Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Immunity ; 56(12): 2790-2802.e6, 2023 Dec 12.
Article in English | MEDLINE | ID: mdl-38091952

ABSTRACT

Neurodegenerative diseases (ND) are characterized by progressive loss of neuronal function. Mechanisms of ND pathogenesis are incompletely understood, hampering the development of effective therapies. Langerhans cell histiocytosis (LCH) is an inflammatory neoplastic disorder caused by hematopoietic progenitors expressing mitogen-activated protein kinase (MAPK)-activating mutations that differentiate into senescent myeloid cells that drive lesion formation. Some individuals with LCH subsequently develop progressive and incurable neurodegeneration (LCH-ND). Here, we showed that LCH-ND was caused by myeloid cells that were clonal with peripheral LCH cells. Circulating BRAFV600E+ myeloid cells caused the breakdown of the blood-brain barrier (BBB), enhancing migration into the brain parenchyma where they differentiated into senescent, inflammatory CD11a+ macrophages that accumulated in the brainstem and cerebellum. Blocking MAPK activity and senescence programs reduced peripheral inflammation, brain parenchymal infiltration, neuroinflammation, neuronal damage and improved neurological outcome in preclinical LCH-ND. MAPK activation and senescence programs in circulating myeloid cells represent targetable mechanisms of LCH-ND.


Subject(s)
Histiocytosis, Langerhans-Cell , Proto-Oncogene Proteins B-raf , Humans , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , Histiocytosis, Langerhans-Cell/genetics , Histiocytosis, Langerhans-Cell/pathology , Histiocytosis, Langerhans-Cell/therapy , Brain/metabolism , Myeloid Cells/metabolism , Cell Differentiation
2.
Br J Haematol ; 204(5): 1882-1887, 2024 May.
Article in English | MEDLINE | ID: mdl-38501390

ABSTRACT

Optimal therapeutic approaches for advanced Langerhans cell histiocytosis (LCH) are not known. We assessed the safety and efficacy of combined chemotherapy with MAPK pathway inhibition in 10 patients with refractory systemic disease and/or LCH-associated neurodegeneration. Overall response rate was 9/10 (90%) for the entire cohort: 5/5 (100%) for patients with systemic disease and 6/7 (86%) for patients with central nervous system disease. BRAFV600E+ peripheral blood fraction decreased in 5/6 (83%). Toxicities included fever, skin rash, myalgias, neuropathy, cytopenias and hypocalcaemia. Prospective trials are required to optimize combination strategies, determine potential to achieve cure and compare outcomes to chemotherapy or MAPK inhibitor monotherapy.


Subject(s)
Histiocytosis, Langerhans-Cell , MAP Kinase Signaling System , Protein Kinase Inhibitors , Female , Humans , Male , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Histiocytosis, Langerhans-Cell/drug therapy , MAP Kinase Signaling System/drug effects , Protein Kinase Inhibitors/therapeutic use , Protein Kinase Inhibitors/adverse effects , Protein Kinase Inhibitors/administration & dosage , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins B-raf/genetics , Recurrence , Treatment Outcome
3.
Br J Haematol ; 204(5): 1888-1893, 2024 May.
Article in English | MEDLINE | ID: mdl-38501389

ABSTRACT

Over 50% of patients with systemic LCH are not cured with front-line therapies, and data to guide salvage options are limited. We describe 58 patients with LCH who were treated with clofarabine. Clofarabine monotherapy was active against LCH in this cohort, including heavily pretreated patients with a systemic objective response rate of 92.6%, higher in children (93.8%) than adults (83.3%). BRAFV600E+ variant allele frequency in peripheral blood is correlated with clinical responses. Prospective multicentre trials are warranted to determine optimal dosing, long-term efficacy, late toxicities, relative cost and patient-reported outcomes of clofarabine compared to alternative LCH salvage therapy strategies.


Subject(s)
Clofarabine , Histiocytosis, Langerhans-Cell , Humans , Clofarabine/therapeutic use , Clofarabine/administration & dosage , Histiocytosis, Langerhans-Cell/drug therapy , Male , Female , Adult , Adolescent , Child , Middle Aged , Child, Preschool , Young Adult , Aged , Recurrence , Proto-Oncogene Proteins B-raf/genetics , Infant , Treatment Outcome , Salvage Therapy , Adenine Nucleotides/therapeutic use , Adenine Nucleotides/administration & dosage , Adenine Nucleotides/adverse effects , Arabinonucleosides/therapeutic use , Arabinonucleosides/administration & dosage , Arabinonucleosides/adverse effects
4.
J Emerg Med ; 62(1): 64-71, 2022 01.
Article in English | MEDLINE | ID: mdl-34544622

ABSTRACT

BACKGROUND: Ultrasound inferior vena cava (IVC) diameter has been shown to decrease in response to hemorrhage. IVC diameter cut points to identify moderate and severe blood loss have not been established. OBJECTIVES: This study sought to find ultrasound IVC diameter cut points to identify moderate and severe hemorrhage and assess the performance of these cut points vs. vital sign abnormalities. METHODS: This is a secondary analysis of data from a study that described changes in vital signs and sonographic measurements of the IVC during a lower body negative pressure model of hemorrhage. Using receiver operator curve analyses, optimal cut points for identifying moderate and severe hemorrhage were identified. The ability of these cut points to identify hemorrhage in patients with no vital sign abnormalities was then assessed. RESULTS: In both long- and short-axis views, maximum and minimum IVC diameters (IVCmax and IVCmin) were significantly lower than baseline in severe blood loss. The optimal cut point for IVCmax in both axes was found to be ≤ 0.8 cm. This cut point is able to distinguish between no blood loss vs. moderate blood loss, and no blood loss vs. severe blood loss. The optimal cut point for IVCmin was variable between axes and blood loss severity. IVC diameter cut points obtained were able to identify hemorrhage in patients with no vital sign abnormalities. CONCLUSION: An ultrasound IVCmax of ≤ 0.8 cm may be useful in identifying moderate and severe hemorrhage before vital sign abnormalities are evident.


Subject(s)
Abdomen , Vena Cava, Inferior , Hemorrhage/etiology , Humans , Ultrasonography , Vena Cava, Inferior/diagnostic imaging , Vital Signs
5.
Prehosp Emerg Care ; 25(3): 341-346, 2021.
Article in English | MEDLINE | ID: mdl-32628063

ABSTRACT

INTRODUCTION: Inferior vena cava (IVC) diameter decreases under conditions of hypovolemia. Point-of-care ultrasound (POCUS) may be useful to emergently assess IVC diameter. This study tested the hypothesis that ultrasound measurements of IVC diameter decreases during severe simulated blood loss. METHODS: Blood loss was simulated in 14 healthy men (22 ± 2 years) using lower body negative pressure (LBNP). Pressure within the LBNP chamber was reduced 10 mmHg of LBNP every four minutes until participants experienced pre-syncopal symptoms or until 80 mmHg of LBNP was completed. IVC diameter was imaged with POCUS using B-mode in the long and short axis views between minutes two and four of each stage. RESULTS: Maximum IVC diameter in the long axis view was lower than baseline (1.5 ± 0.4 cm) starting at -20 mmHg of LBNP (1.0 ± 0.3 cm; p < 0.01) and throughout LBNP (p < 0.01). The minimum IVC diameter in the long axis view was lower than baseline (0.9 ± 0.3 cm) at -20 mmHg of LBNP (0.5 ± 0.3 cm; p < 0.01) and throughout LBNP (p < 0.01). Maximum IVC diameter in the short axis view was lower than baseline (0.9 ± 0.2 cm) at 40 mmHg of LBNP (0.6 ± 0.2; p = 0.01) and the final LBNP stage (0.6 ± 0.2 cm; p < 0.01). IVC minimum diameter in the short axis view was lower than baseline (0.5 ± 0.2 cm) at the final LBNP stage (0.3 ± 0.2 cm; p = 0.01). CONCLUSION: These data demonstrate that IVC diameter decreases prior to changes in traditional vital signs during simulated blood loss. Further study is needed to determine the view and diameter threshold that most accurate for identifying hemorrhage requiring emergent intervention.


Subject(s)
Emergency Medical Services , Hypovolemia , Hemorrhage/diagnostic imaging , Humans , Lower Body Negative Pressure , Male , Vena Cava, Inferior/diagnostic imaging
6.
Cancer ; 124(12): 2607-2620, 2018 06 15.
Article in English | MEDLINE | ID: mdl-29624648

ABSTRACT

BACKGROUND: Central nervous system Langerhans cell histiocytosis (CNS-LCH) brain involvement may include mass lesions and/or a neurodegenerative disease (LCH-ND) of unknown etiology. The goal of this study was to define the mechanisms of pathogenesis that drive CNS-LCH. METHODS: Cerebrospinal fluid (CSF) biomarkers including CSF proteins and extracellular BRAFV600E DNA were analyzed in CSF from patients with CNS-LCH lesions compared with patients with brain tumors and other neurodegenerative conditions. Additionally, the presence of BRAFV600E was tested in peripheral mononuclear blood cells (PBMCs) as well as brain biopsies from LCH-ND patients, and the response to BRAF-V600E inhibitor was evaluated in 4 patients with progressive disease. RESULTS: Osteopontin was the only consistently elevated CSF protein in patients with CNS-LCH compared with patients with other brain pathologies. BRAFV600E DNA was detected in CSF of only 2/20 (10%) cases, both with LCH-ND and active lesions outside the CNS. However, BRAFV600E+ PBMCs were detected with significantly higher frequency at all stages of therapy in LCH patients who developed LCH-ND. Brain biopsies of patients with LCH-ND demonstrated diffuse perivascular infiltration by BRAFV600E+ cells with monocyte phenotype (CD14+ CD33+ CD163+ P2RY12- ) and associated osteopontin expression. Three of 4 patients with LCH-ND treated with BRAF-V600E inhibitor experienced significant clinical and radiologic improvement. CONCLUSION: In LCH-ND patients, BRAFV600E+ cells in PBMCs and infiltrating myeloid/monocytic cells in the brain is consistent with LCH-ND as an active demyelinating process arising from a mutated hematopoietic precursor from which LCH lesion CD207+ cells are also derived. Therapy directed against myeloid precursors with activated MAPK signaling may be effective for LCH-ND. Cancer 2018;124:2607-20. © 2018 American Cancer Society.


Subject(s)
Brain Neoplasms/diagnosis , Histiocytosis, Langerhans-Cell/diagnosis , Neurodegenerative Diseases/diagnosis , Osteopontin/cerebrospinal fluid , Proto-Oncogene Proteins B-raf/genetics , Adolescent , Adult , Biomarkers/blood , Biomarkers/cerebrospinal fluid , Biopsy , Brain/pathology , Brain Neoplasms/cerebrospinal fluid , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Child , Child, Preschool , Diagnosis, Differential , Female , Hematopoietic Stem Cells/pathology , Histiocytosis, Langerhans-Cell/cerebrospinal fluid , Histiocytosis, Langerhans-Cell/genetics , Histiocytosis, Langerhans-Cell/pathology , Humans , Infant , Infant, Newborn , Leukocytes, Mononuclear/pathology , MAP Kinase Signaling System , Male , Neurodegenerative Diseases/cerebrospinal fluid , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/pathology , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Retrospective Studies , Young Adult
8.
Blood Adv ; 7(14): 3725-3734, 2023 07 25.
Article in English | MEDLINE | ID: mdl-37042921

ABSTRACT

Overall survival after reduced-intensity conditioning (RIC) allogeneic hematopoietic cell transplantation (HCT) using alemtuzumab, fludarabine, and melphalan is associated with high rates of mixed chimerism (MC) and secondary graft failure (GF). We hypothesized that peritransplantation alemtuzumab levels or specific patterns of inflammation would predict these risks. We assessed samples from the Bone Marrow Transplant Clinical Trials Network 1204 (NCT01998633) to study the impact of alemtuzumab levels and cytokine patterns on MC and impending or established secondary GF (defined as donor chimerism <5% after initial engraftment and/or requirement of cellular intervention). Thirty-three patients with hemophagocytic lymphohistiocytosis (n = 25) and other IEIs (n = 8) who underwent HCTs with T-cell-replete grafts were included. Patients with day 0 alemtuzumab levels ≤0.32 µg/mL had a markedly lower incidence of MC, 14.3%, vs 90.9% in patients with levels >0.32 µg/mL (P = .008). Impending or established secondary GF was only observed in patients with day 0 alemtuzumab levels >0.32 µg/mL (P = .08). Unexpectedly, patients with impending or established secondary GF had lower CXCL9 levels. The cumulative incidence of impending or established secondary GF in patients with a day 14+ CXCL9 level ≤2394 pg/mL (day 14+ median) was 73.6% vs 0% in patients with a level >2394 pg/mL (P = .002). CXCL9 levels inversely correlated with alemtuzumab levels. These data suggest a model in which higher levels of alemtuzumab at day 0 deplete donor T cells, inhibit the graft-versus-marrow reaction (thereby suppressing CXCL9 levels), and adversely affect sustained engraftment in the nonmyeloablative HCT setting. This trial was registered at www.clinicaltrials.gov as #NCT01998633.


Subject(s)
Antibodies, Monoclonal, Humanized , Hematopoietic Stem Cell Transplantation , Humans , Alemtuzumab/therapeutic use , Antibodies, Monoclonal, Humanized/therapeutic use , Melphalan/therapeutic use , Hematopoietic Stem Cell Transplantation/adverse effects , Tissue Donors , Chemokine CXCL9
9.
bioRxiv ; 2023 Oct 11.
Article in English | MEDLINE | ID: mdl-37873371

ABSTRACT

Neurodegenerative diseases (ND) are characterized by progressive loss of neuronal function. Mechanisms of ND pathogenesis are incompletely understood, hampering the development of effective therapies. Langerhans cell histiocytosis (LCH) is an inflammatory neoplastic disorder caused by hematopoietic progenitors expressing MAPK activating mutations that differentiate into senescent myeloid cells that drive lesion formation. Some patients with LCH subsequently develop progressive and incurable neurodegeneration (LCH-ND). Here, we show that LCH-ND is caused by myeloid cells that are clonal with peripheral LCH cells. We discovered that circulating BRAF V600E + myeloid cells cause the breakdown of the blood-brain barrier (BBB), enhancing migration into the brain parenchyma where they differentiate into senescent, inflammatory CD11a + macrophages that accumulate in the brainstem and cerebellum. Blocking MAPK activity and senescence programs reduced parenchymal infiltration, neuroinflammation, neuronal damage and improved neurological outcome in preclinical LCH-ND. MAPK activation and senescence programs in circulating myeloid cells represent novel and targetable mechanisms of ND.

10.
Blood Adv ; 5(17): 3457-3467, 2021 09 14.
Article in English | MEDLINE | ID: mdl-34461635

ABSTRACT

Hemophagocytic lymphohistiocytosis (HLH) is a syndrome characterized by pathologic immune activation in which prompt recognition and initiation of immune suppression is essential for survival. Children with HLH have many overlapping clinical features with critically ill children with sepsis and systemic inflammatory response syndrome (SIRS) in whom alternative therapies are indicated. To determine whether plasma biomarkers could differentiate HLH from other inflammatory conditions and to better define a core inflammatory signature of HLH, concentrations of inflammatory plasma proteins were compared in 40 patients with HLH to 47 pediatric patients with severe sepsis or SIRS. Fifteen of 135 analytes were significantly different in HLH plasma compared with SIRS/sepsis, including increased interferon-γ (IFN-γ)-regulated chemokines CXCL9, CXCL10, and CXCL11. Furthermore, a 2-analyte plasma protein classifier including CXCL9 and interleukin-6 was able to differentiate HLH from SIRS/sepsis. Gene expression in CD8+ T cells and activated monocytes from blood were also enriched for IFN-γ pathway signatures in peripheral blood cells from patients with HLH compared with SIRS/sepsis. This study identifies differential expression of inflammatory proteins as a diagnostic strategy to identify critically ill children with HLH, and comprehensive unbiased analysis of inflammatory plasma proteins and global gene expression demonstrates that IFN-γ signaling is uniquely elevated in HLH. In addition to demonstrating the ability of diagnostic criteria for HLH and sepsis or SIRS to identify groups with distinct inflammatory patterns, results from this study support the potential for prospective evaluation of inflammatory biomarkers to aid in diagnosis of and optimizing therapeutic strategies for children with distinctive hyperinflammatory syndromes.


Subject(s)
Lymphohistiocytosis, Hemophagocytic , Sepsis , Child , Diagnosis, Differential , Humans , Interferon-gamma , Lymphohistiocytosis, Hemophagocytic/diagnosis , Proteome , Sepsis/diagnosis , Systemic Inflammatory Response Syndrome/diagnosis
11.
Nat Med ; 27(5): 851-861, 2021 05.
Article in English | MEDLINE | ID: mdl-33958797

ABSTRACT

Langerhans cell histiocytosis (LCH) is a potentially fatal condition characterized by granulomatous lesions with characteristic clonal mononuclear phagocytes (MNPs) harboring activating somatic mutations in mitogen-activated protein kinase (MAPK) pathway genes, most notably BRAFV600E. We recently discovered that the BRAFV600E mutation can also affect multipotent hematopoietic progenitor cells (HPCs) in multisystem LCH disease. How the BRAFV600E mutation in HPCs leads to LCH is not known. Here we show that enforced expression of the BRAFV600E mutation in early mouse and human multipotent HPCs induced a senescence program that led to HPC growth arrest, apoptosis resistance and a senescence-associated secretory phenotype (SASP). SASP, in turn, promoted HPC skewing toward the MNP lineage, leading to the accumulation of senescent MNPs in tissue and the formation of LCH lesions. Accordingly, elimination of senescent cells using INK-ATTAC transgenic mice, as well as pharmacologic blockade of SASP, improved LCH disease in mice. These results identify senescent cells as a new target for the treatment of LCH.


Subject(s)
Cellular Senescence/genetics , Histiocytosis, Langerhans-Cell/genetics , Histiocytosis, Langerhans-Cell/pathology , Langerhans Cells/pathology , Proto-Oncogene Proteins B-raf/genetics , Animals , Apoptosis/genetics , Cell Proliferation/genetics , Cellular Senescence/drug effects , Cytokines/metabolism , Hematopoietic Stem Cells/pathology , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors
12.
Blood Adv ; 4(1): 87-99, 2020 01 14.
Article in English | MEDLINE | ID: mdl-31899802

ABSTRACT

Langerhans cell histiocytosis (LCH) is a myeloproliferative disorder that is characterized by the inflammatory lesions with pathogenic CD1a+CD207+ dendritic cells (DCs). BRAFV600E and other somatic activating MAPK gene mutations have been identified in differentiating bone marrow and blood myeloid cells, but the origin of the LCH lesion CD1a+CD207+ DCs and mechanisms of lesion formation remain incompletely defined. To identify candidate LCH CD1a+CD207+ DC precursor populations, gene-expression profiles of LCH lesion CD1a+CD207+ DCs were first compared with established gene signatures from human myeloid cell subpopulations. Interestingly, the CD1c+ myeloid DC (mDC) gene signature was most enriched in the LCH CD1a+CD207+ DC transcriptome. Additionally, the BRAFV600E allele was not only localized to CD1a+CD207- DCs and CD1a+CD207+ DCs, but it was also identified in CD1c+ mDCs in LCH lesions. Transcriptomes of CD1a+CD207- DCs were nearly indistinguishable from CD1a+CD207+ DCs (both CD1a+CD207low and CD1a+CD207high subpopulations). Transcription profiles of LCH lesion CD1a+CD207+ DCs and peripheral blood CD1c+ mDCs from healthy donors were compared to identify potential LCH DC-specific biomarkers: HLA-DQB2 expression was significantly increased in LCH lesion CD1a+CD207+ DCs compared with circulating CD1c+ mDCs from healthy donors. HLA-DQB2 antigen was identified on LCH lesion CD1a+CD207- DCs and CD1a+CD207+ DCs as well as on CD1c+(CD1a+CD207-) mDCs, but it was not identified in any other lesion myeloid subpopulations. HLA-DQB2 expression was specific to peripheral blood of patients with BRAFV600E+ peripheral blood mononuclear cells, and HLA-DQB2+CD1c+ blood cells were highly enriched for the BRAFV600E in these patients. These data support a model in which blood CD1c+HLA-DQB2+ mDCs with activated ERK migrate to lesion sites where they differentiate into pathogenic CD1a+CD207+ DCs.


Subject(s)
Histiocytosis, Langerhans-Cell , Leukocytes, Mononuclear , Antigens, CD/genetics , Antigens, CD1/genetics , Biomarkers , Dendritic Cells , Glycoproteins , Histiocytosis, Langerhans-Cell/diagnosis , Histiocytosis, Langerhans-Cell/genetics , Humans , Lectins, C-Type/genetics , Mannose-Binding Lectins/genetics , Myeloid Cells
13.
Cancers (Basel) ; 12(12)2020 Dec 02.
Article in English | MEDLINE | ID: mdl-33276546

ABSTRACT

Hodgkin lymphoma (HL) histopathology is characterized by rare malignant Reed-Sternberg cells among an inflammatory infiltrate. We hypothesized that characteristics of inflammation in pediatric HL lesions would be reflected by the levels of inflammatory cytokines or chemokines in pre-therapy plasma of children with HL. The study objectives were to better define the inflammatory pre-therapy plasma proteome and identify plasma biomarkers associated with extent of disease and clinical outcomes in pediatric HL. Pre-therapy plasma samples were obtained from pediatric subjects with newly diagnosed HL and healthy pediatric controls. Plasma concentrations of 135 cytokines/chemokines were measured with the Luminex platform. Associations between protein concentration and disease characteristics were determined using multivariate permutation tests with false discovery control. Fifty-six subjects with HL (mean age: 13 years, range 3-18) and 47 controls were analyzed. The cytokine/chemokine profiles of subjects with HL were distinct from controls, and unique cytokines/chemokines were associated with high-risk disease (IL-10, TNF-α, IFN-γ, IL-8) and slow early response (CCL13, IFN-λ1, IL-8). TNFSF10 was significantly elevated among those who ultimately relapsed and was significantly associated with worse event-free survival. These biomarkers could be incorporated into biologically based risk stratification to optimize outcomes and minimize toxicities in pediatric HL.

14.
J Exp Med ; 215(1): 319-336, 2018 01 02.
Article in English | MEDLINE | ID: mdl-29263218

ABSTRACT

Langerhans cell histiocytosis (LCH) is an inflammatory myeloid neoplasia characterized by granulomatous lesions containing pathological CD207+ dendritic cells (DCs) with constitutively activated mitogen-activated protein kinase (MAPK) pathway signaling. Approximately 60% of LCH patients harbor somatic BRAFV600E mutations localizing to CD207+ DCs within lesions. However, the mechanisms driving BRAFV600E+ LCH cell accumulation in lesions remain unknown. Here we show that sustained extracellular signal-related kinase activity induced by BRAFV600E inhibits C-C motif chemokine receptor 7 (CCR7)-mediated DC migration, trapping DCs in tissue lesions. Additionally, BRAFV600E increases expression of BCL2-like protein 1 (BCL2L1) in DCs, resulting in resistance to apoptosis. Pharmacological MAPK inhibition restores migration and apoptosis potential in a mouse LCH model, as well as in primary human LCH cells. We also demonstrate that MEK inhibitor-loaded nanoparticles have the capacity to concentrate drug delivery to phagocytic cells, significantly reducing off-target toxicity. Collectively, our results indicate that MAPK tightly suppresses DC migration and augments DC survival, rendering DCs in LCH lesions trapped and resistant to cell death.


Subject(s)
Cell Movement/physiology , Dendritic Cells/metabolism , Dendritic Cells/physiology , Histiocytosis, Langerhans-Cell/metabolism , Langerhans Cells/metabolism , MAP Kinase Signaling System/physiology , Proto-Oncogene Proteins B-raf/metabolism , Animals , Apoptosis/physiology , Histiocytosis, Langerhans-Cell/pathology , Humans , Langerhans Cells/physiology , Mice , Mice, Inbred C57BL , Mutation/physiology , Phagocytosis/physiology
SELECTION OF CITATIONS
SEARCH DETAIL