1
|
Gangadaran P, Onkar A, Rajendran RL, Goenka A, Oh JM, Khan F, Nagarajan AK, Muthu S, Krishnan A, Hong CM, Ahn BC. Noninvasive in vivo imaging of macrophages: understanding tumor microenvironments and delivery of therapeutics. Biomark Res 2025; 13:20. [PMID: 39865337 PMCID: PMC11770947 DOI: 10.1186/s40364-025-00735-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 01/21/2025] [Indexed: 01/28/2025] Open
Abstract
Macrophages are pivotal in the body's defense and response to inflammation. They are present in significant numbers and are widely implicated in various diseases, including cancer. While molecular and histological techniques have advanced our understanding of macrophage biology, their precise function within the cancerous microenvironments remains underexplored. Enhancing our knowledge of macrophages and the dynamics of their extracellular vesicles (EVs) in cancer development can potentially improve therapeutic management. Notably, macrophages have also been harnessed to deliver drugs. Noninvasive in vivo molecular imaging of macrophages is crucial for investigating intricate cellular processes, comprehending the underlying mechanisms of diseases, tracking cells and EVs' migration, and devising macrophage-dependent drug-delivery systems in living organisms. Thus, in vivo imaging of macrophages has become an indispensable tool in biomedical research. The integration of multimodal imaging approaches and the continued development of novel contrast agents hold promise for overcoming current limitations and expanding the applications of macrophage imaging. This study comprehensively reviews several methods for labeling macrophages and various imaging modalities, assessing the merits and drawbacks of each approach. The review concludes by offering insights into the applicability of molecular imaging techniques for real time monitoring of macrophages in preclinical and clinical scenarios.
Collapse
Affiliation(s)
- Prakash Gangadaran
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Sciences, School of Medicine, Kyungpook National University, Daegu, 41944, Korea
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu, 41944, Korea
- Cardiovascular Research Institute, Kyungpook National University, Daegu, 41944, Republic of Korea
| | - Akanksha Onkar
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Ramya Lakshmi Rajendran
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Sciences, School of Medicine, Kyungpook National University, Daegu, 41944, Korea
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu, 41944, Korea
- Cardiovascular Research Institute, Kyungpook National University, Daegu, 41944, Republic of Korea
| | - Anshika Goenka
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, 30322, USA
| | - Ji Min Oh
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu, 41944, Korea
- Cardiovascular Research Institute, Kyungpook National University, Daegu, 41944, Republic of Korea
| | - Fatima Khan
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
| | - ArulJothi Kandasamy Nagarajan
- Department of Genetic Engineering, SRM Institute of Science and Technology, Kattankulathur, Chengalpattu, 603203, Tamilnadu, India
| | - Sathish Muthu
- Department of Orthopaedics, Government Medical College, Tamil Nadu, 639004, Karur, India
- Department of Biotechnology, Faculty of Engineering, Karpagam Academy of Higher Education, Tamil Nadu, 641021, Coimbatore, India
| | - Anand Krishnan
- Precision Medicine and Integrated Nano-Diagnostics (P-MIND) Research Group, Office of the Dean, Faculty of Health Sciences, University of the Free State, Bloemfontein, 9300, South Africa
| | - Chae Moon Hong
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu, 41944, Korea.
- Cardiovascular Research Institute, Kyungpook National University, Daegu, 41944, Republic of Korea.
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, 41944, Korea.
| | - Byeong-Cheol Ahn
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Sciences, School of Medicine, Kyungpook National University, Daegu, 41944, Korea.
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu, 41944, Korea.
- Cardiovascular Research Institute, Kyungpook National University, Daegu, 41944, Republic of Korea.
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, 41944, Korea.
| |
Collapse
|
2
|
Lee K, Niku S, Koo SJ, Belezzuoli E, Guma M. Molecular imaging for evaluation of synovitis associated with osteoarthritis: a narrative review. Arthritis Res Ther 2024; 26:25. [PMID: 38229205 PMCID: PMC10790518 DOI: 10.1186/s13075-023-03258-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 12/28/2023] [Indexed: 01/18/2024] Open
Abstract
Recent evidence highlights the role of low-grade synovial inflammation in the progression of osteoarthritis (OA). Inflamed synovium of OA joints detected by imaging modalities are associated with subsequent progression of OA. In this sense, detecting and quantifying synovitis of OA by imaging modalities may be valuable in predicting OA progressors as well as in improving our understanding of OA progression. Of the several imaging modalities, molecular imaging such as positron emission tomography (PET) and single-photon emission computed tomography (SPECT) has an advantage of visualizing the cellular or subcellular events of the tissues. Depending on the radiotracers used, molecular imaging method can potentially detect and visualize various aspects of synovial inflammation. This narrative review summarizes the recent progresses of imaging modalities in assessing inflammation and OA synovitis and focuses on novel radiotracers. Recent studies about imaging modalities including ultrasonography (US), magnetic resonance imaging (MRI), and molecular imaging that were used to detect and quantify inflammation and OA synovitis are summarized. Novel radiotracers specifically targeting the components of inflammation have been developed. These tracers may show promise in detecting inflamed synovium of OA and help in expanding our understanding of OA progression.
Collapse
Affiliation(s)
- Kwanghoon Lee
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
- Department of Medicine, Dongguk University Ilsan Hospital, Goyang, Korea
| | - Soheil Niku
- Nuclear Medicine Service, Jennifer Moreno VA San Diego Healthcare System, San Diego, CA, USA
| | - Sonya J Koo
- Department of Radiology, West Los Angeles VA Medical Center, Los Angeles, CA, USA
| | - Ernest Belezzuoli
- Nuclear Medicine Service, Jennifer Moreno VA San Diego Healthcare System, San Diego, CA, USA
- Department of Radiology, University of California San Diego, La Jolla, CA, USA
| | - Monica Guma
- Department of Medicine, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
3
|
Morimoto M, Till NA, Bertozzi CR. Tumor Immune Cell Targeting Chimeras (TICTACs) For Targeted Depletion of Macrophage-Associated Checkpoint Receptors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.06.570444. [PMID: 38106036 PMCID: PMC10723462 DOI: 10.1101/2023.12.06.570444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
Immune cells in the tumor microenvironment are not only powerful regulators of immunosuppression and tumorigenesis, but also represent a dominant cell type, with tumor-associated macrophages (TAMs) comprising up to 50% of total cell mass in solid tumors. Immunotherapies such as immune checkpoint inhibitors (ICIs) derive their efficacy from this cancer-immune cell interface, however, immune-related adverse events resulting from systemic blockade remain a significant challenge. To address this need for potent, yet highly tumor-specific immunotherapies, we developed Tumor-Immune Cell Targeting Chimeras (TICTACs), antibody conjugates that are capable of selectively depleting immune checkpoint receptors such as SIRPa from the surface of TAMs. These chimeric molecules consist of a synthetic glycan ligand that binds the C-type lectin CD206, a well-established TAM marker, conjugated to a non-blocking antibody that binds but does not inhibit the checkpoint receptor. By engaging CD206, which constitutively recycles between the plasma membrane and early endosomes, TICTACs facilitate robust removal of the checkpoint receptors from the surface of CD206high macrophages, while having no effect on CD206low macrophages. By decoupling antibody selectivity from its blocking function, we present a new paradigm for developing highly tumor-specific immunotherapies.
Collapse
Affiliation(s)
- Mariko Morimoto
- Department of Chemistry and Sarafan ChEM-H, Stanford University, Stanford CA, USA
| | - Nicholas A Till
- Department of Chemistry and Sarafan ChEM-H, Stanford University, Stanford CA, USA
| | - Carolyn R Bertozzi
- Department of Chemistry and Sarafan ChEM-H, Stanford University, Stanford CA, USA
- Howard Hughes Medical Institute, Stanford University, Stanford CA, USA
| |
Collapse
|
4
|
Savchenko IV, Zlotnikov ID, Kudryashova EV. Biomimetic Systems Involving Macrophages and Their Potential for Targeted Drug Delivery. Biomimetics (Basel) 2023; 8:543. [PMID: 37999184 PMCID: PMC10669405 DOI: 10.3390/biomimetics8070543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 09/10/2023] [Accepted: 11/10/2023] [Indexed: 11/25/2023] Open
Abstract
The concept of targeted drug delivery can be described in terms of the drug systems' ability to mimic the biological objects' property to localize to target cells or tissues. For example, drug delivery systems based on red blood cells or mimicking some of their useful features, such as long circulation in stealth mode, have been known for decades. On the contrary, therapeutic strategies based on macrophages have gained very limited attention until recently. Here, we review two biomimetic strategies associated with macrophages that can be used to develop new therapeutic modalities: first, the mimicry of certain types of macrophages (i.e., the use of macrophages, including tumor-associated or macrophage-derived particles as a carrier for the targeted delivery of therapeutic agents); second, the mimicry of ligands, naturally absorbed by macrophages (i.e., the use of therapeutic agents specifically targeted at macrophages). We discuss the potential applications of biomimetic systems involving macrophages for new advancements in the treatment of infections, inflammatory diseases, and cancer.
Collapse
Affiliation(s)
| | | | - Elena V. Kudryashova
- Faculty of Chemistry, Lomonosov Moscow State University, Leninskie Gory, 1/3, 119991 Moscow, Russia (I.D.Z.)
| |
Collapse
|
5
|
Jin H, Liu X, Liu HX. Biological function, regulatory mechanism, and clinical application of mannose in cancer. Biochim Biophys Acta Rev Cancer 2023; 1878:188970. [PMID: 37657682 DOI: 10.1016/j.bbcan.2023.188970] [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: 04/19/2023] [Revised: 08/15/2023] [Accepted: 08/15/2023] [Indexed: 09/03/2023]
Abstract
Studies examining the regulatory roles and clinical applications of monosaccharides other than glucose in cancer have been neglected. Mannose, a common type of monosaccharide found in human body fluids and tissues, primarily functions in protein glycosylation rather than carbohydrate metabolism. Recent research has demonstrated direct anticancer effects of mannose in vitro and in vivo. Simply supplementing cell culture medium or drinking water with mannose achieved these effects. Moreover, mannose enhances the effectiveness of current cancer treatments including chemotherapy, radiotherapy, targeted therapy, and immune therapy. Besides the advancements in basic research on the anticancer effects of mannose, recent studies have reported its application as a biomarker for cancer or in the delivery of anticancer drugs using mannose-modified drug delivery systems. This review discusses the progress made in understanding the regulatory roles of mannose in cancer progression, the mechanisms underlying its anticancer effects, and its current application in cancer diagnosis and treatment.
Collapse
Affiliation(s)
- Haoyi Jin
- Department of Thoracic Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, 110042, Liaoning, China
| | - Xi Liu
- Department of Urology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, 110042, Liaoning, China
| | - Hong-Xu Liu
- Department of Thoracic Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, 110042, Liaoning, China; Department of Urology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, 110042, Liaoning, China.
| |
Collapse
|
6
|
Yoon J, Le XT, Kim J, Lee H, Nguyen NT, Lee WT, Lee ES, Oh KT, Choi HG, Youn YS. Macrophage-reprogramming upconverting nanoparticles for enhanced TAM-mediated antitumor therapy of hypoxic breast cancer. J Control Release 2023; 360:482-495. [PMID: 37423526 DOI: 10.1016/j.jconrel.2023.07.009] [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: 02/26/2023] [Revised: 06/10/2023] [Accepted: 07/06/2023] [Indexed: 07/11/2023]
Abstract
In an attempt to achieve antitumor effects by switching the phenotype of macrophages from the tumor-promoting M2 type to the tumor-suppressing M1 type, we fabricated mannose-decorated/macrophage membrane-coated, silica-layered NaErF4@NaLuF4 upconverting nanoparticles (UCNPs) co-doped with perfluorocarbon (PFC)/chlorin e6 (Ce6) and loaded with paclitaxel (PTX) (UCNP@mSiO2-PFC/Ce6@RAW-Man/PTX: ∼61 nm; -11.6 mV). These nanoparticles were designed to have two major functionalities, (i) efficient singlet oxygen generation aided by an oxygen supply and (ii) good targeting to tumor-associated macrophage (TAMs) (M2-type), to induce polarization to M1 type macrophages that release proinflammatory cytokines and suppress breast cancers. The primary UCNPs consisted of lanthanide elements (erbium and lutetium) in a core@shell structure, and they facilely emitted 660 nm light in response to a deep-penetrating 808 nm near-infrared laser. Moreover, the UCNPs@mSiO2-PFC/Ce6@RAW-Man/PTX were able to release O2 and generate 1O2 because of the co-doped PFC/Ce6 and upconversion. Our nanocarriers' excellent uptake to RAW 264.7 macrophage cells (M2 type) and efficient M1-type polarization activity were clearly demonstrated using qRT-PCR and immunofluorescence-based confocal laser scanning microscopy. Our nanocarriers displayed significant cytotoxicity to 4T1 cells in 2D culture and 3D co-culture systems of 4T1/RAW 264.7 cells. More importantly, UCNPs@mSiO2-PFC/Ce6@RAW-Man/PTX (+808 nm laser) noticeably suppressed tumor growth in 4T1-xenografted mice, compared with the other treatment groups (332.4 vs. 709.5-1185.5 mm3). We attribute this antitumor efficacy to the prominent M1-type macrophage polarization caused by our nanocarriers through efficient ROS/O2 generation and targeting of M2-type TAMs via mannose ligands on coated macrophage-membrane.
Collapse
Affiliation(s)
- Johyun Yoon
- School of Pharmacy, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do 16419, Republic of Korea
| | - Xuan Thien Le
- School of Pharmacy, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do 16419, Republic of Korea
| | - Juho Kim
- School of Pharmacy, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do 16419, Republic of Korea
| | - Hyunjun Lee
- School of Pharmacy, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do 16419, Republic of Korea
| | - Nguyen Thi Nguyen
- School of Pharmacy, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do 16419, Republic of Korea
| | - Woo Tak Lee
- School of Pharmacy, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do 16419, Republic of Korea
| | - Eun Seong Lee
- Department of Biotechnology and Department of Biomedical-Chemical Engineering, Catholic University of Korea, 43 Jibong-ro, Bucheon-si, Gyeonggi-do 14662, Republic of Korea
| | - Kyung Taek Oh
- College of Pharmacy, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul 06974, Republic of Korea
| | - Han-Gon Choi
- College of Pharmacy, Hanyang University, 55, Hanyangdaehak-ro, Sangnok-gu, Ansan 15588, Republic of Korea
| | - Yu Seok Youn
- School of Pharmacy, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do 16419, Republic of Korea.
| |
Collapse
|
7
|
Ahmadi M, Emzhik M, Mosayebnia M. Nanoparticles labeled with gamma-emitting radioisotopes: an attractive approach for in vivo tracking using SPECT imaging. Drug Deliv Transl Res 2023; 13:1546-1583. [PMID: 36811810 DOI: 10.1007/s13346-023-01291-1] [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] [Accepted: 01/03/2023] [Indexed: 02/24/2023]
Abstract
Providing accurate molecular imaging of the body and biological process is critical for diagnosing disease and personalizing treatment with the minimum side effects. Recently, diagnostic radiopharmaceuticals have gained more attention in precise molecular imaging due to their high sensitivity and appropriate tissue penetration depth. The fate of these radiopharmaceuticals throughout the body can be traced using nuclear imaging systems, including single-photon emission computed tomography (SPECT) and positron emission tomography (PET) modalities. In this regard, nanoparticles are attractive platforms for delivering radionuclides into targets because they can directly interfere with the cell membranes and subcellular organelles. Moreover, applying radiolabeled nanomaterials can decrease their toxicity concerns because radiopharmaceuticals are usually administrated at low doses. Therefore, incorporating gamma-emitting radionuclides into nanomaterials can provide imaging probes with valuable additional properties compared to the other carriers. Herein, we aim to review (1) the gamma-emitting radionuclides used for labeling different nanomaterials, (2) the approaches and conditions adopted for their radiolabeling, and (3) their application. This study can help researchers to compare different radiolabeling methods in terms of stability and efficiency and choose the best way for each nanosystem.
Collapse
Affiliation(s)
- Mahnaz Ahmadi
- Department of Pharmaceutics and Pharmaceutical Nanotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Marjan Emzhik
- Department of Pharmaceutics and Pharmaceutical Nanotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mona Mosayebnia
- Department of Pharmaceutical Chemistry and Radiopharmacy, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Niayesh Junction, Vali-E-Asr Ave, Tehran, 14155-6153, Iran.
| |
Collapse
|
8
|
Xia W, Singh N, Goel S, Shi S. Molecular Imaging of Innate Immunity and Immunotherapy. Adv Drug Deliv Rev 2023; 198:114865. [PMID: 37182699 DOI: 10.1016/j.addr.2023.114865] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 04/17/2023] [Accepted: 05/03/2023] [Indexed: 05/16/2023]
Abstract
The innate immune system plays a key role as the first line of defense in various human diseases including cancer, cardiovascular and inflammatory diseases. In contrast to tissue biopsies and blood biopsies, in vivo imaging of the innate immune system can provide whole body measurements of immune cell location and function and changes in response to disease progression and therapy. Rationally developed molecular imaging strategies can be used in evaluating the status and spatio-temporal distributions of the innate immune cells in near real-time, mapping the biodistribution of novel innate immunotherapies, monitoring their efficacy and potential toxicities, and eventually for stratifying patients that are likely to benefit from these immunotherapies. In this review, we will highlight the current state-of-the-art in noninvasive imaging techniques for preclinical imaging of the innate immune system particularly focusing on cell trafficking, biodistribution, as well as pharmacokinetics and dynamics of promising immunotherapies in cancer and other diseases; discuss the unmet needs and current challenges in integrating imaging modalities and immunology and suggest potential solutions to overcome these barriers.
Collapse
Affiliation(s)
- Wenxi Xia
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT 84112, United States
| | - Neetu Singh
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT 84112, United States
| | - Shreya Goel
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT 84112, United States; Department of Biomedical Engineering, University of Utah, Salt Lake City, UT 84112, United States; Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, UT 84112, United States
| | - Sixiang Shi
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT 84112, United States; Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, UT 84112, United States.
| |
Collapse
|
9
|
Low HY, Yang CT, Xia B, He T, Lam WWC, Ng DCE. Radiolabeled Liposomes for Nuclear Imaging Probes. Molecules 2023; 28:molecules28093798. [PMID: 37175207 PMCID: PMC10180453 DOI: 10.3390/molecules28093798] [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: 03/17/2023] [Revised: 04/17/2023] [Accepted: 04/26/2023] [Indexed: 05/15/2023] Open
Abstract
Quantitative nuclear imaging techniques are in high demand for various disease diagnostics and cancer theranostics. The non-invasive imaging modality requires radiotracing through the radioactive decay emission of the radionuclide. Current preclinical and clinical radiotracers, so-called nuclear imaging probes, are radioisotope-labeled small molecules. Liposomal radiotracers have been rapidly developing as novel nuclear imaging probes. The physicochemical properties and structural characteristics of liposomes have been elucidated to address their long circulation and stability as radiopharmaceuticals. Various radiolabeling methods for synthesizing radionuclides onto liposomes and synthesis strategies have been summarized to render them biocompatible and enable specific targeting. Through a variety of radionuclide labeling methods, radiolabeled liposomes for use as nuclear imaging probes can be obtained for in vivo biodistribution and specific targeting studies. The advantages of radiolabeled liposomes including their use as potential clinical nuclear imaging probes have been highlighted. This review is a comprehensive overview of all recently published liposomal SPECT and PET imaging probes.
Collapse
Affiliation(s)
- Ho Ying Low
- Department of Nuclear Medicine and Molecular Imaging, Radiological Sciences Division, Singapore General Hospital, Outram Road, Singapore 169608, Singapore
| | - Chang-Tong Yang
- Department of Nuclear Medicine and Molecular Imaging, Radiological Sciences Division, Singapore General Hospital, Outram Road, Singapore 169608, Singapore
- Duke-NUS Medical School, 8 College Road, Singapore 169857, Singapore
| | - Bin Xia
- School of Chemistry and Chemical Engineering, Hefei University of Technology, Hefei 230009, China
| | - Tao He
- School of Chemistry and Chemical Engineering, Hefei University of Technology, Hefei 230009, China
| | - Winnie Wing Chuen Lam
- Department of Nuclear Medicine and Molecular Imaging, Radiological Sciences Division, Singapore General Hospital, Outram Road, Singapore 169608, Singapore
- Duke-NUS Medical School, 8 College Road, Singapore 169857, Singapore
| | - David Chee Eng Ng
- Department of Nuclear Medicine and Molecular Imaging, Radiological Sciences Division, Singapore General Hospital, Outram Road, Singapore 169608, Singapore
- Duke-NUS Medical School, 8 College Road, Singapore 169857, Singapore
| |
Collapse
|
10
|
Kimura A, Utsumi S, Shimokawa A, Nishimori R, Hosoi R, Stewart NJ, Imai H, Fujiwara H. Targeted Imaging of Lung Cancer with Hyperpolarized 129Xe MRI Using Surface-Modified Iron Oxide Nanoparticles as Molecular Contrast Agents. Cancers (Basel) 2022; 14:cancers14246070. [PMID: 36551556 PMCID: PMC9776850 DOI: 10.3390/cancers14246070] [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: 11/15/2022] [Revised: 12/07/2022] [Accepted: 12/07/2022] [Indexed: 12/14/2022] Open
Abstract
Hyperpolarized 129Xe (HP 129Xe) MRI enables functional imaging of various lung diseases but has been scarcely applied to lung cancer imaging. The aim of this study is to investigate the feasibility of targeted imaging of lung cancer with HP 129Xe MRI using surface-modified iron oxide nanoparticles (IONPs) as molecular targeting contrast agents. A mouse model of lung cancer (LC) was induced in nine mice by intra-peritoneal injection of urethane. Three months after the urethane administration, the mice underwent lung imaging with HP 129Xe MRI at baseline (0 h). Subsequently, the LC group was divided into two sub-groups: mice administered with polyethylene glycol-coated IONPs (PEG-IONPs, n = 4) and folate-conjugated dextran-coated IONPs (FA@Dex-IONPs, n = 5). The mice were imaged at 3, 6, and 24 h after the intravenous injection of IONPs. FA@Dex-IONPs mice showed a 25% reduction in average signal intensity at cancer sites at 3 h post injection, and a 24% reduction at 24 h post injection. On the other hand, in PEG-IONPs mice, while a signal reduction of approximately 28% was observed at cancer sites at 3 to 6 h post injection, the signal intensity was unchanged from that of the baseline at 24 h. Proton MRI of LC mice (n = 3) was able to detect cancer five months after urethane administration, i.e., later than HP 129Xe MRI (3 months). Furthermore, a significant decrease in averaged 1H T2 values at cancer sites was observed at only 6 h post injection of FA@Dex-IONPs (p < 0.05). As such, the targeted delivery of IONPs to cancer tissue was successfully imaged with HP 129Xe MRI, and their surface modification with folate likely has a high affinity with LC, which causes overexpression of folate receptors.
Collapse
Affiliation(s)
- Atsuomi Kimura
- Department of Medical Physics and Engineering, Area of Medical Imaging Technology and Science, Division of Health Sciences, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
- Correspondence: ; Tel.: +81-6-6879-2578
| | - Seiya Utsumi
- Department of Medical Physics and Engineering, Area of Medical Imaging Technology and Science, Division of Health Sciences, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
| | - Akihiro Shimokawa
- Department of Medical Physics and Engineering, Area of Medical Imaging Technology and Science, Division of Health Sciences, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
| | - Renya Nishimori
- Department of Medical Physics and Engineering, Area of Medical Imaging Technology and Science, Division of Health Sciences, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
| | - Rie Hosoi
- Department of Medical Physics and Engineering, Area of Medical Imaging Technology and Science, Division of Health Sciences, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
| | - Neil J. Stewart
- POLARIS, Imaging Sciences, Department of Infection, Immunity & Cardiovascular Disease, University of Sheffield, Sheffield S10 2TA, UK
| | - Hirohiko Imai
- Division of Systems Informatics, Department of Systems Science, Graduate School of Informatics, Kyoto University, Kyoto 606-8561, Japan
| | - Hideaki Fujiwara
- Department of Medical Physics and Engineering, Area of Medical Imaging Technology and Science, Division of Health Sciences, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
| |
Collapse
|
11
|
Nguyen A, Kumar S, Kulkarni AA. Nanotheranostic Strategies for Cancer Immunotherapy. SMALL METHODS 2022; 6:e2200718. [PMID: 36382571 PMCID: PMC11056828 DOI: 10.1002/smtd.202200718] [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: 06/02/2022] [Revised: 09/13/2022] [Indexed: 06/16/2023]
Abstract
Despite advancements in cancer immunotherapy, heterogeneity in tumor response impose barriers to successful treatments and accurate prognosis. Effective therapy and early outcome detection are critical as toxicity profiles following immunotherapies can severely affect patients' quality of life. Existing imaging techniques, including positron emission tomography, computed tomography, magnetic resonance imaging, or multiplexed imaging, are often used in clinics yet suffer from limitations in the early assessment of immune response. Conventional strategies to validate immune response mainly rely on the Response Evaluation Criteria in Solid Tumors (RECIST) and the modified iRECIST for immuno-oncology drug trials. However, accurate monitoring of immunotherapy efficacy is challenging since the response does not always follow conventional RECIST criteria due to delayed and variable kinetics in immunotherapy responses. Engineered nanomaterials for immunotherapy applications have significantly contributed to overcoming these challenges by improving drug delivery and dynamic imaging techniques. This review summarizes challenges in recent immune-modulation approaches and traditional imaging tools, followed by emerging developments in three-in-one nanoimmunotheranostic systems co-opting nanotechnology, immunotherapy, and imaging. In addition, a comprehensive overview of imaging modalities in recent cancer immunotherapy research and a brief outlook on how nanotheranostic platforms can potentially advance to clinical translations for the field of immuno-oncology is presented.
Collapse
Affiliation(s)
- Anh Nguyen
- Department of Chemical Engineering, University of Massachusetts, Amherst, MA, USA
| | - Sahana Kumar
- Department of Chemical Engineering, University of Massachusetts, Amherst, MA, USA
| | - Ashish A. Kulkarni
- Department of Chemical Engineering, University of Massachusetts, Amherst, MA, USA
- Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA, USA
| |
Collapse
|
12
|
Chung H, Park JY, Kim K, Yoo RJ, Suh M, Gu GJ, Kim JS, Choi TH, Byun JW, Ju YW, Han W, Ryu HS, Chung G, Hwang DW, Kim Y, Kang HR, Na YR, Choi H, Im HJ, Lee YS, Seok SH. Circulation Time-Optimized Albumin Nanoplatform for Quantitative Visualization of Lung Metastasis via Targeting of Macrophages. ACS NANO 2022; 16:12262-12275. [PMID: 35943956 PMCID: PMC9413422 DOI: 10.1021/acsnano.2c03075] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
The development of molecular imaging probes to identify key cellular changes within lung metastases may lead to noninvasive detection of metastatic lesions in the lung. In this study, we constructed a macrophage-targeted clickable albumin nanoplatform (CAN) decorated with mannose as the targeting ligand using a click reaction to maintain the intrinsic properties of albumin in vivo. We also modified the number of mannose molecules on the CAN and found that mannosylated serum albumin (MSA) harboring six molecules of mannose displayed favorable pharmacokinetics that allowed high-contrast imaging of the lung, rendering it suitable for in vivo visualization of lung metastases. Due to the optimized control of functionalization and surface modification, MSA enhanced blood circulation time and active/passive targeting abilities and was specifically incorporated by mannose receptor (CD206)-expressing macrophages in the metastatic lung. Moreover, extensive in vivo imaging studies using single-photon emission computed tomography (SPECT)/CT and positron emission tomography (PET) revealed that blood circulation of time-optimized MSA can be used to discern metastatic lesions, with a strong correlation between its signal and metastatic burden in the lung.
Collapse
Affiliation(s)
- Hyewon Chung
- Macrophage
Lab, Department of Microbiology and Immunology, and Institute of Endemic
Disease, Seoul National University College
of Medicine, Seoul 03080, Republic of Korea
- Bio-MAX
Institute, Seoul National University, Seoul 03080, Republic
of Korea
| | - Ji Yong Park
- Department
of Biomedical Sciences, Seoul National University
College of Medicine, Seoul 03080, Republic of Korea
- Department
of Nuclear Medicine, Seoul National University
Hospital, Seoul 03080, Republic of Korea
- Cancer
Research Institute, Seoul National University, Seoul 03080, Republic of Korea
- Dental
Research Institute, Seoul National University, Seoul 03080, Republic of Korea
| | - Kyuwan Kim
- Department
of Nuclear Medicine, Seoul National University
Hospital, Seoul 03080, Republic of Korea
- Cancer
Research Institute, Seoul National University, Seoul 03080, Republic of Korea
| | - Ran Ji Yoo
- Department
of Nuclear Medicine, Seoul National University
Hospital, Seoul 03080, Republic of Korea
- Cancer
Research Institute, Seoul National University, Seoul 03080, Republic of Korea
| | - Minseok Suh
- Department
of Molecular Medicine and Biopharmaceutical Sciences, Graduate School
of Convergence Science and Technology, Seoul
National University, Seoul 03080, Republic of Korea
| | - Gyo Jeong Gu
- Macrophage
Lab, Department of Microbiology and Immunology, and Institute of Endemic
Disease, Seoul National University College
of Medicine, Seoul 03080, Republic of Korea
| | - Jin Sil Kim
- Department
of Nuclear Medicine, Seoul National University
Hospital, Seoul 03080, Republic of Korea
| | - Tae Hyeon Choi
- Department
of Nuclear Medicine, Seoul National University
Hospital, Seoul 03080, Republic of Korea
- Department
of Molecular Medicine and Biopharmaceutical Sciences, Graduate School
of Convergence Science and Technology, Seoul
National University, Seoul 03080, Republic of Korea
| | - Jung Woo Byun
- Department
of Nuclear Medicine, Seoul National University
Hospital, Seoul 03080, Republic of Korea
| | - Young Wook Ju
- Department
of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul 03080, Republic
of Korea
| | - Wonshik Han
- Department
of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul 03080, Republic
of Korea
| | - Han Suk Ryu
- Department
of Pathology, Seoul National University
College of Medicine, Seoul 03080, Republic of Korea
| | - Gehoon Chung
- Dental
Research Institute, Seoul National University, Seoul 03080, Republic of Korea
- Department
of Oral Physiology, Seoul National University,
School of Dentistry, Seoul 03080, Republic of Korea
| | - Do Won Hwang
- Department
of Nuclear Medicine, Seoul National University
Hospital, Seoul 03080, Republic of Korea
- Research and Development Center, THERABEST,
Co. Ltd., Seoul 03080, Republic of Korea
| | - Yujin Kim
- Department
of Biomedical Sciences, Seoul National University
College of Medicine, Seoul 03080, Republic of Korea
| | - Hye-Ryun Kang
- Department
of Biomedical Sciences, Seoul National University
College of Medicine, Seoul 03080, Republic of Korea
| | - Yi Rang Na
- Transdisciplinary Department of Medicine
and Advanced Technology, Seoul National
University Hospital, Seoul 03080, Republic of Korea
| | - Hongyoon Choi
- Department
of Nuclear Medicine, Seoul National University
Hospital, Seoul 03080, Republic of Korea
| | - Hyung-Jun Im
- Department
of Molecular Medicine and Biopharmaceutical Sciences, Graduate School
of Convergence Science and Technology, Seoul
National University, Seoul 03080, Republic of Korea
- Research Institute for Convergence Science, Seoul National University, Seoul 08823, Republic of Korea
| | - Yun-Sang Lee
- Department
of Biomedical Sciences, Seoul National University
College of Medicine, Seoul 03080, Republic of Korea
- Department
of Nuclear Medicine, Seoul National University
Hospital, Seoul 03080, Republic of Korea
- Cancer
Research Institute, Seoul National University, Seoul 03080, Republic of Korea
| | - Seung Hyeok Seok
- Macrophage
Lab, Department of Microbiology and Immunology, and Institute of Endemic
Disease, Seoul National University College
of Medicine, Seoul 03080, Republic of Korea
- Department
of Biomedical Sciences, Seoul National University
College of Medicine, Seoul 03080, Republic of Korea
| |
Collapse
|
13
|
van der Heide CD, Dalm SU. Radionuclide imaging and therapy directed towards the tumor microenvironment: a multi-cancer approach for personalized medicine. Eur J Nucl Med Mol Imaging 2022; 49:4616-4641. [PMID: 35788730 PMCID: PMC9606105 DOI: 10.1007/s00259-022-05870-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 06/09/2022] [Indexed: 12/19/2022]
Abstract
Targeted radionuclide theranostics is becoming more and more prominent in clinical oncology. Currently, most nuclear medicine compounds researched for cancer theranostics are directed towards targets expressed in only a small subset of cancer types, limiting clinical applicability. The identification of cancer-specific targets that are (more) universally expressed will allow more cancer patients to benefit from these personalized nuclear medicine–based interventions. A tumor is not merely a collection of cancer cells, it also comprises supporting stromal cells embedded in an altered extracellular matrix (ECM), together forming the tumor microenvironment (TME). Since the TME is less genetically unstable than cancer cells, and TME phenotypes can be shared between cancer types, it offers targets that are more universally expressed. The TME is characterized by the presence of altered processes such as hypoxia, acidity, and increased metabolism. Next to the ECM, the TME consists of cancer-associated fibroblasts (CAFs), macrophages, endothelial cells forming the neo-vasculature, immune cells, and cancer-associated adipocytes (CAAs). Radioligands directed at the altered processes, the ECM, and the cellular components of the TME have been developed and evaluated in preclinical and clinical studies for targeted radionuclide imaging and/or therapy. In this review, we provide an overview of the TME targets and their corresponding radioligands. In addition, we discuss what developments are needed to further explore the TME as a target for radionuclide theranostics, with the hopes of stimulating the development of novel TME radioligands with multi-cancer, or in some cases even pan-cancer, application.
Collapse
Affiliation(s)
| | - Simone U Dalm
- Department of Radiology & Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands.
| |
Collapse
|
14
|
van der Geest KSM, Sandovici M, Nienhuis PH, Slart RHJA, Heeringa P, Brouwer E, Jiemy WF. Novel PET Imaging of Inflammatory Targets and Cells for the Diagnosis and Monitoring of Giant Cell Arteritis and Polymyalgia Rheumatica. Front Med (Lausanne) 2022; 9:902155. [PMID: 35733858 PMCID: PMC9207253 DOI: 10.3389/fmed.2022.902155] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 05/13/2022] [Indexed: 12/26/2022] Open
Abstract
Giant cell arteritis (GCA) and polymyalgia rheumatica (PMR) are two interrelated inflammatory diseases affecting patients above 50 years of age. Patients with GCA suffer from granulomatous inflammation of medium- to large-sized arteries. This inflammation can lead to severe ischemic complications (e.g., irreversible vision loss and stroke) and aneurysm-related complications (such as aortic dissection). On the other hand, patients suffering from PMR present with proximal stiffness and pain due to inflammation of the shoulder and pelvic girdles. PMR is observed in 40-60% of patients with GCA, while up to 21% of patients suffering from PMR are also affected by GCA. Due to the risk of ischemic complications, GCA has to be promptly treated upon clinical suspicion. The treatment of both GCA and PMR still heavily relies on glucocorticoids (GCs), although novel targeted therapies are emerging. Imaging has a central position in the diagnosis of GCA and PMR. While [18F]fluorodeoxyglucose (FDG)-positron emission tomography (PET) has proven to be a valuable tool for diagnosis of GCA and PMR, it possesses major drawbacks such as unspecific uptake in cells with high glucose metabolism, high background activity in several non-target organs and a decrease of diagnostic accuracy already after a short course of GC treatment. In recent years, our understanding of the immunopathogenesis of GCA and, to some extent, PMR has advanced. In this review, we summarize the current knowledge on the cellular heterogeneity in the immunopathology of GCA/PMR and discuss how recent advances in specific tissue infiltrating leukocyte and stromal cell profiles may be exploited as a source of novel targets for imaging. Finally, we discuss prospective novel PET radiotracers that may be useful for the diagnosis and treatment monitoring in GCA and PMR.
Collapse
Affiliation(s)
- Kornelis S. M. van der Geest
- Department of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Maria Sandovici
- Department of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Pieter H. Nienhuis
- Department of Nuclear Medicine and Molecular Imaging, Medical Imaging Center, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Riemer H. J. A. Slart
- Department of Nuclear Medicine and Molecular Imaging, Medical Imaging Center, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
- Department of Biomedical Photonic Imaging Group, University of Twente, Enschede, Netherlands
| | - Peter Heeringa
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Elisabeth Brouwer
- Department of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - William F. Jiemy
- Department of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| |
Collapse
|
15
|
Tuguntaev RG, Hussain A, Fu C, Chen H, Tao Y, Huang Y, Liu L, Liang XJ, Guo W. Bioimaging guided pharmaceutical evaluations of nanomedicines for clinical translations. J Nanobiotechnology 2022; 20:236. [PMID: 35590412 PMCID: PMC9118863 DOI: 10.1186/s12951-022-01451-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 05/05/2022] [Indexed: 11/25/2022] Open
Abstract
Nanomedicines (NMs) have emerged as an efficient approach for developing novel treatment strategies against a variety of diseases. Over the past few decades, NM formulations have received great attention, and a large number of studies have been performed in this field. Despite this, only about 60 nano-formulations have received industrial acceptance and are currently available for clinical use. Their in vivo pharmaceutical behavior is considered one of the main challenges and hurdles for the effective clinical translation of NMs, because it is difficult to monitor the pharmaceutic fate of NMs in the biological environment using conventional pharmaceutical evaluations. In this context, non-invasive imaging modalities offer attractive solutions, providing the direct monitoring and quantification of the pharmacokinetic and pharmacodynamic behavior of labeled NMs in a real-time manner. Imaging evaluations have great potential for revealing the relationship between the physicochemical properties of NMs and their pharmaceutical profiles in living subjects. In this review, we introduced imaging techniques that can be used for in vivo NM evaluations. We also provided an overview of various studies on the influence of key parameters on the in vivo pharmaceutical behavior of NMs that had been visualized in a non-invasive and real-time manner.
Collapse
Affiliation(s)
- Ruslan G Tuguntaev
- Department of Minimally Invasive Interventional Radiology, Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences & the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510260, People's Republic of China
| | - Abid Hussain
- Advanced Research Institute of Multidisciplinary Science, School of Life Science, School of Medical Technology (Institute of Engineering Medicine), Key Laboratory of Molecular Medicine and Biotherapy, Key Laboratory of Medical Molecular Science and Pharmaceutics Engineering, Beijing Institute of Technology, Beijing, 100081, China
| | - Chenxing Fu
- Department of Minimally Invasive Interventional Radiology, Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences & the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510260, People's Republic of China
| | - Haoting Chen
- Department of Minimally Invasive Interventional Radiology, Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences & the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510260, People's Republic of China
| | - Ying Tao
- Department of Minimally Invasive Interventional Radiology, Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences & the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510260, People's Republic of China
| | - Yan Huang
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Nantong University, Nantong, 226001, China
| | - Lu Liu
- Chinese Academy of Sciences (CAS) Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, 100190, People's Republic of China.
| | - Xing-Jie Liang
- Chinese Academy of Sciences (CAS) Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, 100190, People's Republic of China.
| | - Weisheng Guo
- Department of Minimally Invasive Interventional Radiology, Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences & the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510260, People's Republic of China.
| |
Collapse
|
16
|
Padmakumar A, Koyande NP, Rengan AK. The Role of Hitchhiking in Cancer Therapeutics – A review. ADVANCED THERAPEUTICS 2022. [DOI: 10.1002/adtp.202200042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Affiliation(s)
- Ananya Padmakumar
- Department of Biomedical Engineering Indian Institute of Technology Hyderabad Sangareddy 502284 India
| | - Navami Prabhakar Koyande
- Department of Biomedical Engineering Indian Institute of Technology Hyderabad Sangareddy 502284 India
| | - Aravind Kumar Rengan
- Department of Biomedical Engineering Indian Institute of Technology Hyderabad Sangareddy 502284 India
| |
Collapse
|
17
|
Cui R, Wang L, Zhang D, Zhang K, Dou J, Dong L, Zhang Y, Wu J, Tan L, Yu J, Liang P. Combination therapy using microwave ablation and D-mannose-chelated iron oxide nanoparticles inhibits hepatocellular carcinoma progression. Acta Pharm Sin B 2022; 12:3475-3485. [PMID: 36176908 PMCID: PMC9513490 DOI: 10.1016/j.apsb.2022.05.026] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Revised: 05/02/2022] [Accepted: 05/11/2022] [Indexed: 12/16/2022] Open
Abstract
Despite being a common therapy for hepatocellular carcinoma (HCC), insufficient thermal ablation can leave behind tumor residues that can cause recurrence. This is believed to augment M2 inflammatory macrophages that usually play a pro-tumorigenic role. To address this problem, we designed d-mannose-chelated iron oxide nanoparticles (man-IONPs) to polarize M2-like macrophages into the antitumor M1 phenotype. In vitro and in vivo experiments demonstrated that man-IONPs specifically targeted M2-like macrophages and accumulated in peri-ablation zones after macrophage infiltration was augmented under insufficient microwave ablation (MWA). The nanoparticles simultaneously induced polarization of pro-tumorigenic M2 macrophages into antitumor M1 phenotypes, enabling the transformation of the immunosuppressive microenvironment into an immunoactivating one. Post-MWA macrophage polarization exerted robust inhibitory effects on HCC progression in a well-established orthotopic liver cancer mouse model. Thus, combining thermal ablation with man-IONPs can salvage residual tumors after insufficient MWA. These results have strong potential for clinical translation.
Collapse
|
18
|
Hegi-Johnson F, Rudd S, Hicks RJ, De Ruysscher D, Trapani JA, John T, Donnelly P, Blyth B, Hanna G, Everitt S, Roselt P, MacManus MP. Imaging immunity in patients with cancer using positron emission tomography. NPJ Precis Oncol 2022; 6:24. [PMID: 35393508 PMCID: PMC8989882 DOI: 10.1038/s41698-022-00263-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 02/24/2022] [Indexed: 12/26/2022] Open
Abstract
Immune checkpoint inhibitors and related molecules can achieve tumour regression, and even prolonged survival, for a subset of cancer patients with an otherwise dire prognosis. However, it remains unclear why some patients respond to immunotherapy and others do not. PET imaging has the potential to characterise the spatial and temporal heterogeneity of both immunotherapy target molecules and the tumor immune microenvironment, suggesting a tantalising vision of personally-adapted immunomodulatory treatment regimens. Personalised combinations of immunotherapy with local therapies and other systemic therapies, would be informed by immune imaging and subsequently modified in accordance with therapeutically induced immune environmental changes. An ideal PET imaging biomarker would facilitate the choice of initial therapy and would permit sequential imaging in time-frames that could provide actionable information to guide subsequent therapy. Such imaging should provide either prognostic or predictive measures of responsiveness relevant to key immunotherapy types but, most importantly, guide key decisions on initiation, continuation, change or cessation of treatment to reduce the cost and morbidity of treatment while enhancing survival outcomes. We survey the current literature, focusing on clinically relevant immune checkpoint immunotherapies, for which novel PET tracers are being developed, and discuss what steps are needed to make this vision a reality.
Collapse
Affiliation(s)
- Fiona Hegi-Johnson
- Department of Radiation Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- The Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
| | - Stacey Rudd
- Department of Chemistry, University of Melbourne, Melbourne, VIC, Australia
| | - Rodney J Hicks
- The Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
- Department of Cancer Imaging, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Dirk De Ruysscher
- Department of Radiation Oncology (Maastro), GROW School for Oncology, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Joseph A Trapani
- The Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Thomas John
- The Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Paul Donnelly
- Department of Chemistry, University of Melbourne, Melbourne, VIC, Australia
| | - Benjamin Blyth
- The Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
| | - Gerard Hanna
- Department of Radiation Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- The Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
| | - Sarah Everitt
- Department of Radiation Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- The Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
| | - Peter Roselt
- Department of Cancer Imaging, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Michael P MacManus
- Department of Radiation Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.
- The Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia.
| |
Collapse
|
19
|
Chen X, Niu W, Du Z, Zhang Y, Su D, Gao X. 64Cu radiolabeled nanomaterials for positron emission tomography (PET) imaging. CHINESE CHEM LETT 2022. [DOI: 10.1016/j.cclet.2022.02.070] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
|
20
|
Li X, Wang R, Zhang Y, Han S, Gan Y, Liang Q, Ma X, Rong P, Wang W, Li W. Molecular imaging of tumor-associated macrophages in cancer immunotherapy. Ther Adv Med Oncol 2022; 14:17588359221076194. [PMID: 35251314 PMCID: PMC8891912 DOI: 10.1177/17588359221076194] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 01/10/2022] [Indexed: 12/20/2022] Open
Abstract
Tumor-associated macrophages (TAMs), the most abundant inflammatory cell group in the tumor microenvironment, play an essential role in tumor immune regulation. The infiltration degree of TAMs in the tumor microenvironment is closely related to tumor growth and metastasis, and TAMs have become a promising target in tumor immunotherapy. Molecular imaging is a new interdisciplinary subject that combines medical imaging technology with molecular biology, nuclear medicine, radiation medicine, and computer science. The latest progress in molecular imaging allows the biological processes of cells to be visualized in vivo, which makes it possible to better understand the density and distribution of macrophages in the tumor microenvironment. This review mainly discusses the application of targeting TAM in tumor immunotherapy and the imaging characteristics and progress of targeting TAM molecular probes using various imaging techniques.
Collapse
Affiliation(s)
- Xiaoying Li
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, People’s Republic of China
- Cell Transplantation and Gene Therapy Institute, The Third Xiangya Hospital, Central South University, Changsha, People’s Republic of China
| | - Ruike Wang
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, People’s Republic of China
- Cell Transplantation and Gene Therapy Institute, The Third Xiangya Hospital, Central South University, Changsha, People’s Republic of China
| | - Yangnan Zhang
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, People’s Republic of China
- Cell Transplantation and Gene Therapy Institute, The Third Xiangya Hospital, Central South University, Changsha, People’s Republic of China
| | - Shuangze Han
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, People’s Republic of China
- Cell Transplantation and Gene Therapy Institute, The Third Xiangya Hospital, Central South University, Changsha, People’s Republic of China
| | - Yu Gan
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, People’s Republic of China
- Cell Transplantation and Gene Therapy Institute, The Third Xiangya Hospital, Central South University, Changsha, People’s Republic of China
| | - Qi Liang
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, People’s Republic of China
- Cell Transplantation and Gene Therapy Institute, The Third Xiangya Hospital, Central South University, Changsha, People’s Republic of China
| | - Xiaoqian Ma
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, People’s Republic of China
- Cell Transplantation and Gene Therapy Institute, The Third Xiangya Hospital, Central South University, Changsha, People’s Republic of China
| | - Pengfei Rong
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha 410013, Hunan, People’s Republic of China
- Cell Transplantation and Gene Therapy Institute, The Third Xiangya Hospital, Central South University, Changsha, People’s Republic of China
| | - Wei Wang
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha 410013, Hunan, People’s Republic of China
- Cell Transplantation and Gene Therapy Institute, The Third Xiangya Hospital, Central South University, Changsha, People’s Republic of China
| | - Wei Li
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha 410013, Hunan, People’s Republic of China
- Cell Transplantation and Gene Therapy Institute, The Third Xiangya Hospital, Central South University, Changsha, People’s Republic of China
| |
Collapse
|
21
|
Rehman MU, Khan A, Imtiyaz Z, Ali S, Makeen HA, Rashid S, Arafah A. Current Nano-therapeutic Approaches Ameliorating Inflammation in Cancer Progression. Semin Cancer Biol 2022; 86:886-908. [DOI: 10.1016/j.semcancer.2022.02.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Revised: 01/22/2022] [Accepted: 02/03/2022] [Indexed: 12/12/2022]
|
22
|
Song Y, Huang Y, Zhou F, Ding J, Zhou W. Macrophage-targeted nanomedicine for chronic diseases immunotherapy. CHINESE CHEM LETT 2022. [DOI: 10.1016/j.cclet.2021.08.090] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
23
|
Ravi Kiran AVVV, Kusuma Kumari G, Krishnamurthy PT, Khaydarov RR. Tumor microenvironment and nanotherapeutics: intruding the tumor fort. Biomater Sci 2021; 9:7667-7704. [PMID: 34673853 DOI: 10.1039/d1bm01127h] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Over recent years, advancements in nanomedicine have allowed new approaches to diagnose and treat tumors. Nano drug delivery systems exploit the enhanced permeability and retention (EPR) effect and enter the tumor tissue's interstitial space. However, tumor barriers play a crucial role, and cause inefficient EPR or the homing effect. Mounting evidence supports the hypothesis that the components of the tumor microenvironment, such as the extracellular matrix, and cellular and physiological components collectively or cooperatively hinder entry and distribution of drugs, and therefore, limit the theragnostic applications of cancer nanomedicine. This abnormal tumor microenvironment plays a pivotal role in cancer nanomedicine and was recently recognized as a promising target for improving nano-drug delivery and their therapeutic outcomes. Strategies like passive or active targeting, stimuli-triggered nanocarriers, and the modulation of immune components have shown promising results in achieving anticancer efficacy. The present review focuses on the tumor microenvironment and nanoparticle-based strategies (polymeric, inorganic and organic nanoparticles) for intruding the tumor barrier and improving therapeutic effects.
Collapse
Affiliation(s)
- Ammu V V V Ravi Kiran
- Department of Pharmacology, JSS College of Pharmacy (JSS Academy of Higher Education and Research), Ooty, Tamil Nadu, 643001, India
| | - Garikapati Kusuma Kumari
- Department of Pharmacology, JSS College of Pharmacy (JSS Academy of Higher Education and Research), Ooty, Tamil Nadu, 643001, India
| | - Praveen T Krishnamurthy
- Department of Pharmacology, JSS College of Pharmacy (JSS Academy of Higher Education and Research), Ooty, Tamil Nadu, 643001, India
| | - Renat R Khaydarov
- Institute of Nuclear Physics, Uzbekistan Academy of Sciences, Tashkent, 100047, Uzbekistan.
| |
Collapse
|
24
|
Functionally Heterogenous Macrophage Subsets in the Pathogenesis of Giant Cell Arteritis: Novel Targets for Disease Monitoring and Treatment. J Clin Med 2021; 10:jcm10214958. [PMID: 34768479 PMCID: PMC8585092 DOI: 10.3390/jcm10214958] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 10/20/2021] [Accepted: 10/23/2021] [Indexed: 12/19/2022] Open
Abstract
Giant cell arteritis (GCA) is a granulomatous large-vessel vasculitis that affects adults above 50 years of age. In GCA, circulating monocytes are recruited to the inflamed arteries. With cues from the vascular microenvironment, they differentiate into macrophages and play important roles in the pathogenesis of GCA via pro-inflammatory cytokine production and vascular remodeling. However, a deeper understanding of macrophage heterogeneity in GCA pathogenesis is needed to assist the development of novel diagnostic tools and targeted therapies. Here, we review the current knowledge on macrophage heterogeneity and diverse functions of macrophage subsets in the pathogenesis of GCA. We next discuss the possibility to exploit their heterogeneity as a source of novel biomarkers and as targets for nuclear imaging. Finally, we discuss novel macrophage-targeted therapies and future directions for targeting these cells in GCA.
Collapse
|
25
|
Cheng Y, Song S, Wu P, Lyu B, Qin M, Sun Y, Sun A, Mu L, Xu F, Zhang L, Wang J, Zhang Q. Tumor Associated Macrophages and TAMs-Based Anti-Tumor Nanomedicines. Adv Healthc Mater 2021; 10:e2100590. [PMID: 34292673 DOI: 10.1002/adhm.202100590] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 06/08/2021] [Indexed: 12/14/2022]
Abstract
As an important part of tumor microenvironment, tumor associated macrophages (TAMs) play a vital role in the occurrence, development, invasion, and metastasis of many malignant tumors and can significantly promote the formation of tumor blood vessels and lymphatic vessels, hence TAMs are greatly associated with poor prognosis. The research on nanomedicine has achieved huge progress, and nano-drugs have been widely utilized to treat various diseases through different mechanisms. Therefore, developing nano-drugs that are based on TAMs-associated anti-tumor mechanisms to effectively suppress tumor growth is expected to be a promising research filed. This paper introduces relevant information about TAMs in terms of their origin, and their roles in tumor genesis, development and metastasis. Furthermore, TAMs-related anti-tumor nano-drugs are summarized. Specifically, a wide range of nano-drugs targeting at TAMs are introduced, and categorized according to their therapeutic mechanisms toward tumors. Additionally, various nano delivery platforms using TAMs as cell carriers which aim at inhibiting tumor growth are reviewed. These two parts elucidate that the exploration of nanomedicine is essential to the study on TAMs-related anti-tumor strategies. This review is also intended to provide novel ideas for in-depth investigation on anti-tumor molecular mechanisms and nano-drug delivery systems based on TAMs.
Collapse
Affiliation(s)
- Yuxi Cheng
- State Key Laboratory of Natural and Biomimetic Drugs Peking University Beijing 100191 China
| | - Siyang Song
- State Key Laboratory of Natural and Biomimetic Drugs Peking University Beijing 100191 China
| | - Peiyao Wu
- State Key Laboratory of Natural and Biomimetic Drugs Peking University Beijing 100191 China
- School of Pharmacy Shenyang Pharmaceutical University Shenyang 110016 China
| | - Bochen Lyu
- State Key Laboratory of Natural and Biomimetic Drugs Peking University Beijing 100191 China
| | - Mengmeng Qin
- State Key Laboratory of Natural and Biomimetic Drugs Peking University Beijing 100191 China
| | - Yanan Sun
- State Key Laboratory of Natural and Biomimetic Drugs Peking University Beijing 100191 China
| | - Aning Sun
- State Key Laboratory of Natural and Biomimetic Drugs Peking University Beijing 100191 China
| | - Limin Mu
- State Key Laboratory of Natural and Biomimetic Drugs Peking University Beijing 100191 China
| | - Fei Xu
- State Key Laboratory of Natural and Biomimetic Drugs Peking University Beijing 100191 China
| | - Lu Zhang
- State Key Laboratory of Natural and Biomimetic Drugs Peking University Beijing 100191 China
| | - Jiancheng Wang
- State Key Laboratory of Natural and Biomimetic Drugs Peking University Beijing 100191 China
| | - Qiang Zhang
- State Key Laboratory of Natural and Biomimetic Drugs Peking University Beijing 100191 China
- School of Pharmacy Shenyang Pharmaceutical University Shenyang 110016 China
| |
Collapse
|
26
|
Nanoparticles to Target and Treat Macrophages: The Ockham's Concept? Pharmaceutics 2021; 13:pharmaceutics13091340. [PMID: 34575416 PMCID: PMC8469871 DOI: 10.3390/pharmaceutics13091340] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 08/15/2021] [Accepted: 08/19/2021] [Indexed: 12/19/2022] Open
Abstract
Nanoparticles are nanomaterials with three external nanoscale dimensions and an average size ranging from 1 to 1000 nm. Nanoparticles have gained notoriety in technological advances due to their tunable physical, chemical, and biological characteristics. However, the administration of functionalized nanoparticles to living beings is still challenging due to the rapid detection and blood and tissue clearance by the mononuclear phagocytic system. The major exponent of this system is the macrophage. Regardless the nanomaterial composition, macrophages can detect and incorporate foreign bodies by phagocytosis. Therefore, the simplest explanation is that any injected nanoparticle will be probably taken up by macrophages. This explains, in part, the natural accumulation of most nanoparticles in the spleen, lymph nodes, and liver (the main organs of the mononuclear phagocytic system). For this reason, recent investigations are devoted to design nanoparticles for specific macrophage targeting in diseased tissues. The aim of this review is to describe current strategies for the design of nanoparticles to target macrophages and to modulate their immunological function involved in different diseases with special emphasis on chronic inflammation, tissue regeneration, and cancer.
Collapse
|
27
|
Inflammation during Lung Cancer Progression and Ethyl Pyruvate Treatment Observed by Pulmonary Functional Hyperpolarized 129Xe MRI in Mice. CONTRAST MEDIA & MOLECULAR IMAGING 2021; 2021:9918702. [PMID: 34257627 PMCID: PMC8261185 DOI: 10.1155/2021/9918702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 06/08/2021] [Accepted: 06/17/2021] [Indexed: 11/17/2022]
Abstract
This study aimed to assess the suitability of hyperpolarized 129Xe (HPXe) MRI for noninvasive longitudinal evaluation of pulmonary function in preclinical lung cancer models. A mouse model of lung cancer (LC) was induced in 5 mice by intraperitoneal injection of urethane, while a negative-control (NC) mice (N = 5) was prepared by injection of saline solution. Longitudinal HPXe MRI was performed over a 5-month period to monitor lung ventilation and gas exchange. The treatment efficacy of ethyl pyruvate (EP), an anti-inflammatory drug, to the mouse LC model was monitored using HPXe MRI by commencing administration of EP pre (early-phase) and 1-month post (late-phase) injection of urethane (N = 5 mice for each group). Gas-exchange function in LC mice was significantly reduced at 1-month after urethane injection compared with NC mice administered with saline (P < 0.01). Thereafter, it remained consistently lower than that of the NC group for the full 5-month measurement period. In contrast, the ventilation function of the LC model mice was not significantly different to that of the NC mice. Histological analysis revealed alveolar epithelial hyperplasia in LC mice alveoli at 1 month after urethane injection, and adenoma was confirmed 3 months after the injection. The early- and late-phase EP interventions were found to improve HPXe MRI metrics (reduced at 1 month postinjection of urethane) and significantly inhibit tumor growth. These results suggest that HPXe MRI gas-exchange metrics can be used to quantitatively assess changes in the precancerous lesion microenvironment and to evaluate therapeutic efficacy in cancer. Thus, HPXe MRI can be utilized to noninvasively monitor pulmonary pathology during LC progression and can visualize functional changes during therapy.
Collapse
|
28
|
Luo X, Hu D, Gao D, Wang Y, Chen X, Liu X, Zheng H, Sun M, Sheng Z. Metabolizable Near-Infrared-II Nanoprobes for Dynamic Imaging of Deep-Seated Tumor-Associated Macrophages in Pancreatic Cancer. ACS NANO 2021; 15:10010-10024. [PMID: 34060821 DOI: 10.1021/acsnano.1c01608] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Tumor-associated macrophages (TAMs) play a crucial part in cancer evolution. Dynamic imaging of TAMs is of great significance for treatment outcome evaluation and precision tumor therapy. Currently, most fluorescence nanoprobes tend to accumulate in the liver and are difficult to metabolize, which leads to strong background signals and inadequate imaging quality of TAMs nearby the liver such as pancreatic cancer. Herein, we aim to develop metabolizable dextran-indocyanine green (DN-ICG) nanoprobes in the second near-infrared window (NIR-II, 1 000-1 700 nm) for dynamic imaging of TAMs in pancreatic cancer. Compared to free ICG, the NIR-II fluorescence intensity of DN-ICG nanoprobes increased by 279% with significantly improved stability. We demonstrated that DN-ICG nanoprobes could specifically target TAMs through the interaction of dextran with specific ICAM-3-grabbing nonintegrin related 1 (SIGN-R1), which were highly expressed in TAMs. Subsequently, DN-ICG nanoprobes gradually metabolized in the liver yet remained in pancreatic tumor stroma in mouse models, achieving a high signal-to-background ratio (SBR = 7) in deep tissue (∼0.5 cm) NIR-II imaging of TAMs. Moreover, DN-ICG nanoprobes could detect dynamic changes of TAMs induced by low-dose radiotherapy and zoledronic acid. Therefore, the highly biocompatible and biodegradable DN-ICG nanoprobes harbor great potential for precision therapy in pancreatic cancer.
Collapse
Affiliation(s)
- Xinping Luo
- NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, P. R. China
- Paul C. Lauterbur Research Center for Biomedical Imaging, Key Laboratory for Magnetic Resonance and Multimodality Imaging of Guangdong Province, Shenzhen Key Laboratory of Ultrasound Imaging and Therapy, CAS Key Laboratory of Health Informatics, Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, P. R. China
| | - Dehong Hu
- Paul C. Lauterbur Research Center for Biomedical Imaging, Key Laboratory for Magnetic Resonance and Multimodality Imaging of Guangdong Province, Shenzhen Key Laboratory of Ultrasound Imaging and Therapy, CAS Key Laboratory of Health Informatics, Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, P. R. China
| | - Duyang Gao
- Paul C. Lauterbur Research Center for Biomedical Imaging, Key Laboratory for Magnetic Resonance and Multimodality Imaging of Guangdong Province, Shenzhen Key Laboratory of Ultrasound Imaging and Therapy, CAS Key Laboratory of Health Informatics, Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, P. R. China
| | - Yuenan Wang
- Department of Radiaton Oncology, Peking University Shenzhen Hospital, No. 1120, Lianhua Road, Futian District, Shenzhen, Guangdong Province 518036, P. R. China
| | - Xinhua Chen
- Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University, Hangzhou 310003, P. R. China
| | - Xin Liu
- Paul C. Lauterbur Research Center for Biomedical Imaging, Key Laboratory for Magnetic Resonance and Multimodality Imaging of Guangdong Province, Shenzhen Key Laboratory of Ultrasound Imaging and Therapy, CAS Key Laboratory of Health Informatics, Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, P. R. China
| | - Hairong Zheng
- Paul C. Lauterbur Research Center for Biomedical Imaging, Key Laboratory for Magnetic Resonance and Multimodality Imaging of Guangdong Province, Shenzhen Key Laboratory of Ultrasound Imaging and Therapy, CAS Key Laboratory of Health Informatics, Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, P. R. China
| | - Minjie Sun
- NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, P. R. China
| | - Zonghai Sheng
- Paul C. Lauterbur Research Center for Biomedical Imaging, Key Laboratory for Magnetic Resonance and Multimodality Imaging of Guangdong Province, Shenzhen Key Laboratory of Ultrasound Imaging and Therapy, CAS Key Laboratory of Health Informatics, Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, P. R. China
| |
Collapse
|
29
|
Moody AS, Dayton PA, Zamboni WC. Imaging methods to evaluate tumor microenvironment factors affecting nanoparticle drug delivery and antitumor response. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2021; 4:382-413. [PMID: 34796317 PMCID: PMC8597952 DOI: 10.20517/cdr.2020.94] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 01/07/2021] [Accepted: 01/28/2021] [Indexed: 11/24/2022]
Abstract
Standard small molecule and nanoparticulate chemotherapies are used for cancer treatment; however, their effectiveness remains highly variable. One reason for this variable response is hypothesized to be due to nonspecific drug distribution and heterogeneity of the tumor microenvironment, which affect tumor delivery of the agents. Nanoparticle drugs have many theoretical advantages, but due to variability in tumor microenvironment (TME) factors, the overall drug delivery to tumors and associated antitumor response are low. The nanotechnology field would greatly benefit from a thorough analysis of the TME factors that create these physiological barriers to tumor delivery and treatment in preclinical models and in patients. Thus, there is a need to develop methods that can be used to reveal the content of the TME, determine how these TME factors affect drug delivery, and modulate TME factors to increase the tumor delivery and efficacy of nanoparticles. In this review, we will discuss TME factors involved in drug delivery, and how biomedical imaging tools can be used to evaluate tumor barriers and predict drug delivery to tumors and antitumor response.
Collapse
Affiliation(s)
- Amber S. Moody
- UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA
- UNC Lineberger Comprehensive Cancer Center, Chapel Hill, NC 27599, USA
- Carolina Institute for Nanomedicine, Chapel Hill, NC 27599, USA
- Joint Department of Biomedical Engineering, University of North Carolina and North Carolina State University, Chapel Hill, NC 27599, USA
| | - Paul A. Dayton
- UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA
- UNC Lineberger Comprehensive Cancer Center, Chapel Hill, NC 27599, USA
- Joint Department of Biomedical Engineering, University of North Carolina and North Carolina State University, Chapel Hill, NC 27599, USA
| | - William C. Zamboni
- UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA
- UNC Lineberger Comprehensive Cancer Center, Chapel Hill, NC 27599, USA
- Carolina Institute for Nanomedicine, Chapel Hill, NC 27599, USA
| |
Collapse
|
30
|
Martin JD, Miyazaki T, Cabral H. Remodeling tumor microenvironment with nanomedicines. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2021; 13:e1730. [PMID: 34124849 DOI: 10.1002/wnan.1730] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 05/21/2021] [Accepted: 05/22/2021] [Indexed: 12/17/2022]
Abstract
The tumor microenvironment (TME) has been recognized as a major contributor to cancer malignancy and therapeutic resistance. Thus, strategies directed to re-engineer the TME are emerging as promising approaches for improving the efficacy of antitumor therapies by enhancing tumor perfusion and drug delivery, as well as alleviating the immunosuppressive TME. In this regard, nanomedicine has shown great potential for developing effective treatments capable of re-modeling the TME by controlling drug action in a spatiotemporal manner and allowing long-lasting modulatory effects on the TME. Herein, we review recent progress on TME re-engineering by using nanomedicine, particularly focusing on formulations controlling TME characteristics through targeted interaction with cellular components of the TME. Importantly, the TME should be re-engineering to a quiescent phenotype rather than be destroyed. Finally, immediate challenges and future perspectives of TME-re-engineering nanomedicines are discussed, anticipating further innovation in this growing field. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease.
Collapse
Affiliation(s)
| | - Takuya Miyazaki
- Kanagawa Institute of Industrial Science and Technology, Ebina, Japan
| | - Horacio Cabral
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
31
|
Zhang K, Sun Y, Wu S, Zhou M, Zhang X, Zhou R, Zhang T, Gao Y, Chen T, Chen Y, Yao X, Watanabe Y, Tian M, Zhang H. Systematic imaging in medicine: a comprehensive review. Eur J Nucl Med Mol Imaging 2021; 48:1736-1758. [PMID: 33210241 DOI: 10.1007/s00259-020-05107-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Accepted: 11/08/2020] [Indexed: 01/05/2023]
Abstract
Systematic imaging can be broadly defined as the systematic identification and characterization of biological processes at multiple scales and levels. In contrast to "classical" diagnostic imaging, systematic imaging emphasizes on detecting the overall abnormalities including molecular, functional, and structural alterations occurring during disease course in a systematic manner, rather than just one aspect in a partial manner. Concomitant efforts including improvement of imaging instruments, development of novel imaging agents, and advancement of artificial intelligence are warranted for achievement of systematic imaging. It is undeniable that scientists and radiologists will play a predominant role in directing this burgeoning field. This article introduces several recent developments in imaging modalities and nanoparticles-based imaging agents, and discusses how systematic imaging can be achieved. In the near future, systematic imaging which combines multiple imaging modalities with multimodal imaging agents will pave a new avenue for comprehensive characterization of diseases, successful achievement of image-guided therapy, precise evaluation of therapeutic effects, and rapid development of novel pharmaceuticals, with the final goal of improving human health-related outcomes.
Collapse
Affiliation(s)
- Kai Zhang
- Department of Nuclear Medicine and PET center, The Second Hospital of Zhejiang University School of Medicine, No. 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China
- Laboratory for Pathophysiological and Health Science, RIKEN Center for Biosystems Dynamics Research, 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo, 650-0047, Japan
| | - Yujie Sun
- State Key Laboratory of Membrane Biology, Biodynamic Optical Imaging Center, School of Life Sciences, Peking University, Beijing, China
| | - Shuang Wu
- Department of Nuclear Medicine and PET center, The Second Hospital of Zhejiang University School of Medicine, No. 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China
| | - Min Zhou
- Department of Nuclear Medicine and PET center, The Second Hospital of Zhejiang University School of Medicine, No. 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China
- Institute of Translational Medicine, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xiaohui Zhang
- Department of Nuclear Medicine and PET center, The Second Hospital of Zhejiang University School of Medicine, No. 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China
| | - Rui Zhou
- Department of Nuclear Medicine and PET center, The Second Hospital of Zhejiang University School of Medicine, No. 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China
| | - Tingting Zhang
- Department of Nuclear Medicine and PET center, The Second Hospital of Zhejiang University School of Medicine, No. 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China
| | - Yuanxue Gao
- Department of Nuclear Medicine and PET center, The Second Hospital of Zhejiang University School of Medicine, No. 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China
| | - Ting Chen
- Department of Nuclear Medicine and PET center, The Second Hospital of Zhejiang University School of Medicine, No. 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China
| | - Yao Chen
- Department of Nuclear Medicine and PET center, The Second Hospital of Zhejiang University School of Medicine, No. 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China
| | - Xin Yao
- Department of Gastroenterology, The First Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Yasuyoshi Watanabe
- Laboratory for Pathophysiological and Health Science, RIKEN Center for Biosystems Dynamics Research, 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo, 650-0047, Japan.
| | - Mei Tian
- Department of Nuclear Medicine and PET center, The Second Hospital of Zhejiang University School of Medicine, No. 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China.
| | - Hong Zhang
- Department of Nuclear Medicine and PET center, The Second Hospital of Zhejiang University School of Medicine, No. 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China.
- Key Laboratory for Biomedical Engineering of Ministry of Education, Zhejiang University, Hangzhou, China.
- The College of Biomedical Engineering and Instrument Science of Zhejiang University, Hangzhou, China.
| |
Collapse
|
32
|
Hah YS, Koo KC. Immunology and Immunotherapeutic Approaches for Advanced Renal Cell Carcinoma: A Comprehensive Review. Int J Mol Sci 2021; 22:ijms22094452. [PMID: 33923219 PMCID: PMC8123195 DOI: 10.3390/ijms22094452] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Revised: 04/19/2021] [Accepted: 04/20/2021] [Indexed: 12/25/2022] Open
Abstract
Renal cell carcinoma (RCC) is a malignant tumor associated with various tumor microenvironments (TMEs). The immune system is activated by the development of cancer and drives T cell anti-tumor response. CD8 T cells are known to improve clinical outcomes and sensitivity to immunotherapy, and play a crucial role against tumors. In contrast, tumor-associated macrophages (TAMs) suppress immunity against malignancy and lead to tumor progression. TAMs are promoted from damaged TMEs and mount proinflammatory responses to pathogens. Initial immunotherapy consists of interferon-α and interleukin-2. However, response to such therapy is unclear in most patients, and it is associated with high levels of toxicity. Immune checkpoint inhibitors (ICIs), which up-regulate immune responses by blocking the programed cell death protein 1 (PD-1) receptor, the ligand of PD-1, or cytotoxic T-lymphocyte-associated protein 4 T cells, have led to a new era of immunotherapy. Furthermore, combination strategies with ICIs have proven effective through several randomized controlled trials. We expect the next generation of immunotherapy to lead to better outcomes based on ongoing trials and inspire new therapeutic strategies.
Collapse
Affiliation(s)
- Yoon-Soo Hah
- Department of Urology, Catholic University of Daegu School of Medicine, Daegu 42472, Korea;
| | - Kyo-Chul Koo
- Department of Urology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul 06229, Korea
- Correspondence: ; Tel.: +82-2-2019-3470
| |
Collapse
|
33
|
Positron Emission Tomography Imaging of Macrophages in Cancer. Cancers (Basel) 2021; 13:cancers13081921. [PMID: 33923410 PMCID: PMC8072570 DOI: 10.3390/cancers13081921] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 04/13/2021] [Accepted: 04/13/2021] [Indexed: 12/14/2022] Open
Abstract
Macrophages are large phagocytic cells that can be classified as a type of white blood cell and may be either mobile or stationary in tissues. The presence of macrophages in essentially every major disease makes them attractive candidates to serve as therapeutic targets and diagnostic biomarkers. Macrophages that are found in the microenvironment of solid tumors are referred to as tumor-associated macrophages (TAMs) and have been shown to influence chemoresistance, immune regulation, tumor initiation and tumor growth. The imaging of TAMs through Positron Emission Tomography (PET) has the potential to provide valuable information on cancer biology, tumor progression, and response to therapy. This review will highlight the versatility of macrophage imaging in cancer through the use of PET.
Collapse
|
34
|
Kimm MA, Klenk C, Alunni-Fabbroni M, Kästle S, Stechele M, Ricke J, Eisenblätter M, Wildgruber M. Tumor-Associated Macrophages-Implications for Molecular Oncology and Imaging. Biomedicines 2021; 9:biomedicines9040374. [PMID: 33918295 PMCID: PMC8066018 DOI: 10.3390/biomedicines9040374] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 03/29/2021] [Accepted: 03/31/2021] [Indexed: 12/21/2022] Open
Abstract
Tumor-associated macrophages (TAMs) represent the largest group of leukocytes within the tumor microenvironment (TME) of solid tumors and orchestrate the composition of anti- as well as pro-tumorigenic factors. This makes TAMs an excellent target for novel cancer therapies. The plasticity of TAMs resulting in varying membrane receptors and expression of intracellular proteins allow the specific characterization of different subsets of TAMs. Those markers similarly allow tracking of TAMs by different means of molecular imaging. This review aims to provides an overview of the origin of tumor-associated macrophages, their polarization in different subtypes, and how characteristic markers of the subtypes can be used as targets for molecular imaging and theranostic approaches.
Collapse
Affiliation(s)
- Melanie A. Kimm
- Department of Radiology, University Hospital, LMU Munich, 81377 Munich, Germany; (M.A.K.); (C.K.); (M.A.-F.); (S.K.); (M.S.); (J.R.)
| | - Christopher Klenk
- Department of Radiology, University Hospital, LMU Munich, 81377 Munich, Germany; (M.A.K.); (C.K.); (M.A.-F.); (S.K.); (M.S.); (J.R.)
| | - Marianna Alunni-Fabbroni
- Department of Radiology, University Hospital, LMU Munich, 81377 Munich, Germany; (M.A.K.); (C.K.); (M.A.-F.); (S.K.); (M.S.); (J.R.)
| | - Sophia Kästle
- Department of Radiology, University Hospital, LMU Munich, 81377 Munich, Germany; (M.A.K.); (C.K.); (M.A.-F.); (S.K.); (M.S.); (J.R.)
| | - Matthias Stechele
- Department of Radiology, University Hospital, LMU Munich, 81377 Munich, Germany; (M.A.K.); (C.K.); (M.A.-F.); (S.K.); (M.S.); (J.R.)
| | - Jens Ricke
- Department of Radiology, University Hospital, LMU Munich, 81377 Munich, Germany; (M.A.K.); (C.K.); (M.A.-F.); (S.K.); (M.S.); (J.R.)
| | - Michel Eisenblätter
- Department of Diagnostic and Interventional Radiology, Freiburg University Hospital, 79106 Freiburg, Germany;
| | - Moritz Wildgruber
- Department of Radiology, University Hospital, LMU Munich, 81377 Munich, Germany; (M.A.K.); (C.K.); (M.A.-F.); (S.K.); (M.S.); (J.R.)
- Correspondence: ; Tel.: +49-0-89-4400-76640
| |
Collapse
|
35
|
Pallares R, Abergel RJ. Diagnostic, Prognostic, and Therapeutic Use of Radiopharmaceuticals in the Context of SARS-CoV-2. ACS Pharmacol Transl Sci 2021; 4:1-7. [PMID: 33615159 PMCID: PMC7839413 DOI: 10.1021/acsptsci.0c00186] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Indexed: 01/18/2023]
Abstract
The coronavirus disease 2019 (COVID-19) outbreak has devastated the healthcare systems and economies of over 200 countries in just a few months. The etiological agent of COVID-19, SARS-CoV-2, is a highly contagious virus that can be transmitted by asymptomatic and symptomatic carriers alike. While in vitro testing techniques have allowed for population-wide screening, prognostic tools are required to assess the disease severity and therapeutic response, contributing to improve the patient clinical outcomes. Moreover, no specific antiviral against COVID-19 exists at the time of publication, severely limiting treatment against the infection. Hence, there is an urgent clinical need for innovative therapeutic strategies that may contribute to manage the COVID-19 outbreak and prevent future pandemics. Herein, we critically examine recent diagnostic, prognostic, and therapeutic advancements for COVID-19 in the field of radiopharmaceuticals. First, we summarize the gold standard techniques used to diagnose COVID-19, including in vitro assays and imaging techniques, and then discuss how radionuclide-based nuclear imaging provides complementary information for prognosis and treatment management of infected patients. Second, we introduce new emerging types of radiotherapies that employ radioimmunoconjugates, which have shown selective cytotoxic response in oncological studies, and critically analyze how these compounds could be used as therapeutic agents against SARS-CoV-2. Finally, this Perspective further discusses the emerging applications of radionuclides to study the behavior of pulmonary SARS-CoV-2 aerosol particles.
Collapse
Affiliation(s)
- Roger
M. Pallares
- Chemical
Sciences Division, Lawrence Berkeley National
Laboratory, Berkeley, California 94720, United States
| | - Rebecca J. Abergel
- Chemical
Sciences Division, Lawrence Berkeley National
Laboratory, Berkeley, California 94720, United States
- Department
of Nuclear Engineering, University of California, Berkeley, California 94720, United States
| |
Collapse
|
36
|
Pellico J, Gawne PJ, T M de Rosales R. Radiolabelling of nanomaterials for medical imaging and therapy. Chem Soc Rev 2021; 50:3355-3423. [PMID: 33491714 DOI: 10.1039/d0cs00384k] [Citation(s) in RCA: 141] [Impact Index Per Article: 35.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Nanomaterials offer unique physical, chemical and biological properties of interest for medical imaging and therapy. Over the last two decades, there has been an increasing effort to translate nanomaterial-based medicinal products (so-called nanomedicines) into clinical practice and, although multiple nanoparticle-based formulations are clinically available, there is still a disparity between the number of pre-clinical products and those that reach clinical approval. To facilitate the efficient clinical translation of nanomedicinal-drugs, it is important to study their whole-body biodistribution and pharmacokinetics from the early stages of their development. Integrating this knowledge with that of their therapeutic profile and/or toxicity should provide a powerful combination to efficiently inform nanomedicine trials and allow early selection of the most promising candidates. In this context, radiolabelling nanomaterials allows whole-body and non-invasive in vivo tracking by the sensitive clinical imaging techniques positron emission tomography (PET), and single photon emission computed tomography (SPECT). Furthermore, certain radionuclides with specific nuclear emissions can elicit therapeutic effects by themselves, leading to radionuclide-based therapy. To ensure robust information during the development of nanomaterials for PET/SPECT imaging and/or radionuclide therapy, selection of the most appropriate radiolabelling method and knowledge of its limitations are critical. Different radiolabelling strategies are available depending on the type of material, the radionuclide and/or the final application. In this review we describe the different radiolabelling strategies currently available, with a critical vision over their advantages and disadvantages. The final aim is to review the most relevant and up-to-date knowledge available in this field, and support the efficient clinical translation of future nanomedicinal products for in vivo imaging and/or therapy.
Collapse
Affiliation(s)
- Juan Pellico
- School of Biomedical Engineering & Imaging Sciences, King's College London, St. Thomas' Hospital, London SE1 7EH, UK.
| | | | | |
Collapse
|
37
|
Yuan Y, Long L, Liu J, Lin Y, Peng C, Tang Y, Zhou X, Li S, Zhang C, Li X, Zhou X. The double-edged sword effect of macrophage targeting delivery system in different macrophage subsets related diseases. J Nanobiotechnology 2020; 18:168. [PMID: 33198758 PMCID: PMC7667812 DOI: 10.1186/s12951-020-00721-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Accepted: 10/24/2020] [Indexed: 01/01/2023] Open
Abstract
Background Monocyte/macrophage-targeting delivery systems (MTDSs) have been focused upon as an emerging routine for delivering drugs to treat various macrophage-related diseases. However, the ability of MTDSs to distinguish different macrophage-related diseases and their impact on macrophage function and disease progression have not been systematically revealed, which is important for actively targeted therapeutic or diagnostic strategies. Results Herein, we used dextran-modified polystyrene nanoparticles (DEX-PS) to demonstrate that modification of nanoparticles by dextran can specifically enhance their recognition by M2 macrophages in vitro, but it is obstructed by monocytes in peripheral blood according to in vivo assays. DEX-PS not only targeted and became distributed in tumors, an M2 macrophage-related disease, but was also highly distributed in an M1 macrophage-related disease, namely acute peritonitis. Thus, DEX-PS acts as a double-edged sword in these two different diseases by reeducating macrophages to a pro-inflammatory phenotype. Conclusions Our results suggest that MTDSs, even those designed based on differential expression of receptors on specific macrophage subtypes, lack the ability to distinguish different macrophage subtype-related diseases in vivo. In addition to the potential impact of these carrier materials on macrophage function, studies of MTDSs should pay greater attention to the distribution of nanoparticles in non-target macrophage-infiltrated disease sites and their impact on disease processes.![]()
Collapse
Affiliation(s)
- Yuchuan Yuan
- Department of Pharmaceutics, College of Pharmacy, Army Medical University, Chongqing, 400038, China
| | - Ling Long
- Department of Oncology, Xinqiao Hospital, Army Medical University, Chongqing, 400042, China
| | - Jiaxing Liu
- Department of Pharmaceutics, College of Pharmacy, Army Medical University, Chongqing, 400038, China
| | - Yongyao Lin
- Department of Pharmaceutics, College of Pharmacy, Army Medical University, Chongqing, 400038, China
| | - Cuiping Peng
- Department of Pharmaceutics, College of Pharmacy, Army Medical University, Chongqing, 400038, China
| | - Yue Tang
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 400054, China
| | - Xuemei Zhou
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 400054, China
| | - Shuhui Li
- Department of Clinical Biochemistry, College of Pharmacy, Army Medical University, Chongqing, 400038, China
| | - Chengyuan Zhang
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 400054, China.
| | - Xiaohui Li
- Department of Pharmaceutics, College of Pharmacy, Army Medical University, Chongqing, 400038, China.
| | - Xing Zhou
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 400054, China.
| |
Collapse
|
38
|
Mukhtar M, Ali H, Ahmed N, Munir R, Talib S, Khan AS, Ambrus R. Drug delivery to macrophages: a review of nano-therapeutics targeted approach for inflammatory disorders and cancer. Expert Opin Drug Deliv 2020; 17:1239-1257. [PMID: 32543950 DOI: 10.1080/17425247.2020.1783237] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
INTRODUCTION Macrophages are involved in the normal defense of the body; however, the varying phenotypes of macrophages and imbalance in their ratio lead to the impairment of immune response initiating the production of inflammation. As the role of macrophages in immunological disorders and their surface receptors modulation has already been manifested; hence, macrophages can be exploited to make them a viable candidate for targeted delivery, which was not possible with previously designed conventional therapies for the immune disorders. AREAS COVERED Nanotechnology is a promising, clear cut, efficient, and adequate approach for targeting macrophages. Literature addresses the receptors available for targeting and the novel small dimensional therapeutic delivery vehicles to target them along with a brief overview of the role of macrophages in these diseases. Furthermore, the patents based on this idea are also listed. EXPERT OPINION Targeted drug delivery to macrophages should take into consideration the plasticity of macrophages and their modulation over time in the diseases. A cost-effective scale-up method of development will further facilitate the clinical trials. Besides, the implementation of safety guidelines to target macrophages and the studies of long-term effects of targeted approaches in humans would highly encourage the clinical outcomes.
Collapse
Affiliation(s)
- Mahwash Mukhtar
- Faculty of Pharmacy, Institute of Pharmaceutical Technology and Regulatory Affairs, University of Szeged , Szeged, Hungary.,Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University , Islamabad, Pakistan
| | - Hussain Ali
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University , Islamabad, Pakistan
| | - Naveed Ahmed
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University , Islamabad, Pakistan
| | - Rashid Munir
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University , Islamabad, Pakistan
| | - Sumbal Talib
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University , Islamabad, Pakistan
| | - Anam S Khan
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University , Islamabad, Pakistan
| | - Rita Ambrus
- Faculty of Pharmacy, Institute of Pharmaceutical Technology and Regulatory Affairs, University of Szeged , Szeged, Hungary
| |
Collapse
|
39
|
Copeland BT, Shallal H, Shen C, Pienta KJ, Foss CA, Pomper MG. Imaging and Characterization of Macrophage Distribution in Mouse Models of Human Prostate Cancer. Mol Imaging Biol 2020; 21:1054-1063. [PMID: 30805886 DOI: 10.1007/s11307-019-01318-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
PURPOSE Prostate carcinoma consists of tumor epithelium and malignant stroma. Until recently, diagnostic and therapeutic efforts have focused exclusively on targeting characteristics of the tumor epithelium, ignoring opportunities to target inflammatory infiltrate and extracellular matrix components. Prostate tumors are rich in tumor-associated macrophages (TAMs), which can be either of the cytotoxic M1 or protumorigenic M2 phenotype. We have quantified the proportion of each in seven common human prostate tumor lines grown subcutaneously in athymic nude mice and have imaged macrophage densities in vivo in xenografts derived from these lines. PROCEDURES A panel of seven human prostate cancer xenografts was generated in intact male athymic nude mice reflecting variable expression of the androgen receptor (AR) and prostate-specific membrane antigen (PSMA). Mice were imaged ex vivo using near-infrared fluorescence (NIRF) imaging for PSMA expression and total macrophage densities to enable direct comparison between the two. Tumors were harvested for sectioning and additional staining to delineate M1 and M2 phenotype along with vascular density. RESULTS Macrophage polarization analysis of sections revealed that all xenografts were > 94% M2 phenotype, and the few M1-polarized macrophages present were confined to the periphery. Xenografts displaying the fastest growth were associated with the highest densities of macrophages while the slowest growing tumors were characterized by focal, tumor-infiltrating macrophage densities. Xenograft sections displayed a strong positive spatial relationship between macrophages, vasculature, and PSMA expression. CONCLUSIONS Prostate TAM disposition can be imaged ex vivo and is associated with growth characteristics of a variety of tumor subtypes regardless of PSMA or AR expression.
Collapse
Affiliation(s)
- Ben T Copeland
- The Russell H Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
- Beckman Research Institute, City of Hope, Duarte, CA, 91010, USA
| | - Hassan Shallal
- The Russell H Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Chentian Shen
- The Russell H Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Kenneth J Pienta
- James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Catherine A Foss
- The Russell H Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA.
| | - Martin G Pomper
- The Russell H Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA.
- James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
| |
Collapse
|
40
|
Zhang C, Chen Z, Li W, Liu X, Tang S, Jiang L, Li M, Peng H, Lian M. Influences of different sugar ligands on targeted delivery of liposomes. J Drug Target 2020; 28:789-801. [PMID: 32242754 DOI: 10.1080/1061186x.2020.1744156] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Ligands are an important part of targeted drug delivery systems. Optimised lignads not only improve the target efficiency, but also enhance therapeutical effect of drugs. In our research, five sugar molecules (Mannose, Galactose, Glucose, Malt disaccharide, and Maltotriose) conjugated PEG600-DSPE were synthesised, of which polysaccharides were first discovered by us as sugar ligands to modify liposomes, which interacts with over expressive GLUT on cancer cells. DiO was encapsulated as fluorescent probe to evaluate their cellular uptake abilities of targeting C6 glioma cells, and the distribution in different visceral organs of rats. The results demonstrated that Malt disaccharide and Glucose-PEG600-DSPE had the strong efficiency of cellular uptake by C6 glioma cells. The distribution and accumulation of liposomes showed that different sugars modified liposomes could target different visceral organs in rats. It has provided a novel idea for ligand selectivity and optimisation of nanocarriers for tumour targeted therapy.
Collapse
Affiliation(s)
- Changmei Zhang
- Department of Pharmaceutics, Daqing Campus of Harbin Medical University, Harbin, China
| | - Zhong Chen
- Department of Pharmaceutics, Daqing Campus of Harbin Medical University, Harbin, China
| | - Wenhua Li
- Department of Pharmaceutics, Daqing Campus of Harbin Medical University, Harbin, China
| | - Xiaoying Liu
- Department of Pharmaceutics, Daqing Campus of Harbin Medical University, Harbin, China
| | - Shukun Tang
- Department of Pharmaceutics, Daqing Campus of Harbin Medical University, Harbin, China
| | - Lei Jiang
- Department of Pharmaceutics, Daqing Campus of Harbin Medical University, Harbin, China
| | - Minghui Li
- Department of Pharmaceutics, Daqing Campus of Harbin Medical University, Harbin, China
| | - Haisheng Peng
- Department of Pharmaceutics, Daqing Campus of Harbin Medical University, Harbin, China
| | - Mingming Lian
- Department of Pharmaceutics, Daqing Campus of Harbin Medical University, Harbin, China
| |
Collapse
|
41
|
Cancer Immunoimaging with Smart Nanoparticles. Trends Biotechnol 2020; 38:388-403. [DOI: 10.1016/j.tibtech.2019.11.001] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 10/30/2019] [Accepted: 11/05/2019] [Indexed: 12/31/2022]
|
42
|
Kumar S, Ramesh A, Kulkarni A. Targeting macrophages: a novel avenue for cancer drug discovery. Expert Opin Drug Discov 2020; 15:561-574. [PMID: 32141351 DOI: 10.1080/17460441.2020.1733525] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Introduction: Tumor-associated macrophages (TAMs) make up a significant portion of the tumor microenvironment. Emerging clinical evidence indicate that cytokines present in the tumor microenvironment influence TAMs to play an immunosuppressive role by acquiring a pro-tumoral phenotype. However, TAMs are inherently plastic cells that can be phenotypically reprogrammed to elicit an anti-tumoral response. Therapeutic strategies that focus on targeting TAMs have opened new avenues for drug discoveries.Areas covered: This review discusses recent developments in TAM targeted immunotherapy in both preclinical and clinical settings. This article highlights the potential signaling pathways that can be targeted for macrophage reprogramming and discusses the progress of current clinical trials involved in TAMs targeting. Novel nanoparticle-based drug delivery strategies involved in macrophage-based cancer therapeutics and diagnostics are also discussed.Expert opinion: TAM targeted therapies have limited success in clinics due to reasons such as insufficient inhibition of signaling pathways, lower drug accumulation in the tumor, activation of feedback signaling pathways that induce resistance to monotherapies and systemic dose-related toxicities. Nanoparticle-based delivery platforms could overcome these challenges since they enable encapsulation of multiple drugs that target different signaling pathways and enhance intratumoral delivery and can enable delivery of imaging agents.
Collapse
Affiliation(s)
- Sahana Kumar
- Department of Chemical Engineering, University of Massachusetts, Amherst, MA, USA
| | - Anujan Ramesh
- Department of Chemical Engineering, University of Massachusetts, Amherst, MA, USA.,Department of Biomedical Engineering, University of Massachusetts, Amherst, MA, USA
| | - Ashish Kulkarni
- Department of Chemical Engineering, University of Massachusetts, Amherst, MA, USA.,Department of Biomedical Engineering, University of Massachusetts, Amherst, MA, USA.,Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA, USA
| |
Collapse
|
43
|
Targeting Tumors Using Peptides. Molecules 2020; 25:molecules25040808. [PMID: 32069856 PMCID: PMC7070747 DOI: 10.3390/molecules25040808] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 02/07/2020] [Accepted: 02/10/2020] [Indexed: 12/16/2022] Open
Abstract
To penetrate solid tumors, low molecular weight (Mw < 10 KDa) compounds have an edge over antibodies: their higher penetration because of their small size. Because of the dense stroma and high interstitial fluid pressure of solid tumors, the penetration of higher Mw compounds is unfavored and being small thus becomes an advantage. This review covers a wide range of peptidic ligands—linear, cyclic, macrocyclic and cyclotidic peptides—to target tumors: We describe the main tools to identify peptides experimentally, such as phage display, and the possible chemical modifications to enhance the properties of the identified peptides. We also review in silico identification of peptides and the most salient non-peptidic ligands in clinical stages. We later focus the attention on the current validated ligands available to target different tumor compartments: blood vessels, extracelullar matrix, and tumor associated macrophages. The clinical advances and failures of these ligands and their therapeutic conjugates will be discussed. We aim to present the reader with the state-of-the-art in targeting tumors, by using low Mw molecules, and the tools to identify new ligands.
Collapse
|
44
|
Arlt A, von Bonin F, Rehberg T, Perez-Rubio P, Engelmann JC, Limm K, Reinke S, Dullin C, Sun X, Specht R, Maulhardt M, Linke F, Bunt G, Klapper W, Vockerodt M, Wilting J, Pukrop T, Dettmer K, Gronwald W, Oefner PJ, Spang R, Kube D. High CD206 levels in Hodgkin lymphoma-educated macrophages are linked to matrix-remodeling and lymphoma dissemination. Mol Oncol 2020; 14:571-589. [PMID: 31825135 PMCID: PMC7053241 DOI: 10.1002/1878-0261.12616] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 11/08/2019] [Accepted: 12/09/2019] [Indexed: 12/14/2022] Open
Abstract
Macrophages (Mφ) are abundantly present in the tumor microenvironment and may predict outcome in solid tumors and defined lymphoma subtypes. Mφ heterogeneity, the mechanisms of their recruitment, and their differentiation into lymphoma‐promoting, alternatively activated M2‐like phenotypes are still not fully understood. Therefore, further functional studies are required to understand biological mechanisms associated with human tumor‐associated Mφ (TAM). Here, we show that the global mRNA expression and protein abundance of human Mφ differentiated in Hodgkin lymphoma (HL)‐conditioned medium (CM) differ from those of Mφ educated by conditioned media from diffuse large B‐cell lymphoma (DLBCL) cells or, classically, by macrophage colony‐stimulating factor (M‐CSF). Conditioned media from HL cells support TAM differentiation through upregulation of surface antigens such as CD40, CD163, CD206, and PD‐L1. In particular, RNA and cell surface protein expression of mannose receptor 1 (MRC1)/CD206 significantly exceed the levels induced by classical M‐CSF stimulation in M2‐like Mφ; this is regulated by interleukin 13 to a large extent. Functionally, high CD206 enhances mannose‐dependent endocytosis and uptake of type I collagen. Together with high matrix metalloprotease9 secretion, HL‐TAMs appear to be active modulators of the tumor matrix. Preclinical in ovo models show that co‐cultures of HL cells with monocytes or Mφ support dissemination of lymphoma cells via lymphatic vessels, while tumor size and vessel destruction are decreased in comparison with lymphoma‐only tumors. Immunohistology of human HL tissues reveals a fraction of cases feature large numbers of CD206‐positive cells, with high MRC1 expression being characteristic of HL‐stage IV. In summary, the lymphoma‐TAM interaction contributes to matrix‐remodeling and lymphoma cell dissemination.
Collapse
Affiliation(s)
- Annekatrin Arlt
- Clinic of Hematology and Medical Oncology, University Medical Centre Göttingen, Germany.,Network BMBF eMed MMML-Demonstrators, Regensburg, Germany
| | - Frederike von Bonin
- Clinic of Hematology and Medical Oncology, University Medical Centre Göttingen, Germany
| | - Thorsten Rehberg
- Statistical Bioinformatics, Institute of Functional Genomics, University of Regensburg, Germany
| | - Paula Perez-Rubio
- Network BMBF eMed MMML-Demonstrators, Regensburg, Germany.,Statistical Bioinformatics, Institute of Functional Genomics, University of Regensburg, Germany
| | - Julia C Engelmann
- Network BMBF eMed MMML-Demonstrators, Regensburg, Germany.,Statistical Bioinformatics, Institute of Functional Genomics, University of Regensburg, Germany
| | - Katharina Limm
- Institute of Functional Genomics, University of Regensburg, Germany
| | - Sarah Reinke
- Department of Pathology, Hematopathology Section, UKSH Campus Kiel, Germany
| | - Christian Dullin
- Institute of Diagnostic and Interventional Radiology, University Medical Centre Göttingen, Germany
| | - Xueni Sun
- Institute of Functional Genomics, University of Regensburg, Germany
| | - Rieke Specht
- Clinic of Hematology and Medical Oncology, University Medical Centre Göttingen, Germany
| | - Markus Maulhardt
- Clinic of Hematology and Medical Oncology, University Medical Centre Göttingen, Germany
| | - Franziska Linke
- Clinic of Hematology and Medical Oncology, University Medical Centre Göttingen, Germany
| | - Gertrude Bunt
- Clinical Optical Microscopy, Institute of Neuropathology, University Medical Centre Göttingen, Germany
| | - Wolfram Klapper
- Department of Pathology, Hematopathology Section, UKSH Campus Kiel, Germany
| | - Martina Vockerodt
- Institute of Anatomy and Cell Biology, University Medical Centre Göttingen, Germany
| | - Jörg Wilting
- Network BMBF eMed MMML-Demonstrators, Regensburg, Germany.,Institute of Anatomy and Cell Biology, University Medical Centre Göttingen, Germany
| | - Tobias Pukrop
- Network BMBF eMed MMML-Demonstrators, Regensburg, Germany.,Department of Internal Medicine III, Hematology and Medical Oncology, University Hospital Regensburg, Germany
| | - Katja Dettmer
- Network BMBF eMed MMML-Demonstrators, Regensburg, Germany.,Institute of Functional Genomics, University of Regensburg, Germany
| | - Wolfram Gronwald
- Network BMBF eMed MMML-Demonstrators, Regensburg, Germany.,Institute of Functional Genomics, University of Regensburg, Germany
| | - Peter J Oefner
- Institute of Functional Genomics, University of Regensburg, Germany
| | - Rainer Spang
- Network BMBF eMed MMML-Demonstrators, Regensburg, Germany.,Statistical Bioinformatics, Institute of Functional Genomics, University of Regensburg, Germany
| | - Dieter Kube
- Clinic of Hematology and Medical Oncology, University Medical Centre Göttingen, Germany.,Network BMBF eMed MMML-Demonstrators, Regensburg, Germany
| |
Collapse
|
45
|
Cellular and Extracellular Components in Tumor Microenvironment and Their Application in Early Diagnosis of Cancers. Anal Cell Pathol (Amst) 2020; 2020:6283796. [PMID: 32377504 PMCID: PMC7199555 DOI: 10.1155/2020/6283796] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 12/11/2019] [Accepted: 12/23/2019] [Indexed: 12/11/2022] Open
Abstract
Tumors are surrounded by complex environmental components, including blood and lymph vessels, fibroblasts, endothelial cells, immune cells, cytokines, extracellular vesicles, and extracellular matrix. All the stromal components together with the tumor cells form the tumor microenvironment (TME). In addition, extracellular physical and chemical factors, including extracellular pH, hypoxia, elevated interstitial fluid pressure, and fibrosis, are closely associated with tumor progression, metastasis, immunosuppression, and drug resistance. Cellular and extracellular components in TME contribute to nearly all procedures of carcinogenesis. By summarizing the recent work in this field, we make a comprehensive review on the role of cellular and extracellular components in the process of carcinogenesis and their potential application in early diagnosis of cancer. We hope that a systematic review of the diverse aspects of TME will help both research scientists and clinicians in this field.
Collapse
|
46
|
Pérez-Medina C, Teunissen AJ, Kluza E, Mulder WJ, van der Meel R. Nuclear imaging approaches facilitating nanomedicine translation. Adv Drug Deliv Rev 2020; 154-155:123-141. [PMID: 32721459 DOI: 10.1016/j.addr.2020.07.017] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 07/08/2020] [Accepted: 07/17/2020] [Indexed: 02/07/2023]
Abstract
Nanomedicine approaches can effectively modulate the biodistribution and bioavailability of therapeutic agents, improving their therapeutic index. However, despite the ever-increasing amount of literature reporting on preclinical nanomedicine, the number of nanotherapeutics receiving FDA approval remains relatively low. Several barriers exist that hamper the effective preclinical evaluation and clinical translation of nanotherapeutics. Key barriers include insufficient understanding of nanomedicines' in vivo behavior, inadequate translation from murine models to larger animals, and a lack of patient stratification strategies. Integrating quantitative non-invasive imaging techniques in nanomedicine development offers attractive possibilities to address these issues. Among the available imaging techniques, nuclear imaging by positron emission tomography (PET) and single-photon emission computed tomography (SPECT) are highly attractive in this context owing to their quantitative nature and uncontested sensitivity. In basic and translational research, nuclear imaging techniques can provide critical quantitative information about pharmacokinetic parameters, biodistribution profiles or target site accumulation of nanocarriers and their associated payload. During clinical evaluation, nuclear imaging can be used to select patients amenable to nanomedicine treatment. Here, we review how nuclear imaging-based approaches are increasingly being integrated into nanomedicine development and discuss future developments that will accelerate their clinical translation.
Collapse
|
47
|
Ge J, Zhang Q, Zeng J, Gu Z, Gao M. Radiolabeling nanomaterials for multimodality imaging: New insights into nuclear medicine and cancer diagnosis. Biomaterials 2019; 228:119553. [PMID: 31689672 DOI: 10.1016/j.biomaterials.2019.119553] [Citation(s) in RCA: 93] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 10/15/2019] [Accepted: 10/15/2019] [Indexed: 12/22/2022]
Abstract
Nuclear medicine imaging has been developed as a powerful diagnostic approach for cancers by detecting gamma rays directly or indirectly from radionuclides to construct images with beneficial characteristics of high sensitivity, infinite penetration depth and quantitative capability. Current nuclear medicine imaging modalities mainly include single-photon emission computed tomography (SPECT) and positron emission tomography (PET) that require administration of radioactive tracers. In recent years, a vast number of radioactive tracers have been designed and constructed to improve nuclear medicine imaging performance toward early and accurate diagnosis of cancers. This review will discuss recent progress of nuclear medicine imaging tracers and associated biomedical imaging applications. Radiolabeling nanomaterials for rational development of tracers will be comprehensively reviewed with highlights on radiolabeling approaches (surface coupling, inner incorporation and interface engineering), providing profound understanding on radiolabeling chemistry and the associated imaging functionalities. The applications of radiolabeled nanomaterials in nuclear medicine imaging-related multimodality imaging will also be summarized with typical paradigms described. Finally, key challenges and new directions for future research will be discussed to guide further advancement and practical use of radiolabeled nanomaterials for imaging of cancers.
Collapse
Affiliation(s)
- Jianxian Ge
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Suzhou, 215123, China
| | - Qianyi Zhang
- School of Chemical Engineering and Australian Centre for NanoMedicine (ACN), University of New South Wales, Sydney, NSW, 2052, Australia
| | - Jianfeng Zeng
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Suzhou, 215123, China.
| | - Zi Gu
- School of Chemical Engineering and Australian Centre for NanoMedicine (ACN), University of New South Wales, Sydney, NSW, 2052, Australia.
| | - Mingyuan Gao
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Suzhou, 215123, China; Institute of Chemistry, Chinese Academy of Sciences/School of Chemistry and Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100190, China
| |
Collapse
|
48
|
Mukherjee S, Sonanini D, Maurer A, Daldrup-Link HE. The yin and yang of imaging tumor associated macrophages with PET and MRI. Am J Cancer Res 2019; 9:7730-7748. [PMID: 31695797 PMCID: PMC6831464 DOI: 10.7150/thno.37306] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 08/27/2019] [Indexed: 12/14/2022] Open
Abstract
Tumor associated macrophages (TAM) are key players in the cancer microenvironment. Molecular imaging modalities such as MRI and PET can be used to track and monitor TAM dynamics in tumors non-invasively, based on specific uptake and quantification of MRI-detectable nanoparticles or PET-detectable radiotracers. Particular molecular signatures can be leveraged to target anti-inflammatory TAM, which support tumor growth, and pro-inflammatory TAM, which suppress tumor growth. In addition, TAM-directed imaging probes can be designed to include immune modulating properties, thereby leading to combined diagnostic and therapeutic (theranostic) effects. In this review, we will discuss the complementary role of TAM-directed radiotracers and iron oxide nanoparticles for monitoring cancer immunotherapies with PET and MRI technologies. In addition, we will outline how TAM-directed imaging and therapy is interdependent and can be connected towards improved clinical outcomes
Collapse
|
49
|
Rodell CB, Koch PD, Weissleder R. Screening for new macrophage therapeutics. Theranostics 2019; 9:7714-7729. [PMID: 31695796 PMCID: PMC6831478 DOI: 10.7150/thno.34421] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 06/19/2019] [Indexed: 12/15/2022] Open
Abstract
Myeloid derived macrophages play a key role in many human diseases, and their therapeutic modulation via pharmacological means is receiving considerable attention. Of particular interest is the fact that these cells are i) dynamic phenotypes well suited to therapeutic manipulation and ii) phagocytic, allowing them to be efficiently targeted with nanoformulations. However, it is important to consider that macrophages represent heterogeneous populations of subtypes with often competing biological behaviors and functions. In order to develop next generation therapeutics, it is therefore essential to screen for biological effects through a combination of in vitro and in vivo assays. Here, we review the state-of-the-art techniques, including both cell based screens and in vivo imaging tools that have been developed for assessment of macrophage phenotype. We conclude with a forward-looking perspective on the growing need for noninvasive macrophage assessment and laboratory assays to be put into clinical practice and the potential broader impact of myeloid-targeted therapeutics.
Collapse
|
50
|
Vuong L, Kotecha RR, Voss MH, Hakimi AA. Tumor Microenvironment Dynamics in Clear-Cell Renal Cell Carcinoma. Cancer Discov 2019; 9:1349-1357. [PMID: 31527133 DOI: 10.1158/2159-8290.cd-19-0499] [Citation(s) in RCA: 258] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 07/02/2019] [Accepted: 08/02/2019] [Indexed: 12/30/2022]
Abstract
Renal cell carcinoma stands out as one of the most immune-infiltrated tumors in pan-cancer comparisons. Features of the tumor microenvironment heavily affect disease biology and may affect responses to systemic therapy. With evolving frontline options in the metastatic setting, several immune checkpoint blockade regimens have emerged as efficacious, and there is growing interest in characterizing features of tumor biology that can reproducibly prognosticate patients and/or predict the likelihood of their deriving therapeutic benefit. Herein, we review pertinent characteristics of the tumor microenvironment with dedicated attention to candidate prognostic and predictive signatures as well as possible targets for future drug development. SIGNIFICANCE: Tumor microenvironment features broadly characterizing angiogenesis and inflammatory signatures have shown striking differences in response to immune checkpoint blockade and antiangiogenic agents. Integration of stromal and immune biomarkers may hence produce predictive and prognostic signatures to guide management with existing regimens as well as future drug development.
Collapse
Affiliation(s)
- Lynda Vuong
- Immunogenomics and Precision Oncology Platform, Memorial Sloan Kettering Cancer Center, New York, New York.,Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Ritesh R Kotecha
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Martin H Voss
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - A Ari Hakimi
- Immunogenomics and Precision Oncology Platform, Memorial Sloan Kettering Cancer Center, New York, New York. .,Department of Urology, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|