1
|
Macone A, Cappelletti C, Incocciati A, Piacentini R, Botta S, Boffi A, Bonamore A. Challenges in Exploiting Human H Ferritin Nanoparticles for Drug Delivery: Navigating Physiological Constraints. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2024; 16:e2016. [PMID: 39541599 DOI: 10.1002/wnan.2016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 10/14/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024]
Abstract
Over the past two decades, ferritin has emerged as a promising nanoparticle for drug delivery, catalyzing the development of numerous prototypes capable of encapsulating a wide array of therapeutic agents. These ferritin-based nanoparticles exhibit selectivity for various molecular targets and are distinguished by their potential biocompatibility, unique symmetrical structure, and highly controlled size. The hollow interior of ferritin nanoparticles allows for efficient encapsulation of diverse therapeutic agents, enhancing their delivery and effectiveness. Despite these promising features, the anticipated clinical advancements have yet to be fully realized. As a physiological protein with a central role in both health and disease, ferritin can exert unexpected effects on physiology when employed as a drug delivery system. Many studies have not thoroughly evaluated the pharmacokinetic properties of the ferritin protein shell when administered in vivo, overlooking crucial aspects such as biodistribution, clearance, cellular trafficking, and immune response. Addressing these challenges is crucial for achieving the desired transition from bench to bedside. Biodistribution studies need to account for ferritin's natural accumulation in specific organs (liver, spleen, and kidneys), which may lead to off-target effects. Moreover, the mechanisms of clearance and cellular trafficking must be elucidated to optimize the delivery and reduce potential toxicity of ferritin nanoparticles. Additionally, understanding the immune response elicited by exogenous ferritin is essential to mitigate adverse reactions and enhance therapeutic efficacy. A comprehensive understanding of these physiological constraints, along with innovative solutions, is essential to fully realize the therapeutic potential of ferritin nanoparticles paving the way for their successful clinical translation.
Collapse
Affiliation(s)
- Alberto Macone
- Department of Biochemical Sciences "Alessandro Rossi Fanelli", Sapienza University of Rome, Rome, Italy
| | - Chiara Cappelletti
- Department of Biochemical Sciences "Alessandro Rossi Fanelli", Sapienza University of Rome, Rome, Italy
| | - Alessio Incocciati
- Department of Biochemical Sciences "Alessandro Rossi Fanelli", Sapienza University of Rome, Rome, Italy
| | - Roberta Piacentini
- Department of Biochemical Sciences "Alessandro Rossi Fanelli", Sapienza University of Rome, Rome, Italy
| | - Sofia Botta
- Department of Biochemical Sciences "Alessandro Rossi Fanelli", Sapienza University of Rome, Rome, Italy
| | - Alberto Boffi
- Department of Biochemical Sciences "Alessandro Rossi Fanelli", Sapienza University of Rome, Rome, Italy
| | - Alessandra Bonamore
- Department of Biochemical Sciences "Alessandro Rossi Fanelli", Sapienza University of Rome, Rome, Italy
| |
Collapse
|
2
|
Cotto N, Chauhan N, Adriano B, Chauhan DS, Cabrera M, Chauhan SC, Yallapu MM. Milk Exosome-Glow Nanosystem for Cancer Cellular and Tissue Bioimaging. CHEMICAL & BIOMEDICAL IMAGING 2024; 2:711-720. [PMID: 39483633 PMCID: PMC11522989 DOI: 10.1021/cbmi.4c00040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 07/25/2024] [Accepted: 07/26/2024] [Indexed: 11/03/2024]
Abstract
Milk-derived exosomes are widely used for diagnosis, delivery, imaging, and theranostic applications. Near-Infrared (NIR) based fluorescence bioimaging is an attractive and safer technique that is used for clinical applications. However, almost all NIR imaging agents tend to have poor photostability, short half-life, nonspecific protein binding, and concentration-dependent aggregation(s). Therefore, there is an unmet clinical need to develop newer and safer modalities to package and deliver NIR imaging agents. Bovine milk exosomes are natural, biocompatible, safe, and efficient nanocarriers that facilitate the delivery of micro- and macromolecules. Herein, we developed an exosome-based NIR dye loaded nanoimaging formulation that offers improved solubility and photostability of NIR dye. Following the acetic acid based extracellular vesicle (EV) treatment method, we extracted the bovine milk exosomes from a variety of pasteurized grade milk. The EVs were screened for their physicochemical properties such as particle size and concentration and zeta potential. The stability of these exosomes was also determined under different conditions, including storage temperatures, pH, and salt concentrations. Next, indocyanine green, a model NIR dye was loaded into these exosomes (Exo-Glow) via a sonication method and further assessed for their improved fluorescence intensity and photostability using an IVIS imaging system. Initial screening suggested that size of the selected bovine milk exosomes was ∼100-135 nm with an average particle concentration of 5.8 × 102 particles/mL. Exo-Glow further demonstrated higher fluorescence intensity in cancer cells and tissues when compared to free dye. These results showed that Exo-Glow has the potential to serve as a safer NIR imaging tool for cancer cells/tissues.
Collapse
Affiliation(s)
- Nycol
M. Cotto
- Division
of Immunology and Microbiology, Medicine and Oncology Integrated Service
Unit, School of Medicine, The University
of Texas Rio Grande Valley, McAllen, Texas 78504, United States
- South
Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, Texas 78504, United States
| | - Neeraj Chauhan
- Division
of Immunology and Microbiology, Medicine and Oncology Integrated Service
Unit, School of Medicine, The University
of Texas Rio Grande Valley, McAllen, Texas 78504, United States
- South
Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, Texas 78504, United States
| | - Benilde Adriano
- Division
of Immunology and Microbiology, Medicine and Oncology Integrated Service
Unit, School of Medicine, The University
of Texas Rio Grande Valley, McAllen, Texas 78504, United States
- South
Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, Texas 78504, United States
| | - Deepak S. Chauhan
- Division
of Immunology and Microbiology, Medicine and Oncology Integrated Service
Unit, School of Medicine, The University
of Texas Rio Grande Valley, McAllen, Texas 78504, United States
- South
Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, Texas 78504, United States
| | - Marco Cabrera
- Division
of Immunology and Microbiology, Medicine and Oncology Integrated Service
Unit, School of Medicine, The University
of Texas Rio Grande Valley, McAllen, Texas 78504, United States
- South
Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, Texas 78504, United States
| | - Subhash C. Chauhan
- Division
of Immunology and Microbiology, Medicine and Oncology Integrated Service
Unit, School of Medicine, The University
of Texas Rio Grande Valley, McAllen, Texas 78504, United States
- South
Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, Texas 78504, United States
| | - Murali M. Yallapu
- Division
of Immunology and Microbiology, Medicine and Oncology Integrated Service
Unit, School of Medicine, The University
of Texas Rio Grande Valley, McAllen, Texas 78504, United States
- South
Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, Texas 78504, United States
| |
Collapse
|
3
|
Chen Y, Xu C, Sun M, Zhao G, Wang Z, Lv C. Vertasile ferritin nanocages: Applications in detection and bioimaging. Biosens Bioelectron 2024; 262:116567. [PMID: 39013360 DOI: 10.1016/j.bios.2024.116567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Revised: 06/30/2024] [Accepted: 07/10/2024] [Indexed: 07/18/2024]
Abstract
Food safety and human health remain significant concerns in the food industry. Detecting food contaminants and diagnosing diseases are critical aspects. Ferritin, an iron storage protein widely found in nature, offers unique advantages. Its hollow protein nanocage structure, distinct interfaces, hydrophobic or hydrophilic channels, and B-C loop regions recognized by transferrin receptor 1 make ferritin versatile for detecting heavy metals, free radicals, and bioimaging both in vitro and in vivo. This review summarizes ferritin's general characteristics, its specific properties as biosensors, and its applications in food safety and in vivo imaging. It emphasizes not only ferritin's role in detecting heavy metals like mercury and chemical hazards but also its potential in early diagnosing chronic diseases such as tumors, macrophages, and kidney diseases. Further research into ferritin promises advancements in enhancing food safety and improving human health diagnostics.
Collapse
Affiliation(s)
- Yunqi Chen
- College of Food Science & Nutritional Engineering, China Agricultural University, Beijing Key Laboratory of Functional Food from Plant Resources, Beijing, PR China
| | - Chen Xu
- College of Food Science & Nutritional Engineering, China Agricultural University, Beijing Key Laboratory of Functional Food from Plant Resources, Beijing, PR China
| | - Mingyang Sun
- College of Food Science & Nutritional Engineering, China Agricultural University, Beijing Key Laboratory of Functional Food from Plant Resources, Beijing, PR China
| | - Guanghua Zhao
- College of Food Science & Nutritional Engineering, China Agricultural University, Beijing Key Laboratory of Functional Food from Plant Resources, Beijing, PR China
| | - Zhongjiang Wang
- College of Food Science, Northeast Agricultural University, Haerbin, Heilongjiang Province, PR China.
| | - Chenyan Lv
- College of Food Science & Nutritional Engineering, China Agricultural University, Beijing Key Laboratory of Functional Food from Plant Resources, Beijing, PR China.
| |
Collapse
|
4
|
Jeong S, Lee H, Jeong Y, Cha HJ. Indocyanine Green-Loaded Adhesive Proteinic Nanoparticles for Effective Locoregional and Prolonged Photothermal Anticancer Therapy. Biomacromolecules 2024; 25:6913-6921. [PMID: 39297577 DOI: 10.1021/acs.biomac.4c01143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2024]
Abstract
Conventional anticancer therapies, including surgical resection, radiation, and chemotherapy, are the primary modalities for treating various forms of cancer. However, these treatments often bring significant side effects and risk of recurrence, underscoring the need for more targeted and less invasive therapeutic options. To address this challenge, we developed an adhesive nanoparticle (NP)-based effective anticancer photothermal therapy (PTT) system using bioengineered mussel adhesion protein (MAP). The unique underwater tissue adhesive properties of MAP NPs enabled targeted delivery and prolonged retention at the tumor site, thereby improving therapeutic efficacy. Our innovative indocyanine green (ICG)-loaded MAP NPs (MAP@ICG NPs) demonstrated strong photothermal capability and stability, and potent anticancer activity in vitro. In vivo intratumor injection of the MAP@ICG NPs showed remarkable anticancer PTT effects, effectively reducing tumor growth with minimal damage to surrounding tissues. The development and utilization of this adhesive proteinic NP-based PTT system represent a significant advancement in cancer therapy, offering a promising alternative that combines the precision of NP delivery with effective therapeutic efficacy.
Collapse
Affiliation(s)
- Soyeon Jeong
- Department of Chemical Engineering, Pohang University of Science and Technology, Pohang 37673, Republic of Korea
| | - Hyeokjun Lee
- Department of Chemical Engineering, Pohang University of Science and Technology, Pohang 37673, Republic of Korea
| | - Yeonsu Jeong
- Department of Chemical Engineering, Pohang University of Science and Technology, Pohang 37673, Republic of Korea
| | - Hyung Joon Cha
- Department of Chemical Engineering, Pohang University of Science and Technology, Pohang 37673, Republic of Korea
- Medical Science and Engineering, School of Convergence Science and Technology, Pohang University of Science and Technology, Pohang 37673, Republic of Korea
| |
Collapse
|
5
|
Incocciati A, Cappelletti C, Masciarelli S, Liccardo F, Piacentini R, Giorgi A, Bertuccini L, De Berardis B, Fazi F, Boffi A, Bonamore A, Macone A. Ferritin-based disruptor nanoparticles: A novel strategy to enhance LDL cholesterol clearance via multivalent inhibition of PCSK9-LDL receptor interaction. Protein Sci 2024; 33:e5111. [PMID: 39150051 PMCID: PMC11328107 DOI: 10.1002/pro.5111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 06/24/2024] [Accepted: 06/26/2024] [Indexed: 08/17/2024]
Abstract
Hypercholesterolemia, characterized by elevated low-density lipoprotein (LDL) cholesterol levels, is a significant risk factor for cardiovascular disease. Proprotein convertase subtilisin/kexin type 9 (PCSK9) plays a crucial role in cholesterol metabolism by regulating LDL receptor degradation, making it a therapeutic target for mitigating hypercholesterolemia-associated risks. In this context, we aimed to engineer human H ferritin as a scaffold to present 24 copies of a PCSK9-targeting domain. The rationale behind this protein nanoparticle design was to disrupt the PCSK9-LDL receptor interaction, thereby attenuating the PCSK9-mediated impairment of LDL cholesterol clearance. The N-terminal sequence of human H ferritin was engineered to incorporate a 13-amino acid linear peptide (Pep2-8), which was previously identified as the smallest PCSK9 inhibitor. Exploiting the quaternary structure of ferritin, engineered nanoparticles were designed to display 24 copies of the targeting peptide on their surface, enabling a multivalent binding effect. Extensive biochemical characterization confirmed precise control over nanoparticle size and morphology, alongside robust PCSK9-binding affinity (KD in the high picomolar range). Subsequent efficacy assessments employing the HepG2 liver cell line demonstrated the ability of engineered ferritin's ability to disrupt PCSK9-LDL receptor interaction, thereby promoting LDL receptor recycling on cell surfaces and consequently enhancing LDL uptake. Our findings highlight the potential of ferritin-based platforms as versatile tools for targeting PCSK9 in the management of hypercholesterolemia. This study not only contributes to the advancement of ferritin-based therapeutics but also offers valuable insights into novel strategies for treating cardiovascular diseases.
Collapse
Affiliation(s)
- Alessio Incocciati
- Department of Biochemical Sciences "Alessandro Rossi Fanelli", Sapienza University of Rome, Rome, Italy
| | - Chiara Cappelletti
- Department of Biochemical Sciences "Alessandro Rossi Fanelli", Sapienza University of Rome, Rome, Italy
| | - Silvia Masciarelli
- Department of Anatomical, Histological, Forensic & Orthopaedic Sciences, Section of Histology and Medical Embryology, Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University of Rome, Rome, Italy
| | - Francesca Liccardo
- Department of Anatomical, Histological, Forensic & Orthopaedic Sciences, Section of Histology and Medical Embryology, Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University of Rome, Rome, Italy
| | - Roberta Piacentini
- Department of Biochemical Sciences "Alessandro Rossi Fanelli", Sapienza University of Rome, Rome, Italy
- Center for Life Nano Science at Sapienza, Istituto Italiano di Tecnologia, Rome, Italy
| | - Alessandra Giorgi
- Department of Biochemical Sciences "Alessandro Rossi Fanelli", Sapienza University of Rome, Rome, Italy
| | - Lucia Bertuccini
- Core Facilities, Microscopy Area, Istituto Superiore di Sanita, Rome, Italy
| | - Barbara De Berardis
- National Center for Innovative Technologies in Public Health, Istituto Superiore di Sanità, Rome, Italy
| | - Francesco Fazi
- Department of Anatomical, Histological, Forensic & Orthopaedic Sciences, Section of Histology and Medical Embryology, Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University of Rome, Rome, Italy
| | - Alberto Boffi
- Department of Biochemical Sciences "Alessandro Rossi Fanelli", Sapienza University of Rome, Rome, Italy
| | - Alessandra Bonamore
- Department of Biochemical Sciences "Alessandro Rossi Fanelli", Sapienza University of Rome, Rome, Italy
| | - Alberto Macone
- Department of Biochemical Sciences "Alessandro Rossi Fanelli", Sapienza University of Rome, Rome, Italy
| |
Collapse
|
6
|
Sitia L, Saccomandi P, Bianchi L, Sevieri M, Sottani C, Allevi R, Grignani E, Mazzucchelli S, Corsi F. Combined Ferritin Nanocarriers with ICG for Effective Phototherapy Against Breast Cancer. Int J Nanomedicine 2024; 19:4263-4278. [PMID: 38766663 PMCID: PMC11102096 DOI: 10.2147/ijn.s445334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 03/30/2024] [Indexed: 05/22/2024] Open
Abstract
Introduction Photodynamic Therapy (PDT) is a promising, minimally invasive treatment for cancer with high immunostimulatory potential, no reported drug resistance, and reduced side effects. Indocyanine Green (ICG) has been used as a photosensitizer (PS) for PDT, although its poor stability and low tumor-target specificity strongly limit its efficacy. To overcome these limitations, ICG can be formulated as a tumor-targeting nanoparticle (NP). Methods We nanoformulated ICG into recombinant heavy-ferritin nanocages (HFn-ICG). HFn has a specific interaction with transferrin receptor 1 (TfR1), which is overexpressed in most tumors, thus increasing HFn tumor tropism. First, we tested the properties of HFn-ICG as a PS upon irradiation with a continuous-wave diode laser. Then, we evaluated PDT efficacy in two breast cancer (BC) cell lines with different TfR1 expression levels. Finally, we measured the levels of intracellular endogenous heavy ferritin (H-Fn) after PDT treatment. In fact, it is known that cells undergoing ROS-induced autophagy, as in PDT, tend to increase their ferritin levels as a defence mechanism. By measuring intracellular H-Fn, we verified whether this interplay between internalized HFn and endogenous H-Fn could be used to maximize HFn uptake and PDT efficacy. Results We previously demonstrated that HFn-ICG stabilized ICG molecules and increased their delivery to the target site in vitro and in vivo for fluorescence guided surgery. Here, with the aim of using HFn-ICG for PDT, we showed that HFn-ICG improved treatment efficacy in BC cells, depending on their TfR1 expression. Our data revealed that endogenous H-Fn levels were increased after PDT treatment, suggesting that this defence reaction against oxidative stress could be used to enhance HFn-ICG uptake in cells, increasing treatment efficacy. Conclusion The strong PDT efficacy and peculiar Trojan horse-like mechanism, that we revealed for the first time in literature, confirmed the promising application of HFn-ICG in PDT.
Collapse
Affiliation(s)
- Leopoldo Sitia
- Department of Biomedical and Clinical Sciences, Università degli studi di Milano, Milan, Italy
| | - Paola Saccomandi
- Department of Mechanical Engineering, Politecnico di Milano, Milan, Italy
| | - Leonardo Bianchi
- Department of Mechanical Engineering, Politecnico di Milano, Milan, Italy
| | - Marta Sevieri
- Department of Biomedical and Clinical Sciences, Università degli studi di Milano, Milan, Italy
| | - Cristina Sottani
- Environmental Research Center, Istituti Clinici Scientifici Maugeri IRCCS, Pavia, Italy
| | - Raffaele Allevi
- Department of Biomedical and Clinical Sciences, Università degli studi di Milano, Milan, Italy
| | - Elena Grignani
- Environmental Research Center, Istituti Clinici Scientifici Maugeri IRCCS, Pavia, Italy
| | - Serena Mazzucchelli
- Department of Biomedical and Clinical Sciences, Università degli studi di Milano, Milan, Italy
| | - Fabio Corsi
- Department of Biomedical and Clinical Sciences, Università degli studi di Milano, Milan, Italy
- Istituti Clinici Scientifici Maugeri IRCCS, Pavia, Italy
| |
Collapse
|
7
|
Pop CF, Veys I, Bormans A, Larsimont D, Liberale G. Fluorescence imaging for real-time detection of breast cancer tumors using IV injection of indocyanine green with non-conventional imaging: a systematic review of preclinical and clinical studies of perioperative imaging technologies. Breast Cancer Res Treat 2024; 204:429-442. [PMID: 38182824 PMCID: PMC10959791 DOI: 10.1007/s10549-023-07199-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 11/22/2023] [Indexed: 01/07/2024]
Abstract
BACKGROUND This review summarizes the available data on the effectiveness of indocyanine green fluorescence imaging (ICG-FI) for real-time detection of breast cancer (BC) tumors with perioperative imaging technologies. METHODS PubMed and Scopus databases were exhaustively searched for publications on the use of the real-time ICG-FI evaluation of BC tumors with non-conventional breast imaging technologies. RESULTS Twenty-three studies were included in this review. ICG-FI has been used for BC tumor identification in 12 orthotopic animal tumor experiences, 4 studies on animal assessment, and for 7 human clinical applications. The BC tumor-to-background ratio (TBR) was 1.1-8.5 in orthotopic tumor models and 1.4-3.9 in animal experiences. The detection of primary human BC tumors varied from 40% to 100%. The mean TBR reported for human BC varied from 2.1 to 3.7. In two studies evaluating BC surgical margins, good sensitivity (93.3% and 100%) and specificity (60% and 96%) have been reported, with a negative predictive value of ICG-FI to predict margin involvement intraoperatively of 100% in one study. CONCLUSIONS The use of ICG-FI as a guiding tool for the real-time identification of BC tumors and for the assessment of tumor boundaries is promising. There is great variability between the studies with regard to timing and dose. Further evidence is needed to assess whether ICG-guided BC surgery may be implemented as a standard of care.
Collapse
Affiliation(s)
- C Florin Pop
- Department of Surgical Oncology, Institut Jules Bordet, Université Libre de Bruxelles, Rue Meylemeersch 90, 1070, Brussels, Belgium.
| | - Isabelle Veys
- Department of Surgical Oncology, Institut Jules Bordet, Université Libre de Bruxelles, Rue Meylemeersch 90, 1070, Brussels, Belgium
| | - Anne Bormans
- Institutional Library, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Denis Larsimont
- Department of Pathology, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Gabriel Liberale
- Department of Surgical Oncology, Institut Jules Bordet, Université Libre de Bruxelles, Rue Meylemeersch 90, 1070, Brussels, Belgium
| |
Collapse
|
8
|
Sevieri M, Sottani C, Chesi A, Bonizzi A, Sitia L, Robustelli Della Cuna FS, Grignani E, Corsi F, Mazzucchelli S. Deciphering the Role of H-Ferritin Nanocages in Improving Tumor-Targeted Delivery of Indocyanine Green: Combined Analysis of Murine Tissue Homogenates with UHPLC-MS/MS and Fluorescence. ACS OMEGA 2023; 8:48735-48741. [PMID: 38162787 PMCID: PMC10753538 DOI: 10.1021/acsomega.3c05566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 10/19/2023] [Accepted: 11/22/2023] [Indexed: 01/03/2024]
Abstract
We investigated the relevance of encapsulation in H-ferritin nanocages (HFn) in determining an improved tumor-targeted delivery of indocyanine green (ICG). Since from previous experiments, the administration of HFn loaded with ICG (HFn-ICG) resulted in an increased fluorescence signal of ICG, our aim was to uncover if the nanoformulation could have a major role in driving a specific targeting of the dye to the tumor or rather a protective action on ICG's fluorescence. Here, we took advantage of a combined analysis involving ultrahigh performance liquid chromatography-tandem mass spectrometry (UHPLC-MS/MS) on murine tissue homogenates matched with fluorescence intensities analysis detected by ex vivo optical imaging. The quantification of ICG content performed on different organs over time combined with the fluorescent signal detection confirmed the superior delivery of ICG thanks to the nanoformulation. Our results showed that HFn-ICG drives a real accumulation at the tumor instead of only having a role in the preservation of ICG's fluorescence, further supporting its use as a delivery system of ICG for fluorescence-guided surgery applications in oncology.
Collapse
Affiliation(s)
- Marta Sevieri
- Nanomedicine
Laboratory, Dipartimento di Scienze Biomediche e Cliniche, Università di Milano, Milan 20157, Italy
| | - Cristina Sottani
- Environmental
Research Center, Istituti Clinici Scientifici
Maugeri IRCCS, Pavia 27100, Italy
| | - Arianna Chesi
- Nanomedicine
Laboratory, Dipartimento di Scienze Biomediche e Cliniche, Università di Milano, Milan 20157, Italy
| | - Arianna Bonizzi
- Nanomedicine
Laboratory, Dipartimento di Scienze Biomediche e Cliniche, Università di Milano, Milan 20157, Italy
- Breast
Unit, Istituti Clinici Scientifici Maugeri
IRCCS, Pavia 27100, Italy
| | - Leopoldo Sitia
- Nanomedicine
Laboratory, Dipartimento di Scienze Biomediche e Cliniche, Università di Milano, Milan 20157, Italy
| | | | - Elena Grignani
- Environmental
Research Center, Istituti Clinici Scientifici
Maugeri IRCCS, Pavia 27100, Italy
| | - Fabio Corsi
- Nanomedicine
Laboratory, Dipartimento di Scienze Biomediche e Cliniche, Università di Milano, Milan 20157, Italy
- Breast
Unit, Istituti Clinici Scientifici Maugeri
IRCCS, Pavia 27100, Italy
| | - Serena Mazzucchelli
- Nanomedicine
Laboratory, Dipartimento di Scienze Biomediche e Cliniche, Università di Milano, Milan 20157, Italy
| |
Collapse
|
9
|
Rahate NP, Kapse A, Rahate PV, Nimbhorkar SP. The Wonder Dye: Uses and Implications of Indigocyanine Green in Various Surgeries. Cureus 2023; 15:e46722. [PMID: 38021982 PMCID: PMC10630983 DOI: 10.7759/cureus.46722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 10/09/2023] [Indexed: 12/01/2023] Open
Abstract
Indigocyanine green (ICG) is a fluorophore dye that has been extensively used in recent modern times for bioimaging in numerous surgeries to aid in easier identification of occult and often tricky-to-find anatomical structures. Surgery becomes complex and challenging due to multiple anatomical anomalies, pathological fibrosis, obesity, or previous surgeries. To overcome these obstacles in surgery, the surgeon yearns to know the structures present beyond their white light vision so that while dissecting the organ, they can avoid injuring the critical systems in the vicinity of dissection. Near-infrared (NIR) imaging aids in visualising the tissues at depth/in the area of dissection, thereby preventing any possible surgical catastrophes due to them inadvertently damaging surrounding vital structures. Various advantages in surgeries like gastric sleeve surgery, lymph node and tumour detection, localisation of ureters and biliary tracts, and intraoperative tissue perfusion of flaps have been described in this study. This review article aims to compile a short list of utilities of ICG with NIR imaging in various surgical interventions. The merits and demerits of this imaging technique have been noted. The study points out the uses of ICG fluorescence imaging under different surgical fronts. This review article concludes by comparing the results of studies performed by various authors. Results have been compared to conventional surgical modalities.
Collapse
Affiliation(s)
- Nachiket P Rahate
- Surgery, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Ankita Kapse
- Medicine, Datta Meghe Medical College, Datta Meghe Institute of Higher Education and Research, Nagpur, IND
| | | | - Sakshi P Nimbhorkar
- Medicine, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| |
Collapse
|
10
|
Fadel MG, Smith MJ. ASO Author Reflections: Comparison of Indocyanine Green with Blue Dye in Sentinel Lymph Node Biopsy for Cutaneous Melanoma. Ann Surg Oncol 2023; 30:4341-4342. [PMID: 37014554 PMCID: PMC10250504 DOI: 10.1245/s10434-023-13423-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 03/15/2023] [Indexed: 04/05/2023]
Affiliation(s)
- Michael G Fadel
- The Sarcoma, Melanoma and Rare Tumours Unit, The Royal Marsden Hospital and Institute Cancer of Research, London, United Kingdom.
| | - Myles J Smith
- The Sarcoma, Melanoma and Rare Tumours Unit, The Royal Marsden Hospital and Institute Cancer of Research, London, United Kingdom
| |
Collapse
|
11
|
Shesh BP, Connor JR. A novel view of ferritin in cancer. Biochim Biophys Acta Rev Cancer 2023; 1878:188917. [PMID: 37209958 PMCID: PMC10330744 DOI: 10.1016/j.bbcan.2023.188917] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 05/13/2023] [Accepted: 05/13/2023] [Indexed: 05/22/2023]
Abstract
Since its discovery more than 85 years ago, ferritin has principally been known as an iron storage protein. However, new roles, beyond iron storage, are being uncovered. Novel processes involving ferritin such as ferritinophagy and ferroptosis and as a cellular iron delivery protein not only expand our thinking on the range of contributions of this protein but present an opportunity to target these pathways in cancers. The key question we focus on within this review is whether ferritin modulation represents a useful approach for treating cancers. We discussed novel functions and processes of this protein in cancers. We are not limiting this review to cell intrinsic modulation of ferritin in cancers, but also focus on its utility in the trojan horse approach in cancer therapeutics. The novel functions of ferritin as discussed herein realize the multiple roles of ferritin in cell biology that can be probed for therapeutic opportunities and further research.
Collapse
Affiliation(s)
| | - James R Connor
- Department of Neurosurgery, Penn State Hershey Medical Center, Hershey, PA, USA.
| |
Collapse
|
12
|
Proteins and their functionalization for finding therapeutic avenues in cancer: Current status and future prospective. Biochim Biophys Acta Rev Cancer 2023; 1878:188862. [PMID: 36791920 DOI: 10.1016/j.bbcan.2023.188862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 01/13/2023] [Accepted: 01/13/2023] [Indexed: 02/15/2023]
Abstract
Despite the remarkable advancement in the health care sector, cancer remains the second most fatal disease globally. The existing conventional cancer treatments primarily include chemotherapy, which has been associated with little to severe side effects, and radiotherapy, which is usually expensive. To overcome these problems, target-specific nanocarriers have been explored for delivering chemo drugs. However, recent reports on using a few proteins having anticancer activity and further use of them as drug carriers have generated tremendous attention for furthering the research towards cancer therapy. Biomolecules, especially proteins, have emerged as suitable alternatives in cancer treatment due to multiple favourable properties including biocompatibility, biodegradability, and structural flexibility for easy surface functionalization. Several in vitro and in vivo studies have reported that various proteins derived from animal, plant, and bacterial species, demonstrated strong cytotoxic and antiproliferative properties against malignant cells in native and their different structural conformations. Moreover, surface tunable properties of these proteins help to bind a range of anticancer drugs and target ligands, thus making them efficient delivery agents in cancer therapy. Here, we discuss various proteins obtained from common exogenous sources and how they transform into effective anticancer agents. We also comprehensively discuss the tumor-killing mechanisms of different dietary proteins such as bovine α-lactalbumin, hen egg-white lysozyme, and their conjugates. We also articulate how protein nanostructures can be used as carriers for delivering cancer drugs and theranostics, and strategies to be adopted for improving their in vivo delivery and targeting. We further discuss the FDA-approved protein-based anticancer formulations along with those in different phases of clinical trials.
Collapse
|
13
|
Sevieri M, Pinori M, Chesi A, Bonizzi A, Sitia L, Truffi M, Morasso C, Corsi F, Mazzucchelli S. Novel Bioengineering Strategies to Improve Bioavailability and In Vivo Circulation of H-Ferritin Nanocages by Surface Functionalization. ACS OMEGA 2023; 8:7244-7251. [PMID: 36873018 PMCID: PMC9979315 DOI: 10.1021/acsomega.2c07794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 02/09/2023] [Indexed: 06/18/2023]
Abstract
Due to its unique architecture and innate capability to specifically target cancer cells, ferritin has emerged as an attractive class of biomaterials for drug delivery. In many studies, various chemotherapeutics have been loaded into ferritin nanocages constituted by H-chains of ferritin (HFn), and their related anti-tumor efficacy has been explored by employing different strategies. Despite the multiple advantages and the versatility of HFn-based nanocages, there are still many challenges to face for their reliable implementation as drug nanocarriers in the process of clinical translation. This review aims at providing an overview of the significant efforts expended during recent years to maximize the features of HFn in terms of increased stability and in vivo circulation. The most considerable modification strategies explored to improve bioavailability and pharmacokinetics profiles of HFn-based nanosystems will be discussed herein.
Collapse
Affiliation(s)
- Marta Sevieri
- Nanomedicine
Laboratory, Department of Biomedical and Clinical Sciences, Università degli Studi di Milano, 20157 Milan, Italy
| | - Mattia Pinori
- Nanomedicine
Laboratory, Department of Biomedical and Clinical Sciences, Università degli Studi di Milano, 20157 Milan, Italy
| | - Arianna Chesi
- Nanomedicine
Laboratory, Department of Biomedical and Clinical Sciences, Università degli Studi di Milano, 20157 Milan, Italy
| | - Arianna Bonizzi
- Nanomedicine
Laboratory, Department of Biomedical and Clinical Sciences, Università degli Studi di Milano, 20157 Milan, Italy
| | - Leopoldo Sitia
- Nanomedicine
Laboratory, Department of Biomedical and Clinical Sciences, Università degli Studi di Milano, 20157 Milan, Italy
| | - Marta Truffi
- Nanomedicine
and Molecular Imaging Lab, Istituti Clinici
Scientifici Maugeri IRCCS, 27100 Pavia, Italy
| | - Carlo Morasso
- Nanomedicine
and Molecular Imaging Lab, Istituti Clinici
Scientifici Maugeri IRCCS, 27100 Pavia, Italy
| | - Fabio Corsi
- Nanomedicine
Laboratory, Department of Biomedical and Clinical Sciences, Università degli Studi di Milano, 20157 Milan, Italy
- Nanomedicine
and Molecular Imaging Lab, Istituti Clinici
Scientifici Maugeri IRCCS, 27100 Pavia, Italy
- Breast
Unit, Istituti Clinici Scientifici Maugeri
IRCCS, 27100 Pavia, Italy
| | - Serena Mazzucchelli
- Nanomedicine
Laboratory, Department of Biomedical and Clinical Sciences, Università degli Studi di Milano, 20157 Milan, Italy
| |
Collapse
|
14
|
Ferritin - a multifaceted protein scaffold for biotherapeutics. Exp Mol Med 2022; 54:1652-1657. [PMID: 36192487 PMCID: PMC9527718 DOI: 10.1038/s12276-022-00859-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 06/21/2022] [Accepted: 06/28/2022] [Indexed: 12/29/2022] Open
Abstract
The ferritin nanocage is an endogenous protein that exists in almost all mammals. Its hollow spherical structure that naturally stores iron ions has been diversely exploited by researchers in biotherapeutics. Ferritin has excellent biosafety profiles, and the nanosized particles exhibit rapid dispersion and controlled/sustained release pharmacokinetics. Moreover, the large surface-to-volume ratio and the disassembly/reassembly behavior of the 24 monomer subunits into a sphere allow diverse modifications by chemical and genetic methods on the surface and inner cage of ferritin. Here, we critically review ferritin and its applications. We (i) introduce the application of ferritin in drug delivery; (ii) present an overview of the use of ferritin in imaging and diagnosis for biomedical purposes; (iii) discuss ferritin-based vaccines; and (iv) review ferritin-based agents currently in clinical trials. Although there are no currently approved drugs based on ferritin, this multifunctional protein scaffold shows immense potential in drug development in diverse categories, and ferritin-based drugs have recently entered phase I clinical trials. This golden shortlist of recent developments will be of immediate benefit and interest to researchers studying ferritin and other protein-based biotherapeutics.
Collapse
|
15
|
Lee C. Development of Injectable and Biodegradable Needle-Type Starch Implant for Effective Intratumoral Drug Delivery and Distribution. Int J Nanomedicine 2022; 17:4307-4319. [PMID: 36147547 PMCID: PMC9488191 DOI: 10.2147/ijn.s370194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 08/21/2022] [Indexed: 11/26/2022] Open
Abstract
Introduction Compared to intravenous administration, intratumoral drug administration enables the direct delivery of drugs to tumors and mitigates the systemic absorption of drugs and associated drug-induced side effects. However, intratumoral drug administration presents several challenges. The high interstitial fluid pressure (IFP) of the tumor prevents the retention of drugs within the tumor; thus, significant amounts of the drugs are absorbed systemically through the bloodstream or delivered to non-target sites. To solve this problem, in this study, a drug-enclosed needle-type starch implant was developed that can overcome IFP and remain in the tumor. Methods Injectable needle-type starch implants (NS implants) were prepared by starch gelatinization and drying. The structure, cytotoxicity, and anticancer effects of the NS implants were evaluated. Biodistribution of NS implants was evaluated in pork (in vitro), dissected liver (ex vivo), and 4T1 tumors in mice (in vivo) using a fluorescence imaging device. Results The prepared NS implants exhibited a hydrogel structure after water absorption. NS implants showed effective cytotoxicity and anticancer effects by photothermal therapy (PTT). The NS implant itself has sufficient strength and can be easily injected into a desired area. In vivo, the NS implant continuously delivered drugs to the tumor more effectively and uniformly than conventional hydrogels and solutions. Conclusion This study demonstrated the advantages of needle-type implants. An injectable NS implant can be a new formulation that can effectively deliver drugs and exhibit anticancer effects.
Collapse
Affiliation(s)
- Changkyu Lee
- Department of Biopharmaceutical Engineering, Division of Chemistry and Biotechnology, Dongguk University, Gyeongju, Korea
| |
Collapse
|
16
|
Abstract
Lymph node mapping for tumor micrometastasis is of great significance for the prevention, prognosis, and treatment of cancer. Currently, the traditional clinical detection methods (computed tomography, magnetic resonance imaging, or positron emission tomography/computed tomography) in clinical lymph node mapping still have some inherent disadvantages, which have prompted the development of various fluorescent probes for lymph node mapping. However, the conventional fluorescent probes such as indocyanine green or methylene blue in lymph node mapping are still accompanied by several problems such as impaired surgical field vision due to dye staining or less accumulation and shorter retention time in the lymph node. In a recent achievement, newly designed nanoparticles are prepared with novel properties that could be attractive for lymph node mapping. In this review, we will provide details on the progress of various nanoparticles for lymph node mapping and emphasize other multivariant properties in different nanoparticles, including strong tumor-targeting affinity and specificity, self-luminescence, and even with the function to kill metastatic cancer cells.
Collapse
Affiliation(s)
- Meng Han
- Queen Mary School, Nanchang University, Nanchang, Jiangxi Province 330006, P.R. China
| | - Ruirui Kang
- The Department of Ultrasound, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province 330006, P.R. China
| | - Chunquan Zhang
- The Department of Ultrasound, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province 330006, P.R. China
| |
Collapse
|
17
|
Sottani C, Grignani E, Cottica D, Mazzucchelli S, Sevieri M, Chesi A, Corsi F, Galfrè S, Robustelli Della Cuna FS, Calleri E. Development and Validation of a Bioanalytical UHPLC-MS/MS Method Applied to Murine Liver Tissue for the Determination of Indocyanine Green Loaded in H-Ferritin Nanoparticles. Front Chem 2022; 9:784123. [PMID: 35047479 PMCID: PMC8762227 DOI: 10.3389/fchem.2021.784123] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 11/19/2021] [Indexed: 11/13/2022] Open
Abstract
Indocyanine green (ICG) is one of the most commonly used fluorophores in near-infrared fluorescence-guided techniques. However, the molecule is prone to form aggregates in saline solution with a limited photostability and a moderate fluorescence yield. ICG was thus formulated using protein-based nanoparticles of H-ferritin (HFn) in order to generate a new nanostructure, HFn-ICG. In this study, an ultrahigh performance liquid chromatography-tandem mass spectrometry (UHPLC-MS/MS) system was employed to develop and validate the quantitative analysis of ICG in liver tissue samples from HFn-ICG-treated mice. To precipitate HFn, cold acetone in acidic solution at pH 5.0 was used. The processed liver samples were injected into the UHPLC-MS/MS system for analysis using the positive electrospray ionization mode. Chromatographic separation was achieved on a Waters Acquity UPLC® HSS T3 Column (1.8 μm, 2.1 × 100 mm) with 0.1% formic acid and acetonitrile as the mobile phase with gradient elution. The selected reaction monitoring transitions of m / z 753 → m / z 330 and m / z 827 → m / z 330 were applied for ICG and IR-820 (the internal standard, IS), respectively. The method was selective and linear over a concentration range of 50-1,500 ng/ml. The method was validated for sensitivity, accuracy, precision, extraction recovery, matrix effect, and stability in liver tissue homogenates. ICG extraction recoveries ranged between 85 and 108%. The intra- and inter-day precisions were less than 6.28%. The method was applied to a bio-distribution study to compare the amount of ICG levels from mice treated with HFn-ICG and free ICG. The analyses of the homogenate samples from the two types of treatment showed that the concentration levels of ICG is approximately six-fold higher than those of free ICG (1,411 ± 7.62 ng/ml vs. 235 ± 26.0 ng/ml) at 2 h post injection.
Collapse
Affiliation(s)
- Cristina Sottani
- Environmental Research Center, Istituti Clinici Scientifici Maugeri IRCCS, Pavia, Italy
| | - Elena Grignani
- Environmental Research Center, Istituti Clinici Scientifici Maugeri IRCCS, Pavia, Italy
| | - Danilo Cottica
- Environmental Research Center, Istituti Clinici Scientifici Maugeri IRCCS, Pavia, Italy
| | - Serena Mazzucchelli
- Nanomedicine Laboratory, Department of Biomedical and Clinical Sciences "Luigi Sacco", Milano University, Milan, Italy
| | - Marta Sevieri
- Nanomedicine Laboratory, Department of Biomedical and Clinical Sciences "Luigi Sacco", Milano University, Milan, Italy
| | - Arianna Chesi
- Nanomedicine Laboratory, Department of Biomedical and Clinical Sciences "Luigi Sacco", Milano University, Milan, Italy
| | - Fabio Corsi
- Nanomedicine Laboratory, Department of Biomedical and Clinical Sciences "Luigi Sacco", Milano University, Milan, Italy.,Breast Unit, Istituti Clinici Scientifici Maugeri IRCCS, Pavia, Italy
| | - Sarah Galfrè
- Department of Drug Sciences, University of Pavia, Pavia, Italy
| | | | - Enrica Calleri
- Department of Drug Sciences, University of Pavia, Pavia, Italy
| |
Collapse
|
18
|
Mainini F, Bonizzi A, Sevieri M, Sitia L, Truffi M, Corsi F, Mazzucchelli S. Protein-Based Nanoparticles for the Imaging and Treatment of Solid Tumors: The Case of Ferritin Nanocages, a Narrative Review. Pharmaceutics 2021; 13:pharmaceutics13122000. [PMID: 34959283 PMCID: PMC8708614 DOI: 10.3390/pharmaceutics13122000] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 11/18/2021] [Accepted: 11/19/2021] [Indexed: 12/13/2022] Open
Abstract
Protein nanocages have been studied extensively, due to their unique architecture, exceptional biocompatibility and highly customization capabilities. In particular, ferritin nanocages (FNs) have been employed for the delivery of a vast array of molecules, ranging from chemotherapeutics to imaging agents, among others. One of the main favorable characteristics of FNs is their intrinsic targeting efficiency toward the Transferrin Receptor 1, which is overexpressed in many tumors. Furthermore, genetic manipulation can be employed to introduce novel variants that are able to improve the loading capacity, targeting capabilities and bio-availability of this versatile drug delivery system. In this review, we discuss the main characteristics of FN and the most recent applications of this promising nanotechnology in the field of oncology with a particular emphasis on the imaging and treatment of solid tumors.
Collapse
Affiliation(s)
- Francesco Mainini
- Dipartimento di Scienze Biomediche e Cliniche “L. Sacco”, Università di Milano, 20157 Milano, Italy; (F.M.); (A.B.); (M.S.); (L.S.)
| | - Arianna Bonizzi
- Dipartimento di Scienze Biomediche e Cliniche “L. Sacco”, Università di Milano, 20157 Milano, Italy; (F.M.); (A.B.); (M.S.); (L.S.)
| | - Marta Sevieri
- Dipartimento di Scienze Biomediche e Cliniche “L. Sacco”, Università di Milano, 20157 Milano, Italy; (F.M.); (A.B.); (M.S.); (L.S.)
| | - Leopoldo Sitia
- Dipartimento di Scienze Biomediche e Cliniche “L. Sacco”, Università di Milano, 20157 Milano, Italy; (F.M.); (A.B.); (M.S.); (L.S.)
| | - Marta Truffi
- Istituti Clinici Scientifici Maugeri IRCCS, 27100 Pavia, Italy;
| | - Fabio Corsi
- Dipartimento di Scienze Biomediche e Cliniche “L. Sacco”, Università di Milano, 20157 Milano, Italy; (F.M.); (A.B.); (M.S.); (L.S.)
- Istituti Clinici Scientifici Maugeri IRCCS, 27100 Pavia, Italy;
- Correspondence: (F.C.); (S.M.)
| | - Serena Mazzucchelli
- Dipartimento di Scienze Biomediche e Cliniche “L. Sacco”, Università di Milano, 20157 Milano, Italy; (F.M.); (A.B.); (M.S.); (L.S.)
- Correspondence: (F.C.); (S.M.)
| |
Collapse
|
19
|
Bishnoi S, Rehman S, Dutta SB, De SK, Chakraborty A, Nayak D, Gupta S. Optical-Property-Enhancing Novel Near-Infrared Active Niosome Nanoformulation for Deep-Tissue Bioimaging. ACS OMEGA 2021; 6:22616-22624. [PMID: 34514233 PMCID: PMC8427633 DOI: 10.1021/acsomega.1c02632] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Accepted: 08/11/2021] [Indexed: 05/17/2023]
Abstract
Indocyanine green (ICG) is a clinically approved near-infrared (NIR) contrast agent used in medical diagnosis. However, ICG has not been used to its fullest for biomedical imaging applications due to its low fluorescence quantum yield, aqueous instability, concentration-dependent aggregation, and photo and thermal degradations, leading to quenching of its fluorescence emission. In the present study, a nanosized niosomal formulation, ICGNiosomes (ICGNios), is fabricated to encapsulate and protect ICG from degradation. Interestingly, compared to free ICG, the ICGNios exhibited higher fluorescence quantum yield and fluorescence emission with a bathochromic shift. Also, ICGNios nanoparticles are biocompatible, biodegradable, and readily uptaken by the cells. Furthermore, ICGNios show more enhanced fluorescence intensity through ∼1 cm thick chicken breast tissue compared to free ICG, which showed minimal emission through the same thickness of tissue. Our results suggest that ICGNios could offer a promising platform for deep-tissue NIR in vivo imaging to visualize inaccessible tissue microstructures for disease diagnosis and therapeutics.
Collapse
Affiliation(s)
- Suman Bishnoi
- Department
of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Indore 453552 Madhya Pradesh, India
| | - Sheeba Rehman
- Department
of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Indore 453552 Madhya Pradesh, India
| | - Surjendu Bikash Dutta
- Department
of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Indore 453552 Madhya Pradesh, India
| | - Soumya Kanti De
- Department
of Chemistry, Indian Institute of Technology
Indore, Indore 453552 Madhya Pradesh, India
| | - Anjan Chakraborty
- Department
of Chemistry, Indian Institute of Technology
Indore, Indore 453552 Madhya Pradesh, India
| | - Debasis Nayak
- Department
of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Indore 453552 Madhya Pradesh, India
| | - Sharad Gupta
- Department
of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Indore 453552 Madhya Pradesh, India
- School
of Biotechnology, Jawaharlal Nehru University, New Delhi 110067, India
| |
Collapse
|
20
|
Song N, Zhang J, Zhai J, Hong J, Yuan C, Liang M. Ferritin: A Multifunctional Nanoplatform for Biological Detection, Imaging Diagnosis, and Drug Delivery. Acc Chem Res 2021; 54:3313-3325. [PMID: 34415728 DOI: 10.1021/acs.accounts.1c00267] [Citation(s) in RCA: 83] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Ferritins are spherical iron storage proteins within cells that are composed of a combination of 24 subunits of two types, heavy-chain ferritin (HFn) and light-chain ferritin (LFn). They autoassemble naturally into a spherical hollow nanocage with an outer diameter of 12 nm and an interior cavity that is 8 nm in diameter. In recent years, with the constantly emerging safety issues and the concerns about unfavorable uniformity and indefinite in vivo behavior of traditional nanomedicines, the characteristics of native ferritin nanocages, such as the unique nanocage structure, excellent safety profile, and definite in vivo behavior, make ferritin-based formulations uniquely attractive for nanomedicine development. To date, a variety of cargo molecules, including therapeutic drugs (e.g., cisplatin, carboplatin, paclitaxel, curcumin, atropine, quercetin, gefitinib, daunomycin, epirubicin, doxorubicin, etc.), imaging agents (e.g., fluorescence dyes, radioisotopes, and MRI contrast agents), nucleic acids (e.g., siRNA and miRNA), and metal nanoparticles (e.g., Fe3O4, CeO2, AuPd, CuS, CoPt, FeCo, Ag, etc.) have been loaded into the interior cavity of ferritin nanocages for a broad range of biomedical applications from in vitro biosensing to targeted delivery of cargo molecules in living systems with the aid of modified targeting ligands either genetically or chemically. We reported that human HFn could selectively deliver a large amount of cargo into tumors in vivo via transferrin receptor 1 (TfR1)-mediated tumor-cell-specific targeting followed by rapid internalization. By the use of the intrinsic tumor-targeting property and unique nanocage structure of human HFn, a broad variety of cargo-loaded HFn formulations have been developed for biological analysis, imaging diagnosis, and medicine development. In view of the intrinsic tumor-targeting property, unique nanocage structure, lack of immunogenicity, and definite in vivo behavior, human HFn holds promise to promote therapeutic drugs, diagnostic imaging agents, and targeting moieties into multifunctional nanomedicines.Since the report of the intrinsic tumor-targeting property of human HFn, we have extensively explored human HFn as an ideal nanocarrier for tumor-targeted delivery of anticancer drugs, MRI contrast agents, inorganic nanoparticles, and radioisotopes. In particular, by the use of genetic tools, we also have genetically engineered human HFn nanocages to recognize a broader range of disease biomarkers. In this Account, we systematically review human ferritins from characterizing their tumor-binding property and understanding their mechanism and kinetics for cargo loading to exploring their biomedical applications. We finally discuss the prospect of ferritin-based formulations to become next-generation nanomedicines. We expect that ferritin formulations with unique physicochemical characteristics and intrinsic tumor-targeting property will attract broad interest in fundamental drug research and offer new opportunities for nanomedicine development.
Collapse
Affiliation(s)
- Ningning Song
- Experimental Center of Advanced Materials, School of Materials Science & Engineering, Beijing Institute of Technology, Beijing 100081, China
| | - Jianlin Zhang
- Experimental Center of Advanced Materials, School of Materials Science & Engineering, Beijing Institute of Technology, Beijing 100081, China
| | - Jiao Zhai
- Tung Foundation Biomedical Sciences Centre/Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| | - Juanji Hong
- Experimental Center of Advanced Materials, School of Materials Science & Engineering, Beijing Institute of Technology, Beijing 100081, China
| | - Chang Yuan
- Experimental Center of Advanced Materials, School of Materials Science & Engineering, Beijing Institute of Technology, Beijing 100081, China
| | - Minmin Liang
- Experimental Center of Advanced Materials, School of Materials Science & Engineering, Beijing Institute of Technology, Beijing 100081, China
| |
Collapse
|
21
|
David KI, Ravikumar TS, Sethuraman S, Krishnan UM. Development and evaluation of a multi-functional organic-inorganic nanotheranostic hybrid for pancreatic cancer therapy. Biomed Mater 2021; 16. [PMID: 34298521 DOI: 10.1088/1748-605x/ac177c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 07/23/2021] [Indexed: 11/12/2022]
Abstract
Pancreatic cancer is a highly invasive disease with low survival rates. The high death rates associated with pancreatic cancer are due to multiple factors including late stage diagnosis, multi-drug resistance, invasive nature and restricted access of the therapeutic moiety to the cancer cells due to the stroma. Smart multifunctional nanocarriers that deliver the therapeutic agent in to the cancer tissue as well as enable imaging of the tissue represent an emerging paradigm in cancer therapy. Accurate and reliable detection of cancerous lesions in pancreas is essential for designing appropriate therapeutic strategy to annihilate the highly aggressive pancreatic cancer. A combination of imaging modalities can enhance the reliability of cancer detection. In this context, we report here a hybrid iron oxide-gold nanoparticle with dual contrast enhancing ability for both magnetic resonance imaging (MRI) and micro-computed tomography (micro-CT) that is co-encapsulated with the nucleotide analogue gemcitabine in a chitosan matrix. The theranostic system displayed enhanced cytotoxicity against PanC-1 pancreatic cancer cells when compared to normal cells over 48 h due to differences in cell internalization. The iron oxide-gold hybrid enabled visualization of the theranostic nanoparticle by MRI as well as micro-CT. Further, the magnetocaloric effect of the iron oxide enabled faster release of the chemotherapeutic agent as well as augmented the cytotoxicity by inducing hyperthermia. This system holds promise for further exploration as an integrated diagnostic and therapeutic platform for pancreatic cancer.
Collapse
Affiliation(s)
- Karolyn Infanta David
- Centre for Nanotechnology & Advanced Biomaterials, SASTRA Deemed University, Thanjavur 613401, India.,School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur 613401, India
| | - T S Ravikumar
- Sri Venkateswara Institute of Medical Sciences (SVIMS), Tirupati 517507, India
| | - Swaminathan Sethuraman
- Centre for Nanotechnology & Advanced Biomaterials, SASTRA Deemed University, Thanjavur 613401, India.,School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur 613401, India
| | - Uma Maheswari Krishnan
- Centre for Nanotechnology & Advanced Biomaterials, SASTRA Deemed University, Thanjavur 613401, India.,School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur 613401, India.,School of Arts, Science & Humanities, SASTRA Deemed University, Thanjavur 613401, India
| |
Collapse
|
22
|
Picchio ML, Bergueiro J, Wedepohl S, Minari RJ, Alvarez Igarzabal CI, Gugliotta LM, Cuggino JC, Calderón M. Exploiting cyanine dye J-aggregates/monomer equilibrium in hydrophobic protein pockets for efficient multi-step phototherapy: an innovative concept for smart nanotheranostics. NANOSCALE 2021; 13:8909-8921. [PMID: 33954311 DOI: 10.1039/d0nr09058a] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
After several decades of development in the field of near-infrared (NIR) dyes for photothermal therapy (PTT), indocyanine green (ICG) still remains the only FDA-approved NIR contrast agent. However, upon NIR light irradiation ICG can react with molecular oxygen to form reactive oxygen species and degrade the ICG core, losing the convenient dye properties. In this work, we introduce a new approach for expanding the application of ICG in nanotheranostics, which relies on the confinement of self-organized J-type aggregates in hydrophobic protein domains acting as monomer depots. Upon the fast photobleaching, while the dye is irradiated, this strategy permits the equilibrium-driven monomer replacement after each irradiation cycle that radically increases the systems' effectivity and applicability. Gadolinium-doped casein micelles were designed to prove this novel concept at the same time as endowing the nanosystems with further magnetic resonance imaging (MRI) ability for dual-modal imaging-guided PTT. By teaching a new trick to a very old dog, the clinical prospect of ICG will undoubtedly be boosted laying the foundation for novel therapeutics. It is anticipated that future research could be expanded to other relevant J-aggregates-forming cyanine dyes or nanocrystal formulations of poorly water-soluble photosensitizers.
Collapse
Affiliation(s)
- Matías L Picchio
- Departamento de Química Orgánica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, IPQA, CONICET-UNC, Haya de la Torre y Medina Allende. Ciudad Universitaria, Córdoba, X5000 HUA, Argentina
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Kaku TS, Lim S. Protein nanoparticles in molecular, cellular, and tissue imaging. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2021; 13:e1714. [PMID: 33821568 DOI: 10.1002/wnan.1714] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 02/12/2021] [Accepted: 03/08/2021] [Indexed: 01/10/2023]
Abstract
The quest to develop ideal nanoparticles capable of molecular, cellular, and tissue level imaging is ongoing. Since certain imaging probes and nanoparticles face drawbacks such as low aqueous solubility, increased ROS generation leading to DNA damage, apoptosis, and high cellular/organ toxicities, the development of versatile and biocompatible nanocarriers becomes necessary. Protein nanoparticles (PNPs) are one such promising class of nanocarriers that possess most of the desirable properties of an ideal nanocarrier for bioimaging applications. PNPs demonstrate high aqueous solubility, minimal cytotoxicity, and multi-cargo loading capacity. They are also amenable to surface-functionalization, as well as modulation of their hydrophobicity and hydrophilicity. The use of PNPs for bioimaging applications has made rapid advancements in the past two decades. Being comparatively less explored, the field opens up a plethora of opportunities and focus areas to engineer ideal bioimaging protein nanocarriers. The use of PNPs as carriers of their natural ligands as well as other heavy metals and fluorescent probes, along with drug molecules for combined theranostic applications has been reported. In addition, surface functionalization to impart specificity of targeting the PNPs has been shown to reduce nonspecific cellular interactions, thus reducing systemic toxicity. PNPs have been explored for their application in imaging of numerous cancers, cardiovascular diseases as well as imaging of the brain using near infrared fluorescence (NIRF) imaging, magnetic resonance imaging (MRI), X-ray computed tomography (CT), positron emission tomography (PET), single-photon emission computed tomography (SPECT), ultrasound (US), and photoacoustic (PA) imaging. This article is categorized under: Biology-Inspired Nanomaterials > Protein and Virus-Based Structures Diagnostic Tools > In Vivo Nanodiagnostics and Imaging.
Collapse
Affiliation(s)
- Tanvi Sushil Kaku
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore
| | - Sierin Lim
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore
| |
Collapse
|
24
|
Silva F, Sitia L, Allevi R, Bonizzi A, Sevieri M, Morasso C, Truffi M, Corsi F, Mazzucchelli S. Combined Method to Remove Endotoxins from Protein Nanocages for Drug Delivery Applications: The Case of Human Ferritin. Pharmaceutics 2021; 13:pharmaceutics13020229. [PMID: 33562060 PMCID: PMC7915212 DOI: 10.3390/pharmaceutics13020229] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 01/27/2021] [Accepted: 01/28/2021] [Indexed: 12/13/2022] Open
Abstract
Protein nanocages represent an emerging candidate among nanoscaled delivery systems. Indeed, they display unique features that proved to be very interesting from the nanotechnological point of view such as uniform structure, stability in biological fluids, suitability for surface modification to insert targeting moieties and loading with different drugs and dyes. However, one of the main concerns regards the production as recombinant proteins in E. coli, which leads to a product with high endotoxin contamination, resulting in nanocage immunogenicity and pyrogenicity. Indeed, a main challenge in the development of protein-based nanoparticles is finding effective procedures to remove endotoxins without affecting protein stability, since every intravenous injectable formulation that should be assessed in preclinical and clinical phase studies should display endotoxins concentration below the admitted limit of 5 EU/kg. Different strategies could be employed to achieve such a result, either by using affinity chromatography or detergents. However, these strategies are not applicable to protein nanocages as such and require implementations. Here we propose a combined protocol to remove bacterial endotoxins from nanocages of human H-ferritin, which is one of the most studied and most promising protein-based drug delivery systems. This protocol couples the affinity purification with the Endotrap HD resin to a treatment with Triton X-114. Exploiting this protocol, we were able to obtain excellent levels of purity maintaining good protein recovery rates, without affecting nanocage interactions with target cells. Indeed, binding assay and confocal microscopy experiments confirm that purified H-ferritin retains its capability to specifically recognize cancer cells. This procedure allowed to obtain injectable formulations, which is preliminary to move to a clinical trial.
Collapse
Affiliation(s)
- Filippo Silva
- Dipartimento di Scienze Biomediche e Cliniche “L. Sacco”, Università di Milano, 20157 Milano, Italy; (F.S.); (L.S.); (R.A.); (A.B.); (M.S.)
| | - Leopoldo Sitia
- Dipartimento di Scienze Biomediche e Cliniche “L. Sacco”, Università di Milano, 20157 Milano, Italy; (F.S.); (L.S.); (R.A.); (A.B.); (M.S.)
| | - Raffaele Allevi
- Dipartimento di Scienze Biomediche e Cliniche “L. Sacco”, Università di Milano, 20157 Milano, Italy; (F.S.); (L.S.); (R.A.); (A.B.); (M.S.)
| | - Arianna Bonizzi
- Dipartimento di Scienze Biomediche e Cliniche “L. Sacco”, Università di Milano, 20157 Milano, Italy; (F.S.); (L.S.); (R.A.); (A.B.); (M.S.)
| | - Marta Sevieri
- Dipartimento di Scienze Biomediche e Cliniche “L. Sacco”, Università di Milano, 20157 Milano, Italy; (F.S.); (L.S.); (R.A.); (A.B.); (M.S.)
| | - Carlo Morasso
- Istituti Clinici Scientifici Maugeri IRCCS, 27100 Pavia, Italy; (C.M.); (M.T.)
| | - Marta Truffi
- Istituti Clinici Scientifici Maugeri IRCCS, 27100 Pavia, Italy; (C.M.); (M.T.)
| | - Fabio Corsi
- Dipartimento di Scienze Biomediche e Cliniche “L. Sacco”, Università di Milano, 20157 Milano, Italy; (F.S.); (L.S.); (R.A.); (A.B.); (M.S.)
- Istituti Clinici Scientifici Maugeri IRCCS, 27100 Pavia, Italy; (C.M.); (M.T.)
- Correspondence: (F.C.); (S.M.)
| | - Serena Mazzucchelli
- Dipartimento di Scienze Biomediche e Cliniche “L. Sacco”, Università di Milano, 20157 Milano, Italy; (F.S.); (L.S.); (R.A.); (A.B.); (M.S.)
- Correspondence: (F.C.); (S.M.)
| |
Collapse
|
25
|
Tumor Accumulation and Off-Target Biodistribution of an Indocyanine-Green Fluorescent Nanotracer: An Ex Vivo Study on an Orthotopic Murine Model of Breast Cancer. Int J Mol Sci 2021; 22:ijms22041601. [PMID: 33562574 PMCID: PMC7915532 DOI: 10.3390/ijms22041601] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 02/01/2021] [Accepted: 02/02/2021] [Indexed: 12/15/2022] Open
Abstract
Indocyanine green (ICG) is a near infrared fluorescent tracer used in image-guided surgery to assist surgeons during resection. Despite appearing as a very promising tool for surgical oncology, its employment in this area is limited to lymph node mapping or to laparoscopic surgery, as it lacks tumor targeting specificity. Recently, a nanoformulation of this dye has been proposed with the aim toward tumor targeting specificity in order to expand its employment in surgical oncology. This nanosystem is constituted by 24 monomers of H-Ferritin (HFn), which self-assemble into a spherical cage structure enclosing the indocyanine green fluorescent tracer. These HFn nanocages were demonstrated to display tumor homing due to the specific interaction between the HFn nanocage and transferrin receptor 1, which is overexpressed in most tumor tissues. Here, we provide an ex vivo detailed comparison between the biodistribution of this nanotracer and free ICG, combining the results obtained with the Karl Storz endoscope that is currently used in clinical practice and the quantification of the ICG signal derived from the fluorescence imaging system IVIS Lumina II. These insights demonstrate the suitability of this novel HFn-based nanosystem in fluorescence-guided oncological surgery.
Collapse
|
26
|
Sitia L, Bonizzi A, Mazzucchelli S, Negri S, Sottani C, Grignani E, Rizzuto MA, Prosperi D, Sorrentino L, Morasso C, Allevi R, Sevieri M, Silva F, Truffi M, Corsi F. Selective Targeting of Cancer-Associated Fibroblasts by Engineered H-Ferritin Nanocages Loaded with Navitoclax. Cells 2021; 10:328. [PMID: 33562504 PMCID: PMC7915356 DOI: 10.3390/cells10020328] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 01/29/2021] [Accepted: 02/02/2021] [Indexed: 12/13/2022] Open
Abstract
Cancer-associated fibroblasts (CAFs) are key actors in regulating cancer progression. They promote tumor growth, metastasis formation, and induce drug resistance. For these reasons, they are emerging as potential therapeutic targets. Here, with the aim of developing CAF-targeted drug delivery agents, we functionalized H-ferritin (HFn) nanocages with fibroblast activation protein (FAP) antibody fragments. Functionalized nanocages (HFn-FAP) have significantly higher binding with FAP+ CAFs than with FAP- cancer cells. We loaded HFn-FAP with navitoclax (Nav), an experimental Bcl-2 inhibitor pro-apoptotic drug, whose clinical development is limited by its strong hydrophobicity and toxicity. We showed that Nav is efficiently loaded into HFn (HNav), maintaining its mechanism of action. Incubating Nav-loaded functionalized nanocages (HNav-FAP) with FAP+ cells, we found significantly higher cytotoxicity as compared to non-functionalized HNav. This was correlated with a significantly higher drug release only in FAP+ cells, confirming the specific targeting ability of functionalized HFn. Finally, we showed that HFn-FAP is able to reach the tumor and to target CAFs in a mouse syngeneic model of triple negative breast cancer after intravenous administration. Our data show that HNav-FAP could be a promising tool to enhance specific drug delivery into CAFs, thus opening new therapeutic possibilities focused on tumor microenvironment.
Collapse
Affiliation(s)
- Leopoldo Sitia
- Dipartimento di Scienze Biomediche e Cliniche “L. Sacco”, Università di Milano, 20157 Milan, Italy; (L.S.); (A.B.); (S.M.); (R.A.); (M.S.); (F.S.)
| | - Arianna Bonizzi
- Dipartimento di Scienze Biomediche e Cliniche “L. Sacco”, Università di Milano, 20157 Milan, Italy; (L.S.); (A.B.); (S.M.); (R.A.); (M.S.); (F.S.)
| | - Serena Mazzucchelli
- Dipartimento di Scienze Biomediche e Cliniche “L. Sacco”, Università di Milano, 20157 Milan, Italy; (L.S.); (A.B.); (S.M.); (R.A.); (M.S.); (F.S.)
| | - Sara Negri
- Istituti Clinici Scientifici Maugeri IRCCS, 27100 Pavia, Italy; (S.N.); (C.S.); (E.G.); (C.M.)
| | - Cristina Sottani
- Istituti Clinici Scientifici Maugeri IRCCS, 27100 Pavia, Italy; (S.N.); (C.S.); (E.G.); (C.M.)
| | - Elena Grignani
- Istituti Clinici Scientifici Maugeri IRCCS, 27100 Pavia, Italy; (S.N.); (C.S.); (E.G.); (C.M.)
| | - Maria Antonietta Rizzuto
- Department of Biotechnology and Biosciences, University of Milan-Bicocca, 20126 Milan, Italy; (M.A.R.); (D.P.)
| | - Davide Prosperi
- Department of Biotechnology and Biosciences, University of Milan-Bicocca, 20126 Milan, Italy; (M.A.R.); (D.P.)
| | - Luca Sorrentino
- Colorectal Surgery Unit, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, 20133 Milan, Italy;
| | - Carlo Morasso
- Istituti Clinici Scientifici Maugeri IRCCS, 27100 Pavia, Italy; (S.N.); (C.S.); (E.G.); (C.M.)
| | - Raffaele Allevi
- Dipartimento di Scienze Biomediche e Cliniche “L. Sacco”, Università di Milano, 20157 Milan, Italy; (L.S.); (A.B.); (S.M.); (R.A.); (M.S.); (F.S.)
| | - Marta Sevieri
- Dipartimento di Scienze Biomediche e Cliniche “L. Sacco”, Università di Milano, 20157 Milan, Italy; (L.S.); (A.B.); (S.M.); (R.A.); (M.S.); (F.S.)
| | - Filippo Silva
- Dipartimento di Scienze Biomediche e Cliniche “L. Sacco”, Università di Milano, 20157 Milan, Italy; (L.S.); (A.B.); (S.M.); (R.A.); (M.S.); (F.S.)
| | - Marta Truffi
- Istituti Clinici Scientifici Maugeri IRCCS, 27100 Pavia, Italy; (S.N.); (C.S.); (E.G.); (C.M.)
| | - Fabio Corsi
- Dipartimento di Scienze Biomediche e Cliniche “L. Sacco”, Università di Milano, 20157 Milan, Italy; (L.S.); (A.B.); (S.M.); (R.A.); (M.S.); (F.S.)
- Istituti Clinici Scientifici Maugeri IRCCS, 27100 Pavia, Italy; (S.N.); (C.S.); (E.G.); (C.M.)
| |
Collapse
|