocyclic Compounds/administration & dosage , Adolescent , Adult , Benzylamines , Cyclams , Dose-Response Relationship, Drug , Genetic Therapy/methods , Hematopoietic Stem Cell Mobilization/methods , Hematopoietic Stem Cells/drug effects , Humans , Immunophenotyping , Peripheral Blood Stem Cell Transplantation/methods , Pilot Projects , Young Adult
8.
Mol Ther ; 26(1): 320-328, 2018 01 03.
Article in English | MEDLINE | ID: mdl-29102562

ABSTRACT

Gene therapy currently in development for hemoglobinopathies utilizes ex vivo lentiviral transduction of CD34+ hematopoietic stem and progenitor cells (HSPCs). A small-molecule screen identified prostaglandin E2 (PGE2) as a positive mediator of lentiviral transduction of CD34+ cells. Supplementation with PGE2 increased lentiviral vector (LVV) transduction of CD34+ cells approximately 2-fold compared to control transduction methods with no effect on cell viability. Transduction efficiency was consistently increased in primary CD34+ cells from multiple normal human donors and from patients with ß-thalassemia or sickle cell disease. Notably, PGE2 increased transduction of repopulating human HSPCs in an immune-deficient (nonobese diabetic/severe combined immunodeficiency/interleukin-2 gamma receptor null [NSG]) xenotransplantation mouse model without evidence of in vivo toxicity, lineage bias, or a de novo bias of lentiviral integration sites. These data suggest that PGE2 improves lentiviral transduction and increases vector copy number, therefore resulting in increased transgene expression. As a result, PGE2 may be useful in clinical gene therapy applications using lentivirally modified HSPCs.


Subject(s)
Dinoprostone/metabolism , Genetic Vectors/genetics , Hematopoietic Stem Cells/metabolism , Lentivirus/genetics , Transduction, Genetic , Anemia, Sickle Cell/genetics , Anemia, Sickle Cell/metabolism , Animals , Antigens, CD34/metabolism , Cell Line , Gene Library , Gene Transfer Techniques , Genetic Therapy , Globins/genetics , Humans , Leukocyte Common Antigens/metabolism , Mice , Transgenes , Transplantation, Heterologous , Virus Internalization , beta-Thalassemia/genetics , beta-Thalassemia/metabolism
9.
Curr Gene Ther ; 15(1): 64-81, 2015.
Article in English | MEDLINE | ID: mdl-25429463

ABSTRACT

A previously published clinical trial demonstrated the benefit of autologous CD34(+) cells transduced with a selfinactivating lentiviral vector (HPV569) containing an engineered ß-globin gene (ß(A-T87Q)-globin) in a subject with ß thalassemia major. This vector has been modified to increase transduction efficacy without compromising safety. In vitro analyses indicated that the changes resulted in both increased vector titers (3 to 4 fold) and increased transduction efficacy (2 to 3 fold). An in vivo study in which 58 ß-thalassemic mice were transplanted with vector- or mock-transduced syngenic bone marrow cells indicated sustained therapeutic efficacy. Secondary transplantations involving 108 recipients were performed to evaluate long-term safety. The six month study showed no hematological or biochemical toxicity. Integration site (IS) profile revealed an oligo/polyclonal hematopoietic reconstitution in the primary transplants and reduced clonality in secondary transplants. Tumor cells were detected in the secondary transplant mice in all treatment groups (including the control group), without statistical differences in the tumor incidence. Immunohistochemistry and quantitative PCR demonstrated that tumor cells were not derived from transduced donor cells. This comprehensive efficacy and safety data provided the basis for initiating two clinical trials with this second generation vector (BB305) in Europe and in the USA in patients with ß-thalassemia major and sickle cell disease.


Subject(s)
Anemia, Sickle Cell/therapy , Genetic Therapy/methods , Genetic Vectors , Lentivirus/genetics , beta-Thalassemia/therapy , Anemia, Sickle Cell/genetics , Animals , Antigens, CD34/metabolism , Computational Biology , DNA Damage , Disease Models, Animal , Female , Gene Expression , Gene Transfer Techniques , Hematopoietic Stem Cell Transplantation , Male , Mice , Mice, Inbred C57BL , beta-Thalassemia/genetics
10.
PLoS Pathog ; 9(4): e1003266, 2013.
Article in English | MEDLINE | ID: mdl-23592989

ABSTRACT

PKR-like endoplasmic reticulum (ER) kinase (PERK) is an ER-associated stress sensor protein which phosphorylates eukaryotic initiation factor 2α (eIF2α) to induce translation attenuation in response to ER stress. PERK is also a regulator of lipogenesis during adipocyte differentiation through activation of the cleavage of sterol regulatory element binding protein 1 (SREBP1), resulting in the upregulation of lipogenic enzymes. Our recent studies have shown that human cytomegalovirus (HCMV) infection in human fibroblasts (HF) induces adipocyte-like lipogenesis through the activation of SREBP1. Here, we report that PERK expression is highly increased in HCMV-infected cells and is necessary for HCMV growth. Depletion of PERK, using short hairpin RNA (shRNA), resulted in attenuation of HCMV growth, inhibition of lipid synthesis and reduction of lipogenic gene expression. Examination of the cleavage of SREBP proteins showed PERK depletion inhibited the cleavage of SREBP1, but not SREBP2, in HCMV-infected cells, suggesting different cleavage regulatory mechanisms for SREBP1 and 2. Further studies showed that the depletion of SREBP1, but not SREBP2, reduced lipid synthesis in HCMV infection, suggesting that activation of SREBP1 is sufficient to induce lipogenesis in HCMV infection. The reduction of lipid synthesis by PERK depletion can be partially restored by expressing a Flag-tagged nuclear form of SREBP1a. Our studies also suggest that the induction of PERK in HCMV-infected cells stimulates SREBP1 cleavage by reducing levels of Insig1 (Insulin inducible gene 1) protein; this occurs independent of the phosphorylation of eIF2α. Introduction of an exogenous Insig1-Myc into HCMV infected cells significantly reduced HCMV growth and lipid synthesis. Our data demonstrate that the induction of PERK during HCMV infection is necessary for full activation of lipogenesis; this effect appears to be mediated by limiting the levels of Insig1 thus freeing SREBP1-SCAP complexes for SREBP1 processing.


Subject(s)
Cytomegalovirus Infections/metabolism , Lipogenesis , Sterol Regulatory Element Binding Protein 1/metabolism , eIF-2 Kinase/metabolism , Cell Differentiation , Cells, Cultured , Cytomegalovirus/growth & development , Cytomegalovirus Infections/virology , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Stress , Enzyme Activation , Fibroblasts/virology , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Phosphorylation , RNA Interference , RNA, Small Interfering , Sterol Regulatory Element Binding Protein 2/metabolism , Unfolded Protein Response , eIF-2 Kinase/biosynthesis , eIF-2 Kinase/genetics
11.
Proc Natl Acad Sci U S A ; 109(12): 4544-9, 2012 Mar 20.
Article in English | MEDLINE | ID: mdl-22388749

ABSTRACT

Marine viruses impose a heavy mortality on their host bacteria, whereas at the same time the degree of viral resistance in marine bacteria appears to be high. Antagonistic coevolution--the reciprocal evolutionary change of interacting species--might reconcile these observations, if it leads to rapid and dynamic levels of viral resistance. Here we demonstrate the potential for extensive antagonistic coevolution between the ecologically important marine cyanobacterium Synechococcus and a lytic virus. In a 6-mo-long replicated chemostat experiment, Synechococcus sp. WH7803 and the virus (RIM8) underwent multiple coevolutionary cycles, leading to the rapid diversification of both host and virus. Over the course of the experiment, we detected between 4 and 13 newly evolved viral phenotypes (differing in host range) and between 4 and 11 newly evolved Synechococcus phenotypes (differing in viral resistance) in each chemostat. Genomic analysis of isolates identified several candidate genes in both the host and virus that might influence their interactions. Notably, none of the viral candidates were tail fiber genes, thought to be the primary determinants of host range in tailed bacteriophages, highlighting the difficulty in generalizing results from bacteriophage infecting γ-Proteobacteria. Finally, we show that pairwise virus-host coevolution may have broader community consequences; coevolution in the chemostat altered the sensitivity of Synechoccocus to a diverse suite of viruses, as well as the virus' ability to infect additional Synechococcus strains. Our results indicate that rapid coevolution may contribute to the generation and maintenance of Synechococcus and virus diversity and thereby influence viral-mediated mortality of these key marine bacteria.


Subject(s)
Bacteriophages/genetics , Synechococcus/genetics , Bacteriophages/physiology , Biological Evolution , Evolution, Molecular , Gammaproteobacteria/physiology , Genomics , Marine Biology , Models, Biological , Models, Genetic , Molecular Sequence Data , Phenotype , Seawater , Synechococcus/physiology , Synechococcus/virology , Viruses/genetics , Water Microbiology
12.
Adv Virus Res ; 80: 49-67, 2011.
Article in English | MEDLINE | ID: mdl-21762821

ABSTRACT

Recent studies of human cytomegalovirus (HCMV) infection have demonstrated that the virus significantly alters cellular metabolism, especially the utilization of glucose and glutamine. Glucose is not broken down by the tricarboxylic acid (TCA) cycle in infected cells; instead, it is used biosynthetically for fatty acid synthesis for membranes needed during the infection. In this chapter, we discuss the possibility that HCMV integrates its mechanisms for manipulating cellular signaling and stress responses to induce novel adipocyte-like differentiation in order to alter metabolism so that glucose can be used synthetically, that is, for fatty acids and lipids. This process diverts glucose from the TCA cycle and requires induction of enzymes that can convert glutamine to α-ketoglutarate to maintain the TCA cycle (anaplerosis). We discuss data proposing that the anaplerotic utilization of glutamine may be mediated, in part, by c-Myc activation, and the induction of adipocyte-like differentiation may result from the activation of the endoplasmic reticulum resident kinase PKR-like ER kinase. These alterations in metabolism during HCMV infection are comparable to those seen in many tumor cells. Indeed, the alterations in cellular signaling, stress responses, and metabolism that have been characterized could result in unexpected pathogenesis, potentially implicating HCMV as an agent or subtle cofactor in many maladies. Better understanding of HCMV's effects on cell signaling and metabolism will show how HCMV-mediated modifications of cellular processes relate to pathogenesis and will suggest novel avenues for antiviral therapy.


Subject(s)
Cytomegalovirus/metabolism , Cytomegalovirus/pathogenicity , Glucose/metabolism , Glutamine/metabolism , Host-Pathogen Interactions , Cell Line, Tumor , Endoplasmic Reticulum/metabolism , Fatty Acids/biosynthesis , Humans , Ketoglutaric Acids/metabolism , Pyruvic Acid/metabolism , Signal Transduction , Tricarboxylic Acids/metabolism
13.
J Virol ; 84(21): 11479-86, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20739513

ABSTRACT

The endoplasmic reticulum (ER) chaperone BiP (immunoglobulin binding protein) plays a major role in the control of the unfolded protein response. We have previously shown that BiP levels are dramatically increased during human cytomegalovirus (HCMV) infection, where BiP performs unique roles in viral assembly and egress. We show that BiP mRNA levels increase during infection due to activation of the BiP promoter by the major immediate-early (MIE) proteins. The BiP promoter, like other ER stress-activated promoters, contains endoplasmic reticulum stress elements (ERSEs), which are activated by unfolded protein response (UPR)-induced transcription factors. However, these elements are not needed for MIE protein-mediated transcriptional activation; thus, a virus-specific transcriptional activation mechanism is used. Transcriptional activation results in only a 3- to 4-fold increase in BiP mRNA, suggesting that additional mechanisms for BiP production are utilized. The BiP mRNA contains an internal ribosome entry site (IRES) which increases the level of BiP mRNA translation. We show that utilization of the BiP IRES is dramatically increased in HCMV-infected cells. Utilization of the BiP IRES can be activated by the La autoantigen, also called Sjögren's syndrome antigen B (SSB). We show that SSB/La levels are significantly increased during HCMV infection, and SSB/La depletion causes the loss of BiP IRES utilization and lowers endogenous BiP levels in infected cells. Our data show that BiP levels increase in HCMV-infected cells through the combination of increased BiP gene transcription mediated by the MIE proteins and increased BiP mRNA translation due to SSB/La-induced utilization of the BiP IRES.


Subject(s)
Cytomegalovirus/physiology , Heat-Shock Proteins/genetics , Protein Biosynthesis , Ribosomes/virology , Transcription, Genetic , Transcriptional Activation , Cytomegalovirus Infections , Endoplasmic Reticulum/chemistry , Endoplasmic Reticulum Chaperone BiP , Humans , Molecular Chaperones , Regulatory Sequences, Nucleic Acid
SELECTION OF CITATIONS
SEARCH DETAIL
...