20 results on '"Nizzero, Sara"'
Search Results
2. Enhanced tumor control and survival in preclinical models with adoptive cell therapy preceded by low-dose radiotherapy.
- Author
-
Puebla-Osorio, Nahum, Fowlkes, Natalie Wall, Barsoumian, Hampartsoum B., Xega, Kristina, Srivastava, Gitika, Kettlun-Leyton, Claudia, Nizzero, Sara, Voss, Tiffany, Riad, Thomas S., Wong, Christina, Huang, Ailing, Yun Hu, Mitchell, Joylise, Mingee Kim, Rafiq, Zahid, Kewen He, Sezen, Duygu, Hsu, Ethan, Masrorpour, Fatemeh, and Maleki, Aurian
- Subjects
CHIMERIC antigen receptors ,ATOMIC force microscopy ,TREATMENT effectiveness ,TUMOR growth ,SURVIVAL rate - Abstract
Introduction: Effective infiltration of chimeric antigen receptor T (CAR-T) cells into solid tumors is critical for achieving a robust antitumor response and improving therapeutic outcomes. While CAR-T cell therapies have succeeded in hematologic malignancies, their efficacy in solid tumors remains limited due to poor tumor penetration and an immunosuppressive tumor microenvironment. This study aimed to evaluate the potential of low-dose radiotherapy (LDRT) administered before T-cell therapy to enhance the antitumor effect by promoting CAR-T cell infiltration. We hypothesized that combining LDRT with T-cell therapy would improve tumor control and survival compared to either treatment alone. Methods: We investigated this hypothesis using two NSG mouse models bearing GSU or CAPAN-2 solid tumors. The mice were treated with engineered CAR-T cells targeting guanyl cyclase-C (GCC) or mesothelin as monotherapy or in combination with LDRT. Additionally, we extended this approach to a C57BL/6 mouse model implanted with MC38-gp100+ cells, followed by adoptive transfer of pmel+ T cells before and after LDRT. Tumor growth and survival outcomes were monitored in all models. Furthermore, we employed atomic force microscopy (AFM) in a small cohort to assess the effects of radiotherapy on tumor stiffness and plasticity, exploring the role of tumor nanomechanics as a potential biomarker for treatment efficacy. Results: Our results demonstrated enhanced tumor control and prolonged survival in mice treated with LDRT followed by T-cell therapy across all models. The combination of LDRT with CAR-T or pmel+ T-cell therapy led to superior tumor suppression and survival compared to monotherapy, highlighting the synergistic impact of the combined approach. Additionally, AFM analysis revealed significant changes in tumor stiffness and plasticity in response to LDRT, suggesting that the nanomechanical properties of the tumor may be predictive of therapeutic response. Discussion: The findings of this study highlight the transformative potential of incorporating LDRT as a precursor to adoptive T-cell therapy in solid tumors. By promoting CAR-T and pmel+ T-cell infiltration into the tumor microenvironment, LDRT enhanced tumor control and improved survival outcomes, offering a promising strategy to overcome the challenges associated with CAR-T therapy in solid tumors. Additionally, the changes in tumor nanomechanics observed through AFM suggest that tumor stiffness and plasticity could be biomarkers for predicting treatment outcomes. These results support further investigation into the clinical application of this combined approach to improve the efficacy of cellbased therapies in patients with solid tumors. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
3. Education and Outreach in Physical Sciences in Oncology
- Author
-
Walker, Sierra A., Pham, Anthony, Nizzero, Sara, Kim, Mingee, Riter, Bob, Bletz, Julie, Judge, Sheila, Phillips, Benette, Noble, Dorottya, Murray, Diana, Wetzel, Erin, Samson, Susan, McMahon, Mariah, Flink, Carl, Couch, Jennifer, Tomlin, Claire, Swanson, Kristin, Anderson, Alexander R.A., Odde, David, Shen, Haifa, Hughes, Shannon, Zahir, Nastaran, Enderling, Heiko, and Wolfram, Joy
- Published
- 2021
- Full Text
- View/download PDF
4. A mathematical model for the quantification of a patient’s sensitivity to checkpoint inhibitors and long-term tumour burden
- Author
-
Butner, Joseph D., Wang, Zhihui, Elganainy, Dalia, Al Feghali, Karine A., Plodinec, Marija, Calin, George A., Dogra, Prashant, Nizzero, Sara, Ruiz-Ramírez, Javier, Martin, Geoffrey V., Tawbi, Hussein A., Chung, Caroline, Koay, Eugene J., Welsh, James W., Hong, David S., and Cristini, Vittorio
- Published
- 2021
- Full Text
- View/download PDF
5. Dedifferentiation-mediated stem cell niche maintenance in early-stage ductal carcinoma in situ progression: insights from a multiscale modeling study
- Author
-
Butner, Joseph D., Dogra, Prashant, Chung, Caroline, Ruiz-Ramírez, Javier, Nizzero, Sara, Plodinec, Marija, Li, Xiaoxian, Pan, Ping-Ying, Chen, Shu-hsia, Cristini, Vittorio, Ozpolat, Bulent, Calin, George A., and Wang, Zhihui
- Published
- 2022
- Full Text
- View/download PDF
6. Immunotherapeutic Transport Oncophysics: Space, Time, and Immune Activation in Cancer
- Author
-
Nizzero, Sara, Shen, Haifa, Ferrari, Mauro, and Corradetti, Bruna
- Published
- 2020
- Full Text
- View/download PDF
7. Transport Barriers and Oncophysics in Cancer Treatment
- Author
-
Nizzero, Sara, Ziemys, Arturas, and Ferrari, Mauro
- Published
- 2018
- Full Text
- View/download PDF
8. Bone-targeting nanoparticle to co-deliver decitabine and arsenic trioxide for effective therapy of myelodysplastic syndrome with low systemic toxicity
- Author
-
Wu, Xiaoyan, Hu, Zhenhua, Nizzero, Sara, Zhang, Guodong, Ramirez, Maricela R., Shi, Ce, Zhou, Jin, Ferrari, Mauro, and Shen, Haifa
- Published
- 2017
- Full Text
- View/download PDF
9. Image‐guided mathematical modeling for pharmacological evaluation of nanomaterials and monoclonal antibodies.
- Author
-
Dogra, Prashant, Butner, Joseph D., Nizzero, Sara, Ruiz Ramírez, Javier, Noureddine, Achraf, Peláez, María J., Elganainy, Dalia, Yang, Zhen, Le, Anh‐Dung, Goel, Shreya, Leong, Hon S., Koay, Eugene J., Brinker, C. Jeffrey, Cristini, Vittorio, and Wang, Zhihui
- Abstract
While plasma concentration kinetics has traditionally been the predictor of drug pharmacological effects, it can occasionally fail to represent kinetics at the site of action, particularly for solid tumors. This is especially true in the case of delivery of therapeutic macromolecules (drug‐loaded nanomaterials or monoclonal antibodies), which can experience challenges to effective delivery due to particle size‐dependent diffusion barriers at the target site. As a result, disparity between therapeutic plasma kinetics and kinetics at the site of action may exist, highlighting the importance of target site concentration kinetics in determining the pharmacodynamic effects of macromolecular therapeutic agents. Assessment of concentration kinetics at the target site has been facilitated by non‐invasive in vivo imaging modalities. This allows for visualization and quantification of the whole‐body disposition behavior of therapeutics that is essential for a comprehensive understanding of their pharmacokinetics and pharmacodynamics. Quantitative non‐invasive imaging can also help guide the development and parameterization of mathematical models for descriptive and predictive purposes. Here, we present a review of the application of state‐of‐the‐art imaging modalities for quantitative pharmacological evaluation of therapeutic nanoparticles and monoclonal antibodies, with a focus on their integration with mathematical models, and identify challenges and opportunities. This article is categorized under:Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic DiseaseDiagnostic Tools > in vivo Nanodiagnostics and ImagingNanotechnology Approaches to Biology > Nanoscale Systems in Biology [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
10. Molecular targeting of FATP4 transporter for oral delivery of therapeutic peptide.
- Author
-
Zhenhua Hu, Nizzero, Sara, Goel, Shreya, Hinkle, Louis E., Xiaoyan Wu, Chao Li, Ferrari, Mauro, and Shen, Haifa
- Subjects
- *
FATTY acid-binding proteins , *INTRACELLULAR tracking , *PATIENT compliance - Published
- 2020
- Full Text
- View/download PDF
11. Systematic comparison of methods for determining the in vivo biodistribution of porous nanostructured injectable inorganic particles.
- Author
-
Nizzero, Sara, Li, Feng, Zhang, Guodong, Venuta, Alessandro, Borsoi, Carlotta, Mai, Junhua, Shen, Haifa, Wolfram, Joy, Li, Zheng, Blanco, Elvin, and Ferrari, Mauro
- Subjects
INDUCTIVELY coupled plasma atomic emission spectrometry ,POROUS silicon - Abstract
With a wide variety of biodistribution measurement techniques reported in the literature, it is important to perform side-by-side comparisons of results obtained with different methods on the same particle platform, to determine differences across methods, highlight advantages and disadvantages, and inform methods selection according to specific applications. Inorganic nanostructured particles (INPs) have gained a central role in the development of injectable delivery vectors thanks to their controllable design, biocompatibility, and favorable degradation kinetic. Thus, accurate determination of in vivo biodistribution of INPs is a key aspect of developing and optimizing this class of delivery vectors. In this study, a systematic comparison of spectroscopy (inductively coupled plasma optical emission spectroscopy), fluorescence (in vivo imaging system, confocal microscopy, and plate reader), and radiolabeling (gamma counter)-based techniques is performed to assess the accuracy and sensitivity of biodistribution measurements in mice. Each method is evaluated on porous silicon particles, an established and versatile injectable delivery platform. Biodistribution is evaluated in all major organs and compared in terms of absolute results (%ID/g and %ID/organ when possible) and sensitivity (σ %). Finally, we discuss how these results can be extended to inform method selection for other platforms and specific applications, with an outlook to potential benefit for pre-clinical and clinical studies. Overall, this study presents a new practical guide for selection of in vivo biodistribution methods that yield quantitative results. The significance of this work lies in the use of a single platform to test performances of different biodistribution methods in vivo , with a strict quantitative metric. These results, united with the qualitative comparison of advantages and disadvantages of each technique, are aimed at supporting the rational choice of each different method according to the specific application, to improve the quantitative description of biodistribution results that will be published by others in the future. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
12. A Mathematical Model to Estimate Chemotherapy Concentration at the Tumor-Site and Predict Therapy Response in Colorectal Cancer Patients with Liver Metastases.
- Author
-
Anaya, Daniel A., Dogra, Prashant, Wang, Zhihui, Haider, Mintallah, Ehab, Jasmina, Jeong, Daniel K., Ghayouri, Masoumeh, Lauwers, Gregory Y., Thomas, Kerry, Kim, Richard, Butner, Joseph D., Nizzero, Sara, Ramírez, Javier Ruiz, Plodinec, Marija, Sidman, Richard L., Cavenee, Webster K., Pasqualini, Renata, Arap, Wadih, Fleming, Jason B., and Cristini, Vittorio
- Subjects
ANTINEOPLASTIC agents ,CANCER chemotherapy ,CANCER patients ,COLON tumors ,COMBINED modality therapy ,LIVER tumors ,METASTASIS ,RECTUM tumors ,PREDICTION models ,TREATMENT effectiveness ,STATISTICAL models ,DESCRIPTIVE statistics - Abstract
Simple Summary: It is known that drug transport barriers in the tumor determine drug concentration at the tumor site, causing disparity from the systemic (plasma) drug concentration. However, current clinical standard of care still bases dosage and treatment optimization on the systemic concentration of drugs. Here, we present a proof of concept observational cohort study to accurately estimate drug concentration at the tumor site from mathematical modeling using biologic, clinical, and imaging/perfusion data, and correlate it with outcome in colorectal cancer liver metastases. We demonstrate that drug concentration at the tumor site, not in systemic circulation, can be used as a credible biomarker for predicting chemotherapy outcome, and thus our mathematical modeling approach can be applied prospectively in the clinic to personalize treatment design to optimize outcome. Chemotherapy remains a primary treatment for metastatic cancer, with tumor response being the benchmark outcome marker. However, therapeutic response in cancer is unpredictable due to heterogeneity in drug delivery from systemic circulation to solid tumors. In this proof-of-concept study, we evaluated chemotherapy concentration at the tumor-site and its association with therapy response by applying a mathematical model. By using pre-treatment imaging, clinical and biologic variables, and chemotherapy regimen to inform the model, we estimated tumor-site chemotherapy concentration in patients with colorectal cancer liver metastases, who received treatment prior to surgical hepatic resection with curative-intent. The differential response to therapy in resected specimens, measured with the gold-standard Tumor Regression Grade (TRG; from 1, complete response to 5, no response) was examined, relative to the model predicted systemic and tumor-site chemotherapy concentrations. We found that the average calculated plasma concentration of the cytotoxic drug was essentially equivalent across patients exhibiting different TRGs, while the estimated tumor-site chemotherapeutic concentration (eTSCC) showed a quadratic decline from TRG = 1 to TRG = 5 (p < 0.001). The eTSCC was significantly lower than the observed plasma concentration and dropped by a factor of ~5 between patients with complete response (TRG = 1) and those with no response (TRG = 5), while the plasma concentration remained stable across TRG groups. TRG variations were driven and predicted by differences in tumor perfusion and eTSCC. If confirmed in carefully planned prospective studies, these findings will form the basis of a paradigm shift in the care of patients with potentially curable colorectal cancer and liver metastases. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
13. A pyruvate decarboxylase-mediated therapeutic strategy for mimicking yeast metabolism in cancer cells.
- Author
-
Scott, Bronwyn, Shen, Jianliang, Nizzero, Sara, Boom, Kathryn, Persano, Stefano, Mi, Yu, Liu, Xuewu, Zhao, Yuliang, Blanco, Elvin, Shen, Haifa, Ferrari, Mauro, and Wolfram, Joy
- Subjects
- *
PYRUVATE decarboxylase , *GLYCOLYSIS , *LACTIC acid fermentation , *WARBURG Effect (Oncology) , *BREAST cancer , *CATIONIC lipids - Abstract
Cancer cells have high rates of glycolysis and lactic acid fermentation in order to fuel accelerated rates of cell division (Warburg effect). Here, we present a strategy for merging cancer and yeast metabolism to remove pyruvate, a key intermediate of cancer cell metabolism, and produce the toxic compound acetaldehyde. This approach was achieved by administering the yeast enzyme pyruvate decarboxylase to triple negative breast cancer cells. To overcome the challenges of protein delivery, a nanoparticle-based system consisting of cationic lipids and porous silicon were employed to obtain efficient intracellular uptake. The results demonstrate that the enzyme therapy decreases cancer cell viability through production of acetaldehyde and reduction of lactic acid fermentation. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
14. Sequential deconstruction of composite drug transport in metastatic breast cancer.
- Author
-
Goel, Shreya, Guodong Zhang, Dogra, Prashant, Nizzero, Sara, Cristini, Vittorio, Zhihui Wang, Zhenhua Hu, Zheng Li, Xuewu Liu, Haifa Shen, and Ferrari, Mauro
- Subjects
- *
METASTATIC breast cancer , *PHARMACOKINETICS , *ORGANS (Anatomy) , *FLUORODEOXYGLUCOSE F18 , *APPLIED sciences , *PHYSICAL sciences , *VASCULAR remodeling - Abstract
The article presents a multidisciplinary translational toolbox to evaluate transport and interactions of drug delivery systems (DDS) within metastases. It mentions the challenges to design effective drug delivery systems (DDS) that target metastatic breast cancers (MBC) because of lack of competent imaging and image analysis protocols that suitably capture the interactions between DDS and metastatic lesions.
- Published
- 2020
- Full Text
- View/download PDF
15. Image-guided mathematical modeling for pharmacological evaluation of nanomaterials and monoclonal antibodies.
- Author
-
Dogra P, Butner JD, Nizzero S, Ruiz Ramírez J, Noureddine A, Peláez MJ, Elganainy D, Yang Z, Le AD, Goel S, Leong HS, Koay EJ, Brinker CJ, Cristini V, and Wang Z
- Subjects
- Animals, Cell Line, Tumor, Humans, Mice, Models, Theoretical, Neoplasms diagnostic imaging, Neoplasms drug therapy, Theranostic Nanomedicine, Xenograft Model Antitumor Assays, Antibodies, Monoclonal metabolism, Antibodies, Monoclonal pharmacokinetics, Antibodies, Monoclonal pharmacology, Antibodies, Monoclonal therapeutic use, Image Processing, Computer-Assisted, Nanostructures
- Abstract
While plasma concentration kinetics has traditionally been the predictor of drug pharmacological effects, it can occasionally fail to represent kinetics at the site of action, particularly for solid tumors. This is especially true in the case of delivery of therapeutic macromolecules (drug-loaded nanomaterials or monoclonal antibodies), which can experience challenges to effective delivery due to particle size-dependent diffusion barriers at the target site. As a result, disparity between therapeutic plasma kinetics and kinetics at the site of action may exist, highlighting the importance of target site concentration kinetics in determining the pharmacodynamic effects of macromolecular therapeutic agents. Assessment of concentration kinetics at the target site has been facilitated by non-invasive in vivo imaging modalities. This allows for visualization and quantification of the whole-body disposition behavior of therapeutics that is essential for a comprehensive understanding of their pharmacokinetics and pharmacodynamics. Quantitative non-invasive imaging can also help guide the development and parameterization of mathematical models for descriptive and predictive purposes. Here, we present a review of the application of state-of-the-art imaging modalities for quantitative pharmacological evaluation of therapeutic nanoparticles and monoclonal antibodies, with a focus on their integration with mathematical models, and identify challenges and opportunities. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease Diagnostic Tools > in vivo Nanodiagnostics and Imaging Nanotechnology Approaches to Biology > Nanoscale Systems in Biology., (© 2020 The Authors. WIREs Nanomedicine and Nanobiotechnology published by Wiley Periodicals, Inc.)
- Published
- 2020
- Full Text
- View/download PDF
16. Retraction of the Research Article: "Molecular targeting of FATP4 transporter for oral delivery of therapeutic peptide" by Z. Hu, S. Nizzero, S. Goel, L. E. Hinkle, X. Wu, C. Li, M. Ferrari and H. Shen.
- Author
-
Nizzero S, Goel S, Hinkle LE, Wu X, Li C, Ferrari M, and Shen H
- Abstract
[This retracts the article DOI: 10.1126/sciadv.aba0145.]., (Copyright © 2020, American Association for the Advancement of Science.)
- Published
- 2020
- Full Text
- View/download PDF
17. Molecular targeting of FATP4 transporter for oral delivery of therapeutic peptide.
- Author
-
Hu Z, Nizzero S, Goel S, Hinkle LE, Wu X, Li C, Ferrari M, and Shen H
- Abstract
Low oral bioavailability of peptide drugs has limited their application to parenteral administration, which suffers from poor patient compliance. Here, we show that molecular targeting of the FATP4 transporter is an effective approach to specifically transport long-chain fatty acid (LCFA)-conjugated peptides across the enterocytic membrane and, thus, enables oral delivery of drug peptides. We packaged LCFA-conjugated exendin-4 (LCFA-Ex4) into liposomes and coated with chitosan nanoparticles to form an orally deliverable Ex4 (OraEx4). OraEx4 protected LCFA-Ex4 from damage by the gastric fluid and released LCFA-Ex4 in the intestinal cavity, where LCFA-Ex4 was transported across the enterocyte membrane by the FAPT4 transporter. OraEx4 had a high bioavailability of 24.8% with respect to subcutaneous injection and exhibited a substantial hypoglycemic effect in murine models of diabetes mellitus. Thus, molecular targeting of the FATP4 transporter enhances oral absorption of therapeutic peptides and provides a platform for oral peptide drug development., (Copyright © 2020 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works. Distributed under a Creative Commons Attribution NonCommercial License 4.0 (CC BY-NC).)
- Published
- 2020
- Full Text
- View/download PDF
18. Smeared Multiscale Finite Element Models for Mass Transport and Electrophysiology Coupled to Muscle Mechanics.
- Author
-
Kojic M, Milosevic M, Simic V, Milicevic B, Geroski V, Nizzero S, Ziemys A, Filipovic N, and Ferrari M
- Abstract
Mass transport represents the most fundamental process in living organisms. It includes delivery of nutrients, oxygen, drugs, and other substances from the vascular system to tissue and transport of waste and other products from cells back to vascular and lymphatic network and organs. Furthermore, movement is achieved by mechanical forces generated by muscles in coordination with the nervous system. The signals coming from the brain, which have the character of electrical waves, produce activation within muscle cells. Therefore, from a physics perspective, there exist a number of physical fields within the body, such as velocities of transport, pressures, concentrations of substances, and electrical potential, which is directly coupled to biochemical processes of transforming the chemical into mechanical energy and further internal forces for motion. The overall problems of mass transport and electrophysiology coupled to mechanics can be investigated theoretically by developing appropriate computational models. Due to the enormous complexity of the biological system, it would be almost impossible to establish a detailed computational model for the physical fields related to mass transport, electrophysiology, and coupled fields. To make computational models feasible for applications, we here summarize a concept of smeared physical fields, with coupling among them, and muscle mechanics, which includes dependence on the electrical potential. Accuracy of the smeared computational models, also with coupling to muscle mechanics, is illustrated with simple example, while their applicability is demonstrated on a liver model with tumors present. The last example shows that the introduced methodology is applicable to large biological systems., (Copyright © 2019 Kojic, Milosevic, Simic, Milicevic, Geroski, Nizzero, Ziemys, Filipovic and Ferrari.)
- Published
- 2019
- Full Text
- View/download PDF
19. A chloroquine-induced macrophage-preconditioning strategy for improved nanodelivery.
- Author
-
Wolfram J, Nizzero S, Liu H, Li F, Zhang G, Li Z, Shen H, Blanco E, and Ferrari M
- Subjects
- Animals, Biological Transport drug effects, Cell Line, Cell Survival drug effects, Endocytosis drug effects, Humans, Immunity, Innate drug effects, Kupffer Cells cytology, Kupffer Cells drug effects, Kupffer Cells immunology, Kupffer Cells metabolism, Macrophages cytology, Macrophages immunology, Mice, Chloroquine pharmacology, Drug Carriers metabolism, Macrophages drug effects, Macrophages metabolism, Nanoparticles metabolism
- Abstract
Site-specific localization is critical for improving the therapeutic efficacy and safety of drugs. Nanoparticles have emerged as promising tools for localized drug delivery. However, over 90% of systemically injected nanocarriers typically accumulate in the liver and spleen due to resident macrophages that form the mononuclear phagocyte system. In this study, the clinically approved antimalarial agent chloroquine was shown to reduce nanoparticle uptake in macrophages by suppressing endocytosis. Pretreatment of mice with a clinically relevant dose of chloroquine substantially decreased the accumulation of liposomes and silicon particles in the mononuclear phagocyte system and improved tumoritropic and organotropic delivery. The novel use of chloroquine as a macrophage-preconditioning agent presents a straightforward approach for addressing a major barrier in nanomedicine. Moreover, this priming strategy has broad applicability for improving the biodistribution and performance of particulate delivery systems. Ultimately, this study defines a paradigm for the combined use of macrophage-modulating agents with nanotherapeutics for improved site-specific delivery.
- Published
- 2017
- Full Text
- View/download PDF
20. Single-particle absorption spectroscopy by photothermal contrast.
- Author
-
Yorulmaz M, Nizzero S, Hoggard A, Wang LY, Cai YY, Su MN, Chang WS, and Link S
- Abstract
Removing effects of sample heterogeneity through single-molecule and single-particle techniques has advanced many fields. While background free luminescence and scattering spectroscopy is widely used, recording the absorption spectrum only is rather difficult. Here we present an approach capable of recording pure absorption spectra of individual nanostructures. We demonstrate the implementation of single-particle absorption spectroscopy on strongly scattering plasmonic nanoparticles by combining photothermal microscopy with a supercontinuum laser and an innovative calibration procedure that accounts for chromatic aberrations and wavelength-dependent excitation powers. Comparison of the absorption spectra to the scattering spectra of the same individual gold nanoparticles reveals the blueshift of the absorption spectra, as predicted by Mie theory but previously not detectable in extinction measurements that measure the sum of absorption and scattering. By covering a wavelength range of 300 nm, we are furthermore able to record absorption spectra of single gold nanorods with different aspect ratios. We find that the spectral shift between absorption and scattering for the longitudinal plasmon resonance decreases as a function of nanorod aspect ratio, which is in agreement with simulations.
- Published
- 2015
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.