Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Exp Neurol ; 329: 113316, 2020 07.
Article in English | MEDLINE | ID: mdl-32304749

ABSTRACT

The development and translation of cell therapies have been hindered by an inability to predict and evaluate their efficacy after transplantation. Using an experimental autoimmune encephalomyelitis (EAE) mouse model of multiple sclerosis (MS), we studied attenuation of the diffuse injury characteristic of EAE and MS by transplanted glial-restricted precursor cells (GRPs). We assessed the potential of on-resonance variable delay multiple pulse (onVDMP) chemical exchange saturation transfer (CEST) MRI to visualize this attenuation. Allogeneic GRPs transplanted in the motor cortex or lateral ventricles attenuated paralysis in EAE mice and attenuated differences compared to naïve mice in onVDMP CEST signal 5 days after transplantation near the transplantation site. Histological analysis revealed that transplanted GRPs co-localized with attenuated astrogliosis. Hence, diffuse injury-sensitive onVDMP CEST MRI may complement conventional MRI to locate and monitor tissue regions responsive to GRP therapy.


Subject(s)
Cell Transplantation/methods , Encephalomyelitis, Autoimmune, Experimental/diagnostic imaging , Encephalomyelitis, Autoimmune, Experimental/therapy , Magnetic Resonance Imaging/methods , Neuroglia/transplantation , Animals , Encephalomyelitis, Autoimmune, Experimental/metabolism , Luminescent Measurements/methods , Mice , Mice, Transgenic , Neuroglia/metabolism
2.
Tomography ; 1(2): 91-97, 2015 12.
Article in English | MEDLINE | ID: mdl-26740972

ABSTRACT

Magnetic labeling of stem cells enables their non-invasive detection by magnetic resonance imaging (MRI). Practically, most MRI studies have been limited to visualization of local engraftment as other sources of endogenous hypointense contrast complicate the interpretation of systemic (whole body) cell distribution. In addition, MRI cell tracking is inherently non-quantitative in nature. We report here on the potential of magnetic particle imaging (MPI) as a novel tomographic technique for non-invasive hot spot imaging and quantification of stem cells using superparamagnetic iron oxide (SPIO) tracers. Neural and mesenchymal stem cells, representing small and larger cell bodies, were labeled with three different SPIO tracer formulations, including two preparations that have previously been used in clinical MRI cell tracking studies (Feridex® and Resovist®). Magnetic particle spectroscopy (MPS) measurements demonstrated a linear correlation between MPI signal and iron content, for both homogeneous solutions of free particles in solution and for internalized and aggregated particles in labeled cells over a wide range of concentrations. The overall MP signal ranged from 1×10-3 - 3×10-4 Am2/g Fe, which was equivalent to 2×10-14 - 1×10-15 Am2 per cell, indicating that cell numbers can be quantified with MPI analogous to the use of radiotracers in nuclear medicine or fluorine tracers in 19F MRI. When SPIO-labeled cells were transplanted in mouse brain, they could be readily detected by MPI at a detection threshold of about 5×104 cells, with MPI/MRI overlays showing an excellent agreement between the hypointense MRI areas and MPI hot spots. The calculated tissue MPI signal ratio for 100,000 vs. 50,000 implanted cells was 2.08. Hence, MPI has potential to be further developed for quantitative and easy-to-interpret, tracer-based non-invasive imaging of cells, preferably with MRI as an adjunct anatomical imaging modality.

3.
Cell Transplant ; 23(2): 253-62, 2014 Feb.
Article in English | MEDLINE | ID: mdl-23294627

ABSTRACT

Allografts continue to be used in clinical neurotransplantation studies; hence, it is crucial to understand the mechanisms that govern allograft tolerance. We investigated the impact of transplantation site within the brain on graft survival. Mouse [Friend leukemia virus, strain B (FVB)] glial precursors, transfected with luciferase, were injected (3 × 10(5)) into the forceps minor (FM) or striatum (STR). Immunodeficient rag2(-/-) and immunocompetent BALB/c mice were used as recipients. Magnetic resonance imaging (MRI) confirmed that cells were precisely deposited at the selected coordinates. The graft viability was assessed noninvasively with bioluminescent imaging (BLI) for a period of 16 days. Regardless of implantation site, all grafts (n = 10) deposited in immunodeficient animals revealed excellent survival. In contrast, immunocompetent animals only accepted grafts at the STR site (n = 10), whereas all the FM grafts were rejected (n = 10). To investigate the factors that led to rejection of FM grafts, with acceptance of STR grafts, another group of animals (n = 19) was sacrificed during the prerejection period, on day 5. Near-infrared fluorescence imaging with IRDye 800CW-polyethylene glycol probe displayed similar blood-brain barrier disruption at both graft locations. The morphological distribution of FM grafts was cylindrical, parallel to the needle track, whereas cells transplanted into the STR accumulated along the border between the STR and the corpus callosum. There was significantly less infiltration by both innate and adaptive immune cells in the STR grafts, especially along the calloso-striatal border. With allograft survival being dependent on the transplantation site, the anatomical coordinates of the graft target should always be taken into account as it may determine the success or failure of therapy.


Subject(s)
Brain/metabolism , Transplantation, Homologous/methods , Animals , Cell Survival/physiology , Cells, Cultured , Central Nervous System/cytology , Graft Survival/physiology , Immunohistochemistry , Magnetic Resonance Imaging , Male , Mice
4.
Adv Drug Deliv Rev ; 62(11): 1080-93, 2010 Aug 30.
Article in English | MEDLINE | ID: mdl-20800081

ABSTRACT

Cellular therapy promises to revolutionize medicine, by restoring tissue and organ function, and combating key disorders including cancer. As with all major developments, new tools must be introduced to allow optimization. For cell therapy, the key tool is in vivo imaging for real time assessment of parameters such as cell localization, numbers and viability. Such data is critical to modulate and tailor the therapy for each patient. In this review, we discuss recent work in the field of imaging cell therapies in the clinic, including preclinical work where clinical examples are not yet available. Clinical trials in which transferred cells were imaged using magnetic resonance imaging (MRI), nuclear scintigraphy, single photon emission computed tomography (SPECT), and positron emission tomography (PET) are evaluated from an imaging perspective. Preclinical cell tracking studies that focus on fluorescence and bioluminescence imaging are excluded, as these modalities are generally not applicable to clinical cell tracking. In this review, we assess the advantages and drawbacks of the various imaging techniques available, focusing on immune cells, particularly dendritic cells. Both strategies of prelabeling cells before transplant and the use of an injectable label to target cells in situ are covered. Finally, we discuss future developments, including the emergence of multimodal imaging technology for cell tracking from the preclinical to the clinical realm.


Subject(s)
Cell Tracking/methods , Cell Transplantation/diagnostic imaging , Cell Transplantation/methods , Diagnostic Imaging/methods , Animals , Dendritic Cells/diagnostic imaging , Dendritic Cells/transplantation , Humans , Radionuclide Imaging , T-Lymphocytes/diagnostic imaging , T-Lymphocytes/transplantation
5.
Cytotherapy ; 11(2): 245-55, 2009.
Article in English | MEDLINE | ID: mdl-19152153

ABSTRACT

From 4 to 5 April 2008, international experts met for the second time in Tubingen, Germany, to present and discuss the latest proceedings in research on non-hematopoietic stem cells (NHSC). This report presents issues of basic research including characterization, isolation, good manufacturing practice (GMP)-like production and imaging as well as clinical applications focusing on the regenerative and immunomodulatory capacities of NHSC.


Subject(s)
Adult Stem Cells/cytology , Biomedical Research , Embryonic Stem Cells/cytology , Immunotherapy, Adoptive , Neoplasms/therapy , Adult Stem Cells/physiology , Biomedical Research/ethics , Biomedical Research/methods , Biomedical Research/trends , Cell Culture Techniques , Cell Differentiation , Cell Movement , Cell Transdifferentiation , Diagnostic Imaging , Embryonic Stem Cells/physiology , Gene Expression Profiling , Germany , Hematopoietic Stem Cell Mobilization , Humans , Regenerative Medicine/trends , Stem Cell Niche
7.
Magn Reson Med ; 58(2): 261-9, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17654572

ABSTRACT

LacZ-transfected C17.2 neural stem cells (NSCs) were labeled with the superparamagnetic iron oxide formulation Feridex prior to ICV injection in shi/shi neonates. Feridex labeling did not alter cell differentiation in vitro and in vivo. Initially, MR images obtained at 11.7T correlated closely to NSC distribution as assessed with anti-dextran and anti-beta-galactosidase double-fluorescent immunostaining. However, at 6 days postgrafting there was already a pronounced mismatch between the hypointense MR signal and the histologically determined cell distribution, with a surprisingly sharp cutoff rather than a gradual decrease of signal. Positive in vivo BrdU labeling of NSCs showed that significant cell replication occurred post-transplantation, causing rapid dilution of Feridex particles between mother and daughter cells toward undetectable levels. Neural differentiation experiments demonstrated asymmetric cell division, explaining the observed sharp cutoff. At later time points (2 weeks), the mismatch further increased by the presence of non-cell-associated Feridex particles resulting from active excretion or cell death. These results are a first demonstration of the inability of MRI to track rapidly dividing and self-renewing, asymmetrically dividing SCs. Therefore, MR cell tracking should only be applied for nonproliferating cells or short-term monitoring of highly-proliferative cells, with mitotic symmetry or asymmetry being important for determining its applicability.


Subject(s)
Magnetic Resonance Imaging/methods , Stem Cell Transplantation , Animals , Brain , Cell Division , Cell Line , Demyelinating Diseases/pathology , Demyelinating Diseases/therapy , Dextrans , Ferrosoferric Oxide , Image Processing, Computer-Assisted , Iron , Magnetite Nanoparticles , Mice, Neurologic Mutants , Oxides , Stem Cells/cytology , Transfection
8.
Mol Pharm ; 3(5): 531-8, 2006.
Article in English | MEDLINE | ID: mdl-17009852

ABSTRACT

Alginate-poly-L-lysine-alginate (APA) microcapsules have been explored as vehicles for therapeutic drug and cell delivery. The permselectivity of these capsules provides a unique means of controlled drug release and immunoisolation of encapsulated cells. Immunoisolation is especially attractive as it abrogates the need for chronic immunosuppressive therapy and opens up the possibility for the delivery of numerous cell sources including xenogeneic grafts. APA microcapsules containing cellular therapeutics have proven effective in the short-term treatment of a wide range of diseases requiring enzyme or endocrine replacement therapy, including type I diabetes. If these microcapsules could be noninvasively monitored with X-ray imaging modalities (i.e., fluoroscopy, CT, and digital subtraction angiography), questions such as the ideal transplantation site, the best means of delivery, and the long-term survival of grafts could be better addressed. We have developed two novel alginate-based radiopaque microcapsule formulations containing either barium sulfate (Ba X-Caps) or bismuth sulfate (Bi X-Caps). As compared to conventional, nonradiopaque APA capsules, Ba X-Caps and Bi X-Caps containing human cadaveric islets resulted in a decrease in cellular viability of less than 5% up to 14 days after encapsulation. Both radiopaque capsules were found to be permeable to lectins < or =75 kDa, but were impermeable to lectins > or =120 kDa, thus ensuring the blockage of the penetration of antibodies while allowing free diffusion of insulin and nutrients. The glucose-responsive insulin secretion of the radiopaque encapsulated human islets was found to be unaltered compared to that of unlabeled controls, with human C-peptide levels ranging from 3.21 to 2.87 (Ba X-Caps) and 3.23 to 2.87 (Bi X-Caps) ng/islet at 7 and 14 days postencapsulation, respectively. Using fluoroscopy, both Ba X-Caps and Bi X-Caps could be readily visualized as single radiopaque entities in vitro. Furthermore, following transplantation in vivo in mice and rabbits, single capsules could be identified with no significant change in contrast for at least 2 weeks. This study represents the first attempt at making radiopaque microcapsules for X-ray guided delivery and imaging of cellular therapeutics. While human cadaveric islets were used as a proof-of-principle, these radiopaque capsules may have wide ranging therapeutic applications for a variety of cell types.


Subject(s)
Alginates/pharmacokinetics , Tomography, X-Ray Computed/methods , Alginates/chemistry , Alginates/therapeutic use , Animals , Barium Sulfate/chemistry , Capsules , Delayed-Action Preparations/chemistry , Delayed-Action Preparations/pharmacokinetics , Delayed-Action Preparations/therapeutic use , Female , Glucose/metabolism , Glucuronic Acid/chemistry , Glucuronic Acid/pharmacokinetics , Glucuronic Acid/therapeutic use , Hexuronic Acids/chemistry , Hexuronic Acids/pharmacokinetics , Hexuronic Acids/therapeutic use , Humans , Insulin/metabolism , Islets of Langerhans/cytology , Islets of Langerhans/diagnostic imaging , Islets of Langerhans/metabolism , Islets of Langerhans Transplantation/methods , Mice , Rabbits , Transplantation, Heterologous
9.
Exp Neurol ; 201(1): 49-59, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16764862

ABSTRACT

Neural precursor cell (NPC) transplantation is a promising strategy for treatment of CNS injuries and neurodegenerative disorders because of potential for cell replacement. An important element of future clinical applications is development of a non-invasive procedure to follow NPC fate. We show that neuronal-restricted precursors (NRPs) and glial-restricted precursors (GRPs), NPCs with lineage restrictions for neurons and glia, respectively, can be labeled in vitro with the superparamagnetic iron oxide contrast agent Feridex. Following engraftment into intact adult spinal cord, labeled cells robustly survived in white and gray matter and migrated selectively along white matter tracts up to 5 mm. Localization of cells was reliably established using ex vivo magnetic resonance imaging of spinal cords. Imaging coincided with histological detection of iron and the human alkaline phosphatase transgene in most grafting sites, including the stream of migrating cells. Following transplantation, magnetically labeled cells exhibited mature morphologies and differentiated into neurons, astrocytes, and oligodendrocytes, similar to grafts of unlabeled NRPs and GRPs. Interestingly, Feridex-labeled cells, but not unlabeled cells, induced influx of ED1-positive macrophages/microglia. Small numbers of these phagocytic cells took up iron from grafted cells, while the majority of Feridex label was found in transplanted cells. We conclude that Feridex labeling does not inhibit NPC differentiation and can be used to reliably localize NPCs by MRI following engraftment into adult CNS, with the possible exception of areas of rapidly proliferating cells. The present results are relevant for MR-guided clinical application of transplantation strategies in treatment of spinal cord injury and other CNS pathologies.


Subject(s)
Cell Lineage , Neurons/cytology , Spinal Cord/cytology , Stem Cells/cytology , Alkaline Phosphatase , Animals , Animals, Genetically Modified , Cell Differentiation , Dextrans , Female , Ferrosoferric Oxide , Fluorescent Antibody Technique , GPI-Linked Proteins , Humans , Iron/chemistry , Iron/metabolism , Isoenzymes/genetics , Isoenzymes/metabolism , Macrophages/cytology , Macrophages/metabolism , Magnetic Resonance Imaging/methods , Magnetite Nanoparticles , Microscopy, Confocal , Neuroglia/cytology , Neuroglia/metabolism , Neurons/metabolism , Neurons/transplantation , Oxides/chemistry , Oxides/metabolism , Rats , Rats, Inbred F344 , Rats, Sprague-Dawley , Spinal Cord/metabolism , Spinal Cord/surgery , Stem Cell Transplantation/methods , Stem Cells/metabolism , Time Factors
10.
Magn Reson Med ; 54(4): 769-74, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16161115

ABSTRACT

For cellular MR imaging, conventional approaches to intracellular magnetic labeling of nonphagocytic cells rely on the use of secondary compounds such as transfection agents and prolonged incubation of cells. Magnetoelectroporation (MEP) was investigated as an alternative method to achieve instant (<1 s) endosomal labeling with the FDA-approved formulation Feridex, without the need for adjunct agents or initiating cell cultures. While MEP was harmful at higher voltages or pulse durations, the procedure could be properly calibrated using a pulse of 130 V and 17 ms. Labeling was demonstrated for stem cells from mice, rats, and humans; the uptake of iron was in the picogram range and comparable to values obtained using transfection agents. MEP-labeled stem cells exhibited an unaltered viability, proliferation, and mitochondrial metabolic rate. Labeled mesenchymal stem cells (MSCs) and neural stem cells (NSCs) differentiated into adipogenic, osteogenic, and neural lineages in an identical fashion as unlabeled cells, while containing Feridex particles as demonstrated by double immunofluorescent staining. MEP-labeled NSCs proliferated normally following intrastriatal transplantation and could be readily detected by MR imaging in vivo. As MEP circumvents the use of secondary agents, obviating the need for clinical approval, MEP labeling may be ideally suitable for bedside implementation.


Subject(s)
Drug Delivery Systems/methods , Electroporation/methods , Image Enhancement/methods , Iron , Magnetic Resonance Imaging/methods , Magnetics , Oxides , Stem Cells/cytology , Animals , Cell Differentiation , Cells, Cultured , Contrast Media , Dextrans , Ferrosoferric Oxide , Humans , Magnetite Nanoparticles , Mice , Rats , Staining and Labeling/methods
11.
Neuroimage ; 26(3): 744-54, 2005 Jul 01.
Article in English | MEDLINE | ID: mdl-15955483

ABSTRACT

The application of stem cells as delivery vehicles opens up the opportunity for targeting therapeutic proteins to the damaged or degenerating central nervous system. Neural stem cell (NSC) lines have been shown to engraft, differentiate and correct certain central nervous system diseases. The present study was performed to test the ability of magnetic resonance imaging (MRI) in detecting transplanted NSCs under conditions of limited migration in the normal adult mouse brain versus widespread migration when the cells are transplanted neonatally. The C17.2 NSC line was labeled in vitro with superparamagnetic iron oxide (SPIO) particles and the labeled cells were implanted intracranially. Serial in vivo gradient echo MR imaging was performed using a 4.7 T horizontal bore magnet. High resolution ex vivo images of the isolated brains were performed at 9.4 T, and the presence of iron was correlated with Prussian blue staining in histological sections. Adult animals injected with SPIO-labeled stem cells exhibited hypointense regions near the injection site that were observed up to 32 days after injection. In neonatally transplanted animals, MR signal intensity from transplanted NSCs was not apparent in in vivo imaging but ex vivo MR images revealed small hypointense regions throughout the brain including the olfactory bulbs, cortex and the cerebellum, reflecting the wide distribution of the engrafted cells. These regions were correlated with Prussian blue staining, which confirmed the presence of SPIO particles inside the engrafted cells. We have shown that MRI is capable of differentiating localized and widespread engraftment of C17.2 stem cells in the central nervous system.


Subject(s)
Brain/cytology , Neurons/transplantation , Stem Cell Transplantation , Animals , Animals, Newborn , Cell Line , Cerebral Cortex/anatomy & histology , Cerebral Cortex/cytology , Clone Cells , Ferric Compounds , Ferrocyanides , Hippocampus/anatomy & histology , Hippocampus/cytology , Image Processing, Computer-Assisted , Magnetic Resonance Imaging , Mice , Mice, Inbred C3H , Models, Anatomic , Tissue Fixation , beta-Galactosidase/metabolism
12.
Magn Reson Med ; 51(6): 1163-72, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15170836

ABSTRACT

There is growing interest in delivering cellular agents to infarcted myocardium to prevent postinfarction left ventricular remodeling. MRI can be effectively used to differentiate infarcted from healthy myocardium. MR-guided delivery of cellular agents/therapeutics is appealing because the therapeutics can be precisely targeted to the desired location within the infarct. In this study, a steerable intramyocardial injection catheter that can be actively tracked under MRI was developed and tested. The components of the catheter were arranged to form a loopless RF antenna receiver coil that enabled active tracking. Feasibility studies were performed in canine and porcine myocardial infarction models. Myocardial delayed-enhancement (MDE) imaging identified the infarcted myocardium, and real-time MRI was used to guide left ventricular catheterization from a carotid artery approach. The distal 35 cm of the catheter was seen under MRI with a bright signal at the distal tip of the catheter. The catheter was steered into position, the distal tip was apposed against the infarct, the needle was advanced, and a bolus of MR contrast agent and tissue marker dye was injected intramyocardially, as confirmed by imaging and postmortem histology. A pilot study involving intramyocardial delivery of magnetically labeled stem cells demonstrated the utility of the active injection catheter system.


Subject(s)
Cardiac Catheterization , Injections, Intralesional , Magnetic Resonance Imaging , Mesenchymal Stem Cell Transplantation , Myocardial Infarction/therapy , Myocardium , Animals , Cardiac Catheterization/instrumentation , Catheterization , Contrast Media , Dextrans , Dogs , Equipment Design , Ferrosoferric Oxide , Iron , Magnetite Nanoparticles , Myocardial Infarction/pathology , Myocardium/pathology , Oxides , Phantoms, Imaging , Swine
13.
Nucl Med Commun ; 24(11): 1149-54, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14569169

ABSTRACT

Mesenchymal stem cells (MSCs) have shown therapeutic potential if successfully delivered to the intended site of myocardial infarction. The purpose of this pilot study was to test the feasibility of 111In oxine labelling of MSCs and single photon emission computed tomography (SPECT) imaging after intravenous administration in a porcine model of myocardial infarction. Adult farm pigs (n=2) were subjected to closed chest experimental myocardial infarction. 111In oxine labelled MSCs (1 x 10(7) to 2 x 10(7) cells) were infused intravenously, and SPECT imaging was performed initially and on days 1, 2, 7 and 14. High quality SPECT images were obtained through 2 weeks of imaging. High initial MSC localization occurred in the lungs and slow progressive accumulation occurred in the liver, spleen and bone marrow. Renal activity was mild and persistent throughout imaging. No appreciable accumulation occurred in the myocardium. It is concluded that 111In oxine radiolabelling of MSCs is feasible, and in vivo imaging with SPECT provides a non-invasive method for sequentially monitoring cell trafficking with good spatial resolution. Because intravenous administration of MSCs results in significant lung activity that obscures the assessment of myocardial cell trafficking, alternative routes of administration should be investigated for this application.


Subject(s)
Cardiac Surgical Procedures/methods , Heart/diagnostic imaging , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/diagnostic imaging , Organometallic Compounds , Oxyquinoline/analogs & derivatives , Stem Cells/diagnostic imaging , Tomography, Emission-Computed, Single-Photon/methods , Animals , Cell Movement , Feasibility Studies , Injections, Intravenous , Isotope Labeling/methods , Mesenchymal Stem Cells/physiology , Organ Specificity , Pilot Projects , Swine
14.
Magn Reson Med ; 50(2): 275-82, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12876703

ABSTRACT

The purpose of this study was to investigate the changes in electrostatic and magnetic resonance (MR) properties observed when MR contrast agents (CAs) (Feridex, MION-46L, or G5-dendrimer-DOTA-Gd) are combined with transfection agents (TAs) under various conditions for use as a CA-TA complex basis for cellular labeling and MRI. CAs were incubated with various classes of TAs for 0-48 hr in solutions of varying concentrations and pH values. NMR relaxation rates (1/T(1), 1/T(2)), MRI and zeta potential (ZP) of CA-TA solutions were measured. TAs decreased the 1/T(1) and 1/T(2) of G5-DOTA-Gd, Feridex, and MION-46L by 0-95%. Altering the pH of G5-DOTA-Gd-TA decreased the T(1)-weighted signal intensity (SI) on MRI from 0 to 78%. Measured ZP values for G5-DOTA-Gd, Feridex, and MION-46L were -51, -41, and -2.0 mV, respectively. The TA LV had a negative ZP, while the other TAs had ZPs ranging from +20 to +65 mV. The alteration of the ZP and NMR relaxivities of the MR CAs, Feridex, MION-46L, and G5-DOTA-Gd by TAs has been demonstrated. These results enhance our understanding of the relationship between electrostatic and MR properties.


Subject(s)
Cells, Cultured , Contrast Media , Magnetic Resonance Imaging , Contrast Media/chemistry , Dextrans , Electrochemistry , Ferrosoferric Oxide , Gadolinium , Heterocyclic Compounds/chemistry , Hydrogen-Ion Concentration , Iron/chemistry , Lipids/chemistry , Magnetite Nanoparticles , Organometallic Compounds/chemistry , Oxides/chemistry , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...