Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 159
Filter
1.
Med Image Anal ; 99: 103334, 2024 Sep 03.
Article in English | MEDLINE | ID: mdl-39255733

ABSTRACT

Deep learning has been extensively applied in medical image reconstruction, where Convolutional Neural Networks (CNNs) and Vision Transformers (ViTs) represent the predominant paradigms, each possessing distinct advantages and inherent limitations: CNNs exhibit linear complexity with local sensitivity, whereas ViTs demonstrate quadratic complexity with global sensitivity. The emerging Mamba has shown superiority in learning visual representation, which combines the advantages of linear scalability and global sensitivity. In this study, we introduce MambaMIR, an Arbitrary-Masked Mamba-based model with wavelet decomposition for joint medical image reconstruction and uncertainty estimation. A novel Arbitrary Scan Masking (ASM) mechanism "masks out" redundant information to introduce randomness for further uncertainty estimation. Compared to the commonly used Monte Carlo (MC) dropout, our proposed MC-ASM provides an uncertainty map without the need for hyperparameter tuning and mitigates the performance drop typically observed when applying dropout to low-level tasks. For further texture preservation and better perceptual quality, we employ the wavelet transformation into MambaMIR and explore its variant based on the Generative Adversarial Network, namely MambaMIR-GAN. Comprehensive experiments have been conducted for multiple representative medical image reconstruction tasks, demonstrating that the proposed MambaMIR and MambaMIR-GAN outperform other baseline and state-of-the-art methods in different reconstruction tasks, where MambaMIR achieves the best reconstruction fidelity and MambaMIR-GAN has the best perceptual quality. In addition, our MC-ASM provides uncertainty maps as an additional tool for clinicians, while mitigating the typical performance drop caused by the commonly used dropout.

2.
Med Image Anal ; 98: 103306, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39163786

ABSTRACT

Positron emission tomography (PET) imaging is widely used in medical imaging for analyzing neurological disorders and related brain diseases. Usually, full-dose imaging for PET ensures image quality but raises concerns about potential health risks of radiation exposure. The contradiction between reducing radiation exposure and maintaining diagnostic performance can be effectively addressed by reconstructing low-dose PET (L-PET) images to the same high-quality as full-dose (F-PET). This paper introduces the Multi Pareto Generative Adversarial Network (MPGAN) to achieve 3D end-to-end denoising for the L-PET images of human brain. MPGAN consists of two key modules: the diffused multi-round cascade generator (GDmc) and the dynamic Pareto-efficient discriminator (DPed), both of which play a zero-sum game for n(n∈1,2,3) rounds to ensure the quality of synthesized F-PET images. The Pareto-efficient dynamic discrimination process is introduced in DPed to adaptively adjust the weights of sub-discriminators for improved discrimination output. We validated the performance of MPGAN using three datasets, including two independent datasets and one mixed dataset, and compared it with 12 recent competing models. Experimental results indicate that the proposed MPGAN provides an effective solution for 3D end-to-end denoising of L-PET images of the human brain, which meets clinical standards and achieves state-of-the-art performance on commonly used metrics.


Subject(s)
Brain , Positron-Emission Tomography , Humans , Positron-Emission Tomography/methods , Brain/diagnostic imaging , Signal-To-Noise Ratio , Radiation Dosage , Algorithms , Neural Networks, Computer , Imaging, Three-Dimensional/methods , Image Processing, Computer-Assisted/methods
3.
EJNMMI Phys ; 11(1): 76, 2024 Aug 30.
Article in English | MEDLINE | ID: mdl-39210079

ABSTRACT

BACKGROUND: Positron emission tomography (PET) traditionally uses coincident annihilation photons emitted from a positron interacting with an electron to localize cancer within the body. The formation of positronium (Ps), a bonded electron-positron pair, has not been utilized in clinical applications of PET due to the need to detect either the emission of a prompt gamma ray or the decay of higher-order coincident events. Assessment of the lifetime of the formed Ps, however, can potentially yield additional diagnostic information of the surrounding tissue because Ps properties vary due to void size and molecular composition. To assess the feasibility of measuring Ps lifetimes with a PET scanner, experiments were performed in a Biograph Vision Quadra (Siemens Healthineers). Quadra is a long-axial field-of-view (LA-FOV) PET scanner capable of producing list-mode data from single interaction events. RESULTS: Ortho-Ps (o-Ps) lifetimes were measured for quartz-glass and polycarbonate samples using a 22 Na positron source. Results produced o-Ps lifetimes of 1.538 ± 0.036 ns for the quartz glass and 1.927 ± 0.042 ns for the polycarbonate. Both o-Ps lifetimes were determined using a double-exponential fit to the time-difference distribution between the emission of a prompt gamma ray and the annihilation of the correlated positron. The measured values match within a single standard deviation of previously published results. The quartz-glass samples were additional measured with 82 Rb , 68 Ga and 124 I to validate the lifetime using clinically available sources. A double-exponential fit was initially chosen as a similar methodology to previously published works, however, an exponentially-modified Gaussian distribution fit to each lifetime more-accurately models the data. A Bayesian method was used to estimate the variables of the fit and o-Ps lifetime results are reported using this methodology for the three clinical isotopes: 1.59 ± 0.03 ns for 82 Rb , 1.58 ± 0.07 ns for 68 Ga and 1.62 ± 0.01 ns for 124 I . The impact of scatter and attenuation on the o-Ps lifetime was also assessed by analyzing a water-filled uniform cylinder (20 ϕ × 30 cm 3 ) with an added 82 Rb solution. Lifetimes were extracted for various regions of the cylinder and while there is a shape difference in the lifetime due to scatter, the extracted o-Ps lifetime of the water, 1.815 ± 0.013 ns, agrees with previously published results. CONCLUSION: Overall, the methodology presented in this manuscript demonstrates the repeatability of Ps lifetime measurements with clinically available isotopes in a commercially-available LA-FOV PET scanner. This validation work lays the foundation for future in-vivo patient scans with Quadra.

4.
Adv Sci (Weinh) ; 11(31): e2308307, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39166458

ABSTRACT

Aloperine (ALO), a quinolizidine-type alkaloid isolated from a natural Chinese herb, has shown promising antitumor effects. Nevertheless, its common mechanism of action and specific target remain elusive. Here, it is demonstrated that ALO inhibits the proliferation and migration of non-small cell lung cancer cell lines in vitro and the tumor development in several mouse tumor models in vivo. Mechanistically, ALO inhibits the fusion of autophagosomes with lysosomes and the autophagic flux, leading to the accumulation of sequestosome-1 (SQSTM1) and production of reactive oxygen species (ROS), thereby inducing tumor cell apoptosis and preventing tumor growth. Knockdown of SQSTM1 in cells inhibits ROS production and reverses ALO-induced cell apoptosis. Furthermore, VPS4A is identified as a direct target of ALO, and the amino acids F153 and D263 of VPS4A are confirmed as the binding sites for ALO. Knockout of VPS4A in H1299 cells demonstrates a similar biological effect as ALO treatment. Additionally, ALO enhances the efficacy of the anti-PD-L1/TGF-ß bispecific antibody in inhibiting LLC-derived subcutaneous tumor models. Thus, ALO is first identified as a novel late-stage autophagy inhibitor that triggers tumor cell death by targeting VPS4A.


Subject(s)
Autophagosomes , Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Lysosomes , Quinolizidines , Animals , Mice , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Humans , Autophagosomes/metabolism , Autophagosomes/drug effects , Lung Neoplasms/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Lysosomes/metabolism , Lysosomes/drug effects , Cell Line, Tumor , Quinolizidines/pharmacology , Disease Models, Animal , Vesicular Transport Proteins/metabolism , Vesicular Transport Proteins/genetics , Disease Progression , Cell Proliferation/drug effects , Autophagy/drug effects , Apoptosis/drug effects
5.
Article in English | MEDLINE | ID: mdl-38958680

ABSTRACT

PURPOSE: While sedation is routinely used in pediatric PET examinations to preserve diagnostic quality, it may result in side effects and may affect the radiotracer's biodistribution. This study aims to investigate the feasibility of sedation-free pediatric PET imaging using ultra-fast total-body (TB) PET scanners and deep learning (DL)-based attenuation and scatter correction (ASC). METHODS: This retrospective study included TB PET (uExplorer) imaging of 35 sedated pediatric patients under four years old to determine the minimum effective scanning time. A DL-based ASC method was applied to enhance PET quantification. Both quantitative and qualitative assessments were conducted to evaluate the image quality of ultra-fast DL-ASC PET. Five non-sedated pediatric patients were subsequently used to validate the proposed approach. RESULTS: Comparisons between standard 300-second and ultra-fast 15-second imaging, CT-ASC and DL-ASC ultra-fast 15-second images, as well as DL-ASC ultra-fast 15-second images in non-sedated and sedated patients, showed no significant differences in qualitative scoring, lesion detectability, and quantitative Standard Uptake Value (SUV) (P = ns). CONCLUSIONS: This study demonstrates that pediatric PET imaging can be effectively performed without sedation by combining ultra-fast imaging techniques with a DL-based ASC. This advancement in sedation-free ultra-fast PET imaging holds potential for broader clinical adoption.

6.
Brain Sci ; 14(7)2024 Jul 04.
Article in English | MEDLINE | ID: mdl-39061420

ABSTRACT

The differential diagnosis between atypical Parkinsonian syndromes may be challenging and critical. We aimed to proposed a radiomics-guided deep learning (DL) model to discover interpretable DL features and further verify the proposed model through the differential diagnosis of Parkinsonian syndromes. We recruited 1495 subjects for 18F-fluorodeoxyglucose positron emission tomography (18F-FDG PET) scanning, including 220 healthy controls and 1275 patients diagnosed with idiopathic Parkinson's disease (IPD), multiple system atrophy (MSA), or progressive supranuclear palsy (PSP). Baseline radiomics and two DL models were developed and tested for the Parkinsonian diagnosis. The DL latent features were extracted from the last layer and subsequently guided by radiomics. The radiomics-guided DL model outperformed the baseline radiomics approach, suggesting the effectiveness of the DL approach. DenseNet showed the best diagnosis ability (sensitivity: 95.7%, 90.1%, and 91.2% for IPD, MSA, and PSP, respectively) using retained DL features in the test dataset. The retained DL latent features were significantly associated with radiomics features and could be interpreted through biological explanations of handcrafted radiomics features. The radiomics-guided DL model offers interpretable high-level abstract information for differential diagnosis of Parkinsonian disorders and holds considerable promise for personalized disease monitoring.

7.
Article in English | MEDLINE | ID: mdl-38953933

ABSTRACT

PURPOSE: There is an unmet need for compounds to detect fibrillar forms of alpha-synuclein (αSyn) and 4-repeat tau, which are critical in many neurodegenerative diseases. Here, we aim to develop an efficient surface plasmon resonance (SPR)-based assay to facilitate the characterization of small molecules that can bind these fibrils. METHODS: SPR measurements were conducted to characterize the binding properties of fluorescent ligands/compounds toward recombinant amyloid-beta (Aß)42, K18-tau, full-length 2N4R-tau and αSyn fibrils. In silico modeling was performed to examine the binding pockets of ligands on αSyn fibrils. Immunofluorescence staining of postmortem brain tissue slices from Parkinson's disease patients and mouse models was performed with fluorescence ligands and specific antibodies. RESULTS: We optimized the protocol for the immobilization of Aß42, K18-tau, full-length 2N4R-tau and αSyn fibrils in a controlled aggregation state on SPR-sensor chips and for assessing their binding to ligands. The SPR results from the analysis of binding kinetics suggested the presence of at least two binding sites for all fibrils, including luminescent conjugated oligothiophenes, benzothiazole derivatives, nonfluorescent methylene blue and lansoprazole. In silico modeling studies for αSyn (6H6B) revealed four binding sites with a preference for one site on the surface. Immunofluorescence staining validated the detection of pS129-αSyn positivity in the brains of Parkinson's disease patients and αSyn preformed-fibril injected mice, 6E10-positive Aß in arcAß mice, and AT-8/AT-100-positivity in pR5 mice. CONCLUSION: SPR measurements of small molecules binding to Aß42, K18/full-length 2N4R-tau and αSyn fibrils suggested the existence of multiple binding sites. This approach may provide efficient characterization of compounds for neurodegenerative disease-relevant proteinopathies.

8.
Article in English | MEDLINE | ID: mdl-39042332

ABSTRACT

PURPOSE: Technological advances in instruments have greatly promoted the development of positron emission tomography (PET) scanners. State-of-the-art PET scanners such as uEXPLORER can collect PET images of significantly higher quality. However, these scanners are not currently available in most local hospitals due to the high cost of manufacturing and maintenance. Our study aims to convert low-quality PET images acquired by common PET scanners into images of comparable quality to those obtained by state-of-the-art scanners without the need for paired low- and high-quality PET images. METHODS: In this paper, we proposed an improved CycleGAN (IE-CycleGAN) model for unpaired PET image enhancement. The proposed method is based on CycleGAN, and the correlation coefficient loss and patient-specific prior loss were added to constrain the structure of the generated images. Furthermore, we defined a normalX-to-advanced training strategy to enhance the generalization ability of the network. The proposed method was validated on unpaired uEXPLORER datasets and Biograph Vision local hospital datasets. RESULTS: For the uEXPLORER dataset, the proposed method achieved better results than non-local mean filtering (NLM), block-matching and 3D filtering (BM3D), and deep image prior (DIP), which are comparable to Unet (supervised) and CycleGAN (supervised). For the Biograph Vision local hospital datasets, the proposed method achieved higher contrast-to-noise ratios (CNR) and tumor-to-background SUVmax ratios (TBR) than NLM, BM3D, and DIP. In addition, the proposed method showed higher contrast, SUVmax, and TBR than Unet (supervised) and CycleGAN (supervised) when applied to images from different scanners. CONCLUSION: The proposed unpaired PET image enhancement method outperforms NLM, BM3D, and DIP. Moreover, it performs better than the Unet (supervised) and CycleGAN (supervised) when implemented on local hospital datasets, which demonstrates its excellent generalization ability.

9.
Brain Pathol ; : e13288, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38982662

ABSTRACT

Abnormal alpha-synuclein (αSyn) and iron accumulation in the brain play an important role in Parkinson's disease (PD). Herein, we aim to visualize αSyn inclusions and iron deposition in the brains of M83 (A53T) mouse models of PD in vivo. The fluorescent pyrimidoindole derivative THK-565 probe was characterized by means of recombinant fibrils and brains from 10- to 11-month-old M83 mice. Concurrent wide-field fluorescence and volumetric multispectral optoacoustic tomography (vMSOT) imaging were subsequently performed in vivo. Structural and susceptibility weighted imaging (SWI) magnetic resonance imaging (MRI) at 9.4 T as well as scanning transmission x-ray microscopy (STXM) were performed to characterize the iron deposits in the perfused brains. Immunofluorescence and Prussian blue staining were further performed on brain slices to validate the detection of αSyn inclusions and iron deposition. THK-565 showed increased fluorescence upon binding to recombinant αSyn fibrils and αSyn inclusions in post-mortem brain slices from patients with PD and M83 mice. Administration of THK-565 in M83 mice showed higher cerebral retention at 20 and 40 min post-intravenous injection by wide-field fluorescence compared to nontransgenic littermate mice, in congruence with the vMSOT findings. SWI/phase images and Prussian blue indicated the accumulation of iron deposits in the brains of M83 mice, presumably in the Fe3+ form, as evinced by the STXM results. In conclusion, we demonstrated in vivo mapping of αSyn by means of noninvasive epifluorescence and vMSOT imaging and validated the results by targeting the THK-565 label and SWI/STXM identification of iron deposits in M83 mouse brains ex vivo.

10.
Theranostics ; 14(9): 3404-3422, 2024.
Article in English | MEDLINE | ID: mdl-38948052

ABSTRACT

Radiopharmaceutical therapy (RPT) is a rapidly developing field of nuclear medicine, with several RPTs already well established in the treatment of several different types of cancers. However, the current approaches to RPTs often follow a somewhat inflexible "one size fits all" paradigm, where patients are administered the same amount of radioactivity per cycle regardless of their individual characteristics and features. This approach fails to consider inter-patient variations in radiopharmacokinetics, radiation biology, and immunological factors, which can significantly impact treatment outcomes. To address this limitation, we propose the development of theranostic digital twins (TDTs) to personalize RPTs based on actual patient data. Our proposed roadmap outlines the steps needed to create and refine TDTs that can optimize radiation dose to tumors while minimizing toxicity to organs at risk. The TDT models incorporate physiologically-based radiopharmacokinetic (PBRPK) models, which are additionally linked to a radiobiological optimizer and an immunological modulator, taking into account factors that influence RPT response. By using TDT models, we envisage the ability to perform virtual clinical trials, selecting therapies towards improved treatment outcomes while minimizing risks associated with secondary effects. This framework could empower practitioners to ultimately develop tailored RPT solutions for subgroups and individual patients, thus improving the precision, accuracy, and efficacy of treatments while minimizing risks to patients. By incorporating TDT models into RPTs, we can pave the way for a new era of precision medicine in cancer treatment.


Subject(s)
Neoplasms , Precision Medicine , Radiopharmaceuticals , Humans , Precision Medicine/methods , Neoplasms/therapy , Neoplasms/radiotherapy , Radiopharmaceuticals/therapeutic use , Radiopharmaceuticals/pharmacokinetics
11.
Theranostics ; 14(9): 3623-3633, 2024.
Article in English | MEDLINE | ID: mdl-38948055

ABSTRACT

Introduction: Prostate Specific Membrane Antigen Positron Emission Tomography (PSMA-PET) is routinely used for the staging of patients with prostate cancer, but data on response assessment are sparse and primarily stem from metastatic castration-resistant prostate cancer (mCRPC) patients treated with PSMA radioligand therapy. Still, follow-up PSMA-PET is employed in earlier disease stages in case of clinical suspicion of disease persistence, recurrence or progression to decide if localized or systemic treatment is indicated. Therefore, the prognostic value of PSMA-PET derived tumor volumes in earlier disease stages (i.e., hormone-sensitive prostate cancer (HSPC) and non-[177Lu]Lu-PSMA-617 (LuPSMA) therapy castration resistant prostate cancer (CRPC)) are evaluated in this manuscript. Methods: A total number of 73 patients (6 primary staging, 42 HSPC, 25 CRPC) underwent two (i.e., baseline and follow-up, median interval: 379 days) whole-body [68Ga]Ga-PSMA-11 PET/CT scans between Nov 2014 and Dec 2018. Analysis was restricted to non-LuPSMA therapy patients. PSMA-PETs were retrospectively analyzed and primary tumor, lymph node-, visceral-, and bone metastases were segmented. Body weight-adjusted organ-specific and total tumor volumes (PSMAvol: sum of PET volumes of all lesions) were measured for baseline and follow-up. PSMAvol response was calculated as the absolute difference of whole-body tumor volumes. High metastatic burden (>5 metastases), RECIP 1.0 and PSMA-PET Progression Criteria (PPP) were determined. Survival data were sourced from the cancer registry. Results: The average number of tumor lesions per patient on the initial PET examination was 10.3 (SD 28.4). At baseline, PSMAvol was strongly associated with OS (HR 3.92, p <0.001; n = 73). Likewise, response in PSMAvol was significantly associated with OS (HR 10.48, p < 0.005; n = 73). PPP achieved significance as well (HR 2.19, p <0.05, n = 73). Patients with hormone sensitive disease and poor PSMAvol response (upper quartile of PSMAvol change) in follow-up had shorter outcome (p < 0.05; n = 42). PSMAvol in bones was the most relevant parameter for OS prognostication at baseline and for response assessment (HR 31.11 p < 0.001; HR 32.27, p < 0.001; n = 73). Conclusion: PPP and response in PSMAvol were significantly associated with OS in the present heterogeneous cohort. Bone tumor volume was the relevant miTNM region for OS prognostication. Future prospective evaluation of the performance of organ specific PSMAvol in more homogeneous cohorts seems warranted.


Subject(s)
Positron Emission Tomography Computed Tomography , Prostatic Neoplasms, Castration-Resistant , Humans , Male , Aged , Positron Emission Tomography Computed Tomography/methods , Prostatic Neoplasms, Castration-Resistant/diagnostic imaging , Prostatic Neoplasms, Castration-Resistant/pathology , Middle Aged , Follow-Up Studies , Gallium Radioisotopes , Retrospective Studies , Aged, 80 and over , Prostatic Neoplasms/diagnostic imaging , Prostatic Neoplasms/pathology , Glutamate Carboxypeptidase II/metabolism , Radiopharmaceuticals , Antigens, Surface/metabolism , Gallium Isotopes , Prognosis , Lutetium/therapeutic use , Positron-Emission Tomography/methods , Tumor Burden , Heterocyclic Compounds, 1-Ring/therapeutic use , Dipeptides/therapeutic use
12.
Semin Nucl Med ; 54(4): 460-469, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39013673

ABSTRACT

Radioligand therapy is an emerging and effective treatment option for various types of malignancies, but may be intricately linked to hematological side effects such as anemia, lymphopenia or thrombocytopenia. The safety and efficacy of novel theranostic agents, targeting increasingly complex targets, can be well served by comprehensive dosimetry. However, optimization in patient management and patient selection based on risk-factors predicting adverse events and built upon reliable dose-response relations is still an open demand. In this context, artificial intelligence methods, especially machine learning and deep learning algorithms, may play a crucial role. This review provides an overview of upcoming opportunities for integrating artificial intelligence methods into the field of dosimetry in nuclear medicine by improving bone marrow and blood dosimetry accuracy, enabling early identification of potential hematological risk-factors, and allowing for adaptive treatment planning. It will further exemplify inspirational success stories from neighboring disciplines that may be translated to nuclear medicine practices, and will provide conceptual suggestions for future directions. In the future, we expect artificial intelligence-assisted (predictive) dosimetry combined with clinical parameters to pave the way towards truly personalized theranostics in radioligand therapy.


Subject(s)
Artificial Intelligence , Bone Marrow , Precision Medicine , Radiometry , Humans , Precision Medicine/methods , Bone Marrow/radiation effects , Ligands
13.
Brain Sci ; 14(6)2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38928575

ABSTRACT

Clinical cognitive advancement within the Alzheimer's disease (AD) continuum is intimately connected with sustained accumulation of tau protein pathology. The biological brain age and its gap show great potential for pathological risk and disease severity. In the present study, we applied multivariable linear support vector regression to train a normative brain age prediction model using tau brain images. We further assessed the predicted biological brain age and its gap for patients within the AD continuum. In the AD continuum, evaluated pathologic tau binding was found in the inferior temporal, parietal-temporal junction, precuneus/posterior cingulate, dorsal frontal, occipital, and inferior-medial temporal cortices. The biological brain age gaps of patients within the AD continuum were notably higher than those of the normal controls (p < 0.0001). Significant positive correlations were observed between the brain age gap and global tau protein accumulation levels for mild cognitive impairment (r = 0.726, p < 0.001), AD (r = 0.845, p < 0.001), and AD continuum (r = 0.797, p < 0.001). The pathologic tau-based age gap was significantly linked to neuropsychological scores. The proposed pathologic tau-based biological brain age model could track the tau protein accumulation trajectory of cognitive impairment and further provide a comprehensive quantification index for the tau accumulation risk.

14.
J Neurol Sci ; 462: 123079, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38878650

ABSTRACT

BACKGROUND: Reactive astrocytes play an important role in the development of Alzheimer's disease and primary tauopathies. Here, we aimed to investigate the relationships between reactive astrocytes. Microgliosis and glucose metabolism with Tau and amyloid beta pathology by using multi-tracer imaging in widely used tauopathy and familial Alzheimer's disease mouse models. RESULTS: Positron emission tomography imaging using [18F]PM-PBB3 (tau), [18F]florbetapir (amyloid-beta), [18F]SMBT-1 (monoamine oxidase-B), [18F]DPA-714 (translocator protein) and [18F]fluorodeoxyglucose was carried out in 3- and 7-month-old rTg4510 tau mice, 5 × FAD familial Alzheimer's disease mice and wild-type mice. Immunofluorescence staining was performed to validate the pathological distribution in the mouse brain after in vivo imaging. We found increased regional levels of [18F]PM-PBB3, [18F]SMBT-1, and [18F]DPA-714 and hypoglucose metabolism in the brains of 7-month-old rTg4510 mice compared to age-matched wild-type mice. Increased [18F]SMBT-1 uptake was observed in the brains of 3, 7-month-old 5 × FAD mice, with elevated regional [18F]florbetapir and [18F]DPA-714 uptakes in the brains of 7-month-old 5 × FAD mice, compared to age-matched wild-type mice. Positive correlations were shown between [18F]SMBT-1 and [18F]PM-PBB3, [18F]DPA-714 and [18F]PM-PBB3 in rTg4510 mice, and between [18F]florbetapir and [18F]DPA-714 SUVRs in 5 × FAD mice. CONCLUSION: In summary, these findings provide in vivo evidence that reactive astrocytes, microglial activation, and cerebral hypoglucose metabolism are associated with tau and amyloid pathology development in animal models of tauopathy and familial Alzheimer's disease.


Subject(s)
Alzheimer Disease , Astrocytes , Brain , Disease Models, Animal , Mice, Transgenic , Positron-Emission Tomography , Tauopathies , Animals , Alzheimer Disease/diagnostic imaging , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Alzheimer Disease/genetics , Astrocytes/metabolism , Positron-Emission Tomography/methods , Tauopathies/diagnostic imaging , Tauopathies/metabolism , Tauopathies/pathology , Mice , Brain/diagnostic imaging , Brain/metabolism , Brain/pathology , Fluorine Radioisotopes , Male , Amyloid beta-Peptides/metabolism , Humans , tau Proteins/metabolism , tau Proteins/genetics , Fluorodeoxyglucose F18 , Radiopharmaceuticals
16.
Eur J Nucl Med Mol Imaging ; 51(11): 3450-3460, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38724653

ABSTRACT

BACKGROUND AND OBJECTIVE: Treatment planning through the diagnostic dimension of theranostics provides insights into predicting the absorbed dose of RPT, with the potential to individualize radiation doses for enhancing treatment efficacy. However, existing studies focusing on dose prediction from diagnostic data often rely on organ-level estimations, overlooking intra-organ variations. This study aims to characterize the intra-organ theranostic heterogeneity and utilize artificial intelligence techniques to localize them, i.e. to predict voxel-wise absorbed dose map based on pre-therapy PET. METHODS: 23 patients with metastatic castration-resistant prostate cancer treated with [177Lu]Lu-PSMA I&T RPT were retrospectively included. 48 treatment cycles with pre-treatment PET imaging and at least 3 post-therapeutic SPECT/CT imaging were selected. The distribution of PET tracer and RPT dose was compared for kidney, liver and spleen, characterizing intra-organ heterogeneity differences. Pharmacokinetic simulations were performed to enhance the understanding of the correlation. Two strategies were explored for pre-therapy voxel-wise dosimetry prediction: (1) organ-dose guided direct projection; (2) deep learning (DL)-based distribution prediction. Physical metrics, dose volume histogram (DVH) analysis, and identity plots were applied to investigate the predicted absorbed dose map. RESULTS: Inconsistent intra-organ patterns emerged between PET imaging and dose map, with moderate correlations existing in the kidney (r = 0.77), liver (r = 0.5), and spleen (r = 0.58) (P < 0.025). Simulation results indicated the intra-organ pharmacokinetic heterogeneity might explain this inconsistency. The DL-based method achieved a lower average voxel-wise normalized root mean squared error of 0.79 ± 0.27%, regarding to ground-truth dose map, outperforming the organ-dose guided projection (1.11 ± 0.57%) (P < 0.05). DVH analysis demonstrated good prediction accuracy (R2 = 0.92 for kidney). The DL model improved the mean slope of fitting lines in identity plots (199% for liver), when compared to the theoretical optimal results of the organ-dose approach. CONCLUSION: Our results demonstrated the intra-organ heterogeneity of pharmacokinetics may complicate pre-therapy dosimetry prediction. DL has the potential to bridge this gap for pre-therapy prediction of voxel-wise heterogeneous dose map.


Subject(s)
Antigens, Surface , Glutamate Carboxypeptidase II , Prostatic Neoplasms, Castration-Resistant , Radiometry , Radiopharmaceuticals , Humans , Male , Glutamate Carboxypeptidase II/metabolism , Radiopharmaceuticals/therapeutic use , Radiopharmaceuticals/pharmacokinetics , Prostatic Neoplasms, Castration-Resistant/radiotherapy , Prostatic Neoplasms, Castration-Resistant/diagnostic imaging , Aged , Retrospective Studies , Precision Medicine/methods , Middle Aged , Positron-Emission Tomography/methods , Positron Emission Tomography Computed Tomography/methods
17.
Article in English | MEDLINE | ID: mdl-38805089

ABSTRACT

PURPOSE: This study aimed to comprehensively explore the different metabolic connectivity topological changes in MTLE and NTLE, as well as their association with surgical outcomes. METHODS: This study enrolled a cohort of patients with intractable MTLE and NTLE. Each individual's metabolic connectome, as determined by Kullback-Leibler divergence similarity estimation for the [18F]FDG PET image, was employed to conduct a comprehensive analysis of the cerebral metabolic network. Alterations in network connectivity were assessed by extracting and evaluating the strength of edge and weighted connectivity. By utilizing these two connectivity strength metrics with the cerebellum, we explored the network properties of connectivity and its association with prognosis in surgical patients. RESULTS: Both MTLE and NTLE patients exhibited substantial alterations in the connectivity of the metabolic network at the edge and nodal levels (p < 0.01, FDR corrected). The key disparity between MTLE and NTLE was observed in the cerebellum. In MTLE, there was a predominance of increased connectivity strength in the cerebellum. Whereas, a decrease in cerebellar connectivity was identified in NTLE. It was found that in MTLE, higher edge connectivity and weighted connectivity strength in the contralateral cerebellar hemisphere correlated with improved surgical outcomes. Conversely, in NTLE, a higher edge metabolic connectivity strength in the ipsilateral cerebellar hemisphere suggested a worse surgical prognosis. CONCLUSION: The cerebellum exhibits distinct topological characteristics in the metabolic networks between MTLE and NTLE. The hyper- or hypo-metabolic connectivity in the cerebellum may be a prognostic biomarker of surgical prognosis, which might aid in therapeutic decision-making for TLE individuals.

19.
ACS Chem Neurosci ; 15(11): 2112-2120, 2024 06 05.
Article in English | MEDLINE | ID: mdl-38776461

ABSTRACT

Neuroinflammation plays an important role in Alzheimer's disease and primary tauopathies. The aim of the current study was to map [18F]GSK1482160 for imaging of purinergic P2X7R in Alzheimer's disease and primary tauopathy mouse models. Small animal PET was performed using [18F]GSK1482160 in widely used mouse models of Alzheimer's disease (APP/PS1, 5×FAD, and 3×Tg), 4-repeat tauopathy (rTg4510) mice, and age-matched wild-type mice. Increased uptake of [18F]GSK1482160 was observed in the brains of 7-month-old rTg4510 mice compared to wild-type mice and compared to 3-month-old rTg4510 mice. A positive correlation between hippocampal tau [18F]APN-1607 and [18F]GSK1482160 uptake was found in rTg4510 mice. No significant differences in the uptake of [18F]GSK1482160 was observed for APP/PS1 mice, 5×FAD mice, or 3×Tg mice. Immunofluorescence staining further indicated the distribution of P2X7Rs in the brains of 7-month-old rTg4510 mice with accumulation of tau inclusion. These findings provide in vivo imaging evidence for an increased level of P2X7R in the brains of tauopathy mice.


Subject(s)
Positron-Emission Tomography , Receptors, Purinergic P2X7 , Tauopathies , Animals , Mice , Alzheimer Disease/metabolism , Alzheimer Disease/diagnostic imaging , Brain/metabolism , Brain/diagnostic imaging , Disease Models, Animal , Fluorine Radioisotopes , Mice, Transgenic , Positron-Emission Tomography/methods , Receptors, Purinergic P2X7/metabolism , tau Proteins/metabolism , Tauopathies/diagnostic imaging , Tauopathies/metabolism
20.
Hum Brain Mapp ; 45(7): e26689, 2024 May.
Article in English | MEDLINE | ID: mdl-38703095

ABSTRACT

Tau pathology and its spatial propagation in Alzheimer's disease (AD) play crucial roles in the neurodegenerative cascade leading to dementia. However, the underlying mechanisms linking tau spreading to glucose metabolism remain elusive. To address this, we aimed to examine the association between pathologic tau aggregation, functional connectivity, and cascading glucose metabolism and further explore the underlying interplay mechanisms. In this prospective cohort study, we enrolled 79 participants with 18F-Florzolotau positron emission tomography (PET), 18F-fluorodeoxyglucose PET, resting-state functional, and anatomical magnetic resonance imaging (MRI) images in the hospital-based Shanghai Memory Study. We employed generalized linear regression and correlation analyses to assess the associations between Florzolotau accumulation, functional connectivity, and glucose metabolism in whole-brain and network-specific manners. Causal mediation analysis was used to evaluate whether functional connectivity mediates the association between pathologic tau and cascading glucose metabolism. We examined 22 normal controls and 57 patients with AD. In the AD group, functional connectivity was associated with Florzolotau covariance (ß = .837, r = 0.472, p < .001) and glucose covariance (ß = 1.01, r = 0.499, p < .001). Brain regions with higher tau accumulation tend to be connected to other regions with high tau accumulation through functional connectivity or metabolic connectivity. Mediation analyses further suggest that functional connectivity partially modulates the influence of tau accumulation on downstream glucose metabolism (mediation proportion: 49.9%). Pathologic tau may affect functionally connected neurons directly, triggering downstream glucose metabolism changes. This study sheds light on the intricate relationship between tau pathology, functional connectivity, and downstream glucose metabolism, providing critical insights into AD pathophysiology and potential therapeutic targets.


Subject(s)
Alzheimer Disease , Fluorodeoxyglucose F18 , Magnetic Resonance Imaging , Nerve Net , Positron-Emission Tomography , tau Proteins , Humans , Alzheimer Disease/diagnostic imaging , Alzheimer Disease/metabolism , Alzheimer Disease/physiopathology , Male , Female , Aged , tau Proteins/metabolism , Middle Aged , Nerve Net/diagnostic imaging , Nerve Net/metabolism , Nerve Net/physiopathology , Glucose/metabolism , Connectome , Prospective Studies , Brain/diagnostic imaging , Brain/metabolism , Brain/physiopathology , Aged, 80 and over
SELECTION OF CITATIONS
SEARCH DETAIL