1
|
Jin J, Barnett JD, Mironchik Y, Gross J, Kobayashi H, Levin A, Bhujwalla ZM. Photoimmunotheranostics of epithelioid sarcoma by targeting CD44 or EGFR. Transl Oncol 2024; 45:101966. [PMID: 38663219 PMCID: PMC11063645 DOI: 10.1016/j.tranon.2024.101966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 04/05/2024] [Accepted: 04/16/2024] [Indexed: 05/05/2024] Open
Abstract
Epithelioid sarcoma (ES) is a rare soft tissue neoplasm with high recurrence rates. Wide surgical resection remains the only potential curative treatment. ES presents most commonly on the fingers, hands and forearm, making light-based cancer cell-targeted therapies such as near-infrared photoimmunotherapy (NIR-PIT) that is target-specific, but with limited penetration depth, suitable for ES treatment. We established that CD44 and EGFR were overexpressed in ES patient samples and in the VA-ES-BJ human ES cell line. NIR-PIT of VA-ES-BJ cells using antibody photosensitizer conjugates, prepared by conjugating a CD44 or EGFR monoclonal antibody to the photosensitizer IR700, confirmed that NIR-PIT with both conjugates resulted in cell death. Neither treatment with NIR light alone nor treatment with the conjugates but without NIR light were effective. CD44-IR700-PIT resulted in greater cell death than EGFR-IR700-PIT, consistent with the increased expression of CD44 by VA-ES-BJ cells. In tumors, EGFR-IR700 exhibited a higher tumor-to-normal ratio, as determined by in vivo fluorescence imaging, and a higher anti-tumor growth effect, compared to CD44-IR700. No antitumor effect of the EGFR antibody or the photosensitizer conjugate alone was observed in vivo. Our data support evaluating the use of EGFR-IR700-PIT in the management of ES for detecting and eliminating ES cells in surgical margins, and in the treatment of superficial recurrent tumors.
Collapse
Affiliation(s)
- Jiefu Jin
- Division of Cancer Imaging Research, The Russell H Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - James D Barnett
- Division of Cancer Imaging Research, The Russell H Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Yelena Mironchik
- Division of Cancer Imaging Research, The Russell H Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - John Gross
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Hisataka Kobayashi
- Laboratory of Molecular Theranostics, Molecular Imaging Branch, NCI/NIH, Bethesda, MD, USA
| | - Adam Levin
- Orthopaedic Oncology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Zaver M Bhujwalla
- Division of Cancer Imaging Research, The Russell H Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA; Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Radiation Oncology and Molecular Radiation Sciences, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
2
|
Yang CW, Liu K, Yao CY, Li B, Juhong A, Ullah AKMA, Bumpers H, Qiu Z, Huang X. Active Targeting Hyaluronan Conjugated Nanoprobe for Magnetic Particle Imaging and Near-Infrared Fluorescence Imaging of Breast Cancer and Lung Metastasis. ACS APPLIED MATERIALS & INTERFACES 2024; 16:27055-27064. [PMID: 38757711 PMCID: PMC11145589 DOI: 10.1021/acsami.4c01623] [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: 01/30/2024] [Revised: 05/07/2024] [Accepted: 05/13/2024] [Indexed: 05/18/2024]
Abstract
A major contributing cause to breast cancer related death is metastasis. Moreover, breast cancer metastasis often shows little symptoms until a large area of the organs is occupied by metastatic cancer cells. Breast cancer multimodal imaging is attractive since it integrates advantages from several modalities, enabling more accurate cancer detection. Glycoprotein CD44 is overexpressed on most breast cancer cells and is the primary cell surface receptor for hyaluronan (HA). To facilitate breast cancer diagnosis, we report an indocyanine green (ICG) and HA conjugated iron oxide nanoparticle (NP-ICG-HA), which enabled active targeting to breast cancer by HA-CD44 interaction and detected metastasis with magnetic particle imaging (MPI) and near-infrared fluorescence imaging (NIR-FI). When evaluated in a transgenic breast cancer mouse model, NP-ICG-HA enabled the detection of multiple breast tumors in MPI and NIR-FI, providing more comprehensive images and a diagnosis of breast cancer. Furthermore, NP-ICG-HAs were evaluated in a lung metastasis model. Upon NP-ICG-HA administration, MPI showed clear signals in the lungs, indicating the tumor sites. This is the first time that HA-based NPs have enabled MPI of cancer. NP-ICG-HAs are an attractive platform for noninvasive detection of primary breast cancer and lung metastasis.
Collapse
Affiliation(s)
- Chia-Wei Yang
- Department
of Chemistry, Michigan State University, East Lansing, Michigan 48824, United States
- Institute
for Quantitative Health Science and Engineering, Michigan State University, East Lansing, Michigan 48824, United States
| | - Kunli Liu
- Department
of Chemistry, Michigan State University, East Lansing, Michigan 48824, United States
- Institute
for Quantitative Health Science and Engineering, Michigan State University, East Lansing, Michigan 48824, United States
| | - Cheng-You Yao
- Institute
for Quantitative Health Science and Engineering, Michigan State University, East Lansing, Michigan 48824, United States
- Department
of Electrical and Computer Engineering, Michigan State University, East Lansing, Michigan 48824, United States
| | - Bo Li
- Institute
for Quantitative Health Science and Engineering, Michigan State University, East Lansing, Michigan 48824, United States
- Department
of Electrical and Computer Engineering, Michigan State University, East Lansing, Michigan 48824, United States
| | - Aniwat Juhong
- Institute
for Quantitative Health Science and Engineering, Michigan State University, East Lansing, Michigan 48824, United States
- Department
of Electrical and Computer Engineering, Michigan State University, East Lansing, Michigan 48824, United States
| | - A. K. M. Atique Ullah
- Department
of Chemistry, Michigan State University, East Lansing, Michigan 48824, United States
- Institute
for Quantitative Health Science and Engineering, Michigan State University, East Lansing, Michigan 48824, United States
| | - Harvey Bumpers
- Department
of Surgery, Michigan State University, East Lansing, Michigan 48824, United States
| | - Zhen Qiu
- Institute
for Quantitative Health Science and Engineering, Michigan State University, East Lansing, Michigan 48824, United States
- Department
of Electrical and Computer Engineering, Michigan State University, East Lansing, Michigan 48824, United States
- Department
of Biomedical Engineering, Michigan State
University, East Lansing, Michigan 48824, United States
| | - Xuefei Huang
- Department
of Chemistry, Michigan State University, East Lansing, Michigan 48824, United States
- Institute
for Quantitative Health Science and Engineering, Michigan State University, East Lansing, Michigan 48824, United States
- Department
of Biomedical Engineering, Michigan State
University, East Lansing, Michigan 48824, United States
| |
Collapse
|
3
|
Yang JK, Kwon H, Kim S. Recent advances in light-triggered cancer immunotherapy. J Mater Chem B 2024; 12:2650-2669. [PMID: 38353138 DOI: 10.1039/d3tb02842a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/14/2024]
Abstract
Light-triggered phototherapies, such as photodynamic therapy (PDT) and photothermal therapy (PTT), have shown strong therapeutic efficacy with minimal invasiveness and systemic toxicity, offering opportunities for tumor-specific therapies. Phototherapies not only induce direct tumor cell killing, but also trigger anti-tumor immune responses by releasing various immune-stimulating factors. In recent years, conventional phototherapies have been combined with cancer immunotherapy as synergistic therapeutic modalities to eradicate cancer by exploiting the innate and adaptive immunity. These combined photoimmunotherapies have demonstrated excellent therapeutic efficacy in preventing tumor recurrence and metastasis compared to phototherapy alone. This review covers recent advancements in combined photoimmunotherapy, including photoimmunotherapy (PIT), PDT-combined immunotherapy, and PTT-combined immunotherapy, along with their underlying anti-tumor immune response mechanisms. In addition, the challenges and future research directions for light-triggered cancer immunotherapy are discussed.
Collapse
Affiliation(s)
- Jin-Kyoung Yang
- Department of Chemical Engineering, Dong-eui University, Busan, 47340, Republic of Korea.
| | - Hayoon Kwon
- Chemical & Biological integrative Research Center, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea.
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Republic of Korea
| | - Sehoon Kim
- Chemical & Biological integrative Research Center, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea.
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Republic of Korea
| |
Collapse
|
4
|
Nakajima K, Ogawa M. Near-infrared photoimmunotherapy and anti-cancer immunity. Int Immunol 2024; 36:57-64. [PMID: 37843836 DOI: 10.1093/intimm/dxad042] [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: 04/13/2023] [Accepted: 10/14/2023] [Indexed: 10/17/2023] Open
Abstract
The activation of the anti-cancer immune system is an important strategy to control cancer. A new form of cancer phototherapy, near-infrared photoimmunotherapy (NIR-PIT), was approved for clinical use in 2020 and uses IRDye® 700DX (IR700)-conjugated antibodies and NIR light. After irradiation with NIR light, the antibody-IR700 conjugate forms water-insoluble aggregations on the plasma membrane of target cells. This aggregation causes lethal damage to the plasma membrane, and effectively leads to immunogenic cell death (ICD). Subsequently, ICD activates anti-cancer immune cells such as dendritic cells and cytotoxic T cells. Combination therapy with immune-checkpoint blockade has synergistically improved the anti-cancer effects of NIR-PIT. Additionally, NIR-PIT can eliminate immunosuppressive immune cells in light-irradiated tumors by using specific antibodies against regulatory T cells and myeloid-derived suppressor cells. In addition to cancer-cell-targeted NIR-PIT, such immune-cell-targeted NIR-PIT has shown promising results by activating the anti-cancer immune system. Furthermore, NIR-PIT can be used to manipulate the tumor microenvironment by eliminating only targeted cells in the tumor, and thus it also can be used to gain insight into immunity in basic research.
Collapse
Affiliation(s)
- Kohei Nakajima
- Laboratory of Bioanalysis and Molecular Imaging, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, Hokkaido 060-0812, Japan
| | - Mikako Ogawa
- Laboratory of Bioanalysis and Molecular Imaging, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, Hokkaido 060-0812, Japan
| |
Collapse
|
5
|
Matsuoka K, Yamada M, Fukatsu N, Goto K, Shimizu M, Kato A, Kato Y, Yukawa H, Baba Y, Sato M, Sato K. Contrast-enhanced ultrasound imaging for monitoring the efficacy of near-infrared photoimmunotherapy. EBioMedicine 2023; 95:104737. [PMID: 37558554 PMCID: PMC10505829 DOI: 10.1016/j.ebiom.2023.104737] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 07/13/2023] [Accepted: 07/17/2023] [Indexed: 08/11/2023] Open
Abstract
BACKGROUND Near-infrared photoimmunotherapy (NIR-PIT) is a promising cancer therapy combining NIR-light irradiation with an antibody and IR700DX, a light-sensitive substance, to destroy tumours. However, homogeneous irradiation is difficult because the light varies depending on the distance and tissue environment. Therefore, markers that indicate sufficient irradiation are necessary. Nanoparticles sized 10∼200 nm show enhanced permeation and retention within tumours, which is further enhanced via NIR-PIT (super enhanced permeability and retention, SUPR). We aimed to monitor the effectiveness of NIR-PIT by measuring SUPR. METHODS A xenograft mouse tumour model was established by inoculating human cancer cells in both buttocks of Balb/C-nu/nu mice, and NIR-PIT was performed on only one side. To evaluate SUPR, fluorescent signal examination was performed using QD800-fluorescent nanoparticles and NIR-fluorescent poly (d,l-lactide-co-glycolic acid) (NIR-PLGA) microparticles. Harmonic signals were evaluated using micro-bubbles of the contrast agent Sonazoid and contrast-enhanced ultrasound (CEUS) imaging. The correlation between SUPR immediately after treatment and NIR-PIT effectiveness on the day after treatment was evaluated. FINDINGS QD800 fluorescent signals persisted only in the treated tumours, and the intensity of remaining signals showed high positive correlation with the therapeutic effect. NIR-PLGA fluorescent signals and Sonazoid-derived harmonic signals remained for a longer time in the treated tumours than in the controls, and the kE value of the two-compartment model correlated with NIR-PIT effectiveness. INTERPRETATION SUPR measurement using Sonazoid and CEUS imaging could be easily adapted for clinical use as a therapeutic image-based biomarker for monitoring and confirming of NIR-PIT efficacy. FUNDING This research was supported by ARIM JAPAN of MEXT, the Program for Developing Next-generation Researchers (Japan Science and Technology Agency), KAKEN (18K15923, 21K07217) (JSPS), CREST (JPMJCR19H2, JST), and FOREST-Souhatsu (JST). Mochida Memorial Foundation for Medical and Pharmaceutical Research; Takeda Science Foundation; The Japan Health Foundation; and Princess Takamatsu Cancer Research Fund. Funders only provided financial support and had no role in the study design, data collection, data analysis, interpretation, and writing of the report.
Collapse
Affiliation(s)
- Kohei Matsuoka
- Division of Host Defense Sciences, Dept. of Integrated Health Sciences, Nagoya University Graduate School of Medicine, Japan
| | - Mizuki Yamada
- Division of Host Defense Sciences, Dept. of Integrated Health Sciences, Nagoya University Graduate School of Medicine, Japan
| | - Noriaki Fukatsu
- Nagoya University Institute for Advanced Research, Advanced Analytical and Diagnostic Imaging Center (AADIC)/Medical Engineering Unit (MEU), B3 Unit, Japan
| | - Kyoichi Goto
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Japan
| | - Misae Shimizu
- Nagoya University Institute for Advanced Research, Advanced Analytical and Diagnostic Imaging Center (AADIC)/Medical Engineering Unit (MEU), B3 Unit, Japan
| | - Ayako Kato
- Nagoya University Institute for Advanced Research, Advanced Analytical and Diagnostic Imaging Center (AADIC)/Medical Engineering Unit (MEU), B3 Unit, Japan
| | - Yoshimi Kato
- Nagoya University Institute for Advanced Research, Advanced Analytical and Diagnostic Imaging Center (AADIC)/Medical Engineering Unit (MEU), B3 Unit, Japan
| | - Hiroshi Yukawa
- Nagoya University Institute for Advanced Research, Advanced Analytical and Diagnostic Imaging Center (AADIC)/Medical Engineering Unit (MEU), B3 Unit, Japan; Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Japan; Institute of Nano-Life-Systems, Institutes of Innovation for Future Society, Nagoya University, Japan; National Institutes for Quantum Science and Technology, Institute for Quantum Life Science, Quantum Life and Medical Science, Japan; Development of Quantum-nano Cancer Photoimmunotherapy for Clinical Application of Refractory Cancer, Nagoya University, Japan
| | - Yoshinobu Baba
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Japan; Institute of Nano-Life-Systems, Institutes of Innovation for Future Society, Nagoya University, Japan; National Institutes for Quantum Science and Technology, Institute for Quantum Life Science, Quantum Life and Medical Science, Japan; Development of Quantum-nano Cancer Photoimmunotherapy for Clinical Application of Refractory Cancer, Nagoya University, Japan
| | - Mitsuo Sato
- Division of Host Defense Sciences, Dept. of Integrated Health Sciences, Nagoya University Graduate School of Medicine, Japan
| | - Kazuhide Sato
- Nagoya University Institute for Advanced Research, Advanced Analytical and Diagnostic Imaging Center (AADIC)/Medical Engineering Unit (MEU), B3 Unit, Japan; Institute of Nano-Life-Systems, Institutes of Innovation for Future Society, Nagoya University, Japan; Development of Quantum-nano Cancer Photoimmunotherapy for Clinical Application of Refractory Cancer, Nagoya University, Japan; Nagoya University Graduate School of Medicine, Japan; FOREST-Souhatsu, JST, Tokyo, Japan.
| |
Collapse
|
6
|
Ortega-Forte E, Rovira A, López-Corrales M, Hernández-García A, Ballester FJ, Izquierdo-García E, Jordà-Redondo M, Bosch M, Nonell S, Santana MD, Ruiz J, Marchán V, Gasser G. A near-infrared light-activatable Ru(ii)-coumarin photosensitizer active under hypoxic conditions. Chem Sci 2023; 14:7170-7184. [PMID: 37416722 PMCID: PMC10321499 DOI: 10.1039/d3sc01844j] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 06/08/2023] [Indexed: 07/08/2023] Open
Abstract
Photodynamic therapy (PDT) represents a promising approach for cancer treatment. However, the oxygen dependency of PDT to generate reactive oxygen species (ROS) hampers its therapeutic efficacy, especially against hypoxic solid tumors. In addition, some photosensitizers (PSs) have dark toxicity and are only activatable with short wavelengths such as blue or UV-light, which suffer from poor tissue penetration. Herein, we developed a novel hypoxia-active PS with operability in the near-infrared (NIR) region based on the conjugation of a cyclometalated Ru(ii) polypyridyl complex of the type [Ru(C^N)(N^N)2] to a NIR-emitting COUPY dye. The novel Ru(ii)-coumarin conjugate exhibits water-solubility, dark stability in biological media and high photostability along with advantageous luminescent properties that facilitate both bioimaging and phototherapy. Spectroscopic and photobiological studies revealed that this conjugate efficiently generates singlet oxygen and superoxide radical anions, thereby achieving high photoactivity toward cancer cells upon highly-penetrating 740 nm light irradiation even under hypoxic environments (2% O2). The induction of ROS-mediated cancer cell death upon low-energy wavelength irradiation along with the low dark toxicity exerted by this Ru(ii)-coumarin conjugate could circumvent tissue penetration issues while alleviating the hypoxia limitation of PDT. As such, this strategy could pave the way to the development of novel NIR- and hypoxia-active Ru(ii)-based theragnostic PSs fuelled by the conjugation of tunable, low molecular-weight COUPY fluorophores.
Collapse
Affiliation(s)
- Enrique Ortega-Forte
- Departamento de Química Inorgánica, Universidad de Murcia, Biomedical Research Institute of Murcia (IMIB-Arrixaca) E-30071 Murcia Spain
| | - Anna Rovira
- Departament de Química Inorgànica i Orgànica, Secció de Química Orgànica, Universitat de Barcelona (UB), Institut de Biomedicina de la Universitat de Barcelona (IBUB) Martí i Franquès 1-11 E-08028 Barcelona Spain
| | - Marta López-Corrales
- Departament de Química Inorgànica i Orgànica, Secció de Química Orgànica, Universitat de Barcelona (UB), Institut de Biomedicina de la Universitat de Barcelona (IBUB) Martí i Franquès 1-11 E-08028 Barcelona Spain
| | - Alba Hernández-García
- Departamento de Química Inorgánica, Universidad de Murcia, Biomedical Research Institute of Murcia (IMIB-Arrixaca) E-30071 Murcia Spain
| | - Francisco José Ballester
- Departamento de Química Inorgánica, Universidad de Murcia, Biomedical Research Institute of Murcia (IMIB-Arrixaca) E-30071 Murcia Spain
| | - Eduardo Izquierdo-García
- Departament de Química Inorgànica i Orgànica, Secció de Química Orgànica, Universitat de Barcelona (UB), Institut de Biomedicina de la Universitat de Barcelona (IBUB) Martí i Franquès 1-11 E-08028 Barcelona Spain
- Chimie ParisTech, PSL University, CNRS, Institute of Chemistry for Life and Health Sciences, Laboratory for Inorganic Chemical Biology F-75005 Paris France
| | - Mireia Jordà-Redondo
- Institut Químic de Sarrià, Universitat Ramon Llull Vía Augusta 390 E-08017 Barcelona Spain
| | - Manel Bosch
- Unitat de Microscòpia Òptica Avançada, Centres Científics i Tecnològics, Universitat de Barcelona Av. Diagonal 643 E-08028 Barcelona Spain
| | - Santi Nonell
- Institut Químic de Sarrià, Universitat Ramon Llull Vía Augusta 390 E-08017 Barcelona Spain
| | - María Dolores Santana
- Departamento de Química Inorgánica, Universidad de Murcia, Biomedical Research Institute of Murcia (IMIB-Arrixaca) E-30071 Murcia Spain
| | - José Ruiz
- Departamento de Química Inorgánica, Universidad de Murcia, Biomedical Research Institute of Murcia (IMIB-Arrixaca) E-30071 Murcia Spain
| | - Vicente Marchán
- Departament de Química Inorgànica i Orgànica, Secció de Química Orgànica, Universitat de Barcelona (UB), Institut de Biomedicina de la Universitat de Barcelona (IBUB) Martí i Franquès 1-11 E-08028 Barcelona Spain
| | - Gilles Gasser
- Chimie ParisTech, PSL University, CNRS, Institute of Chemistry for Life and Health Sciences, Laboratory for Inorganic Chemical Biology F-75005 Paris France
| |
Collapse
|
7
|
Yamada M, Matsuoka K, Sato M, Sato K. Recent Advances in Localized Immunomodulation Technology: Application of NIR-PIT toward Clinical Control of the Local Immune System. Pharmaceutics 2023; 15:pharmaceutics15020561. [PMID: 36839882 PMCID: PMC9967863 DOI: 10.3390/pharmaceutics15020561] [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: 12/12/2022] [Revised: 01/27/2023] [Accepted: 02/04/2023] [Indexed: 02/11/2023] Open
Abstract
Current immunotherapies aim to modulate the balance among different immune cell populations, thereby controlling immune reactions. However, they often cause immune overactivation or over-suppression, which makes them difficult to control. Thus, it would be ideal to manipulate immune cells at a local site without disturbing homeostasis elsewhere in the body. Recent technological developments have enabled the selective targeting of cells and tissues in the body. Photo-targeted specific cell therapy has recently emerged among these. Near-infrared photoimmunotherapy (NIR-PIT) has surfaced as a new modality for cancer treatment, which combines antibodies and a photoabsorber, IR700DX. NIR-PIT is in testing as an international phase III clinical trial for locoregional recurrent head and neck squamous cell carcinoma (HNSCC) patients (LUZERA-301, NCT03769506), with a fast-track designation by the United States Food and Drug Administration (US-FDA). In Japan, NIR-PIT for patients with recurrent head and neck cancer was conditionally approved in 2020. Although NIR-PIT is commonly used for cancer therapy, it could also be exploited to locally eliminate certain immune cells with antibodies for a specific immune cell marker. This strategy can be utilized for anti-allergic therapy. Herein, we discuss the recent technological advances in local immunomodulation technology. We introduce immunomodulation technology with NIR-PIT and demonstrate an example of the knockdown of regulatory T cells (Tregs) to enhance local anti-tumor immune reactions.
Collapse
Affiliation(s)
- Mizuki Yamada
- Division of Host Defense Sciences, Department of Integrated Health Sciences, Nagoya University Graduate School of Medicine, Nagoya 461-8673, Japan
| | - Kohei Matsuoka
- Division of Host Defense Sciences, Department of Integrated Health Sciences, Nagoya University Graduate School of Medicine, Nagoya 461-8673, Japan
| | - Mitsuo Sato
- Division of Host Defense Sciences, Department of Integrated Health Sciences, Nagoya University Graduate School of Medicine, Nagoya 461-8673, Japan
| | - Kazuhide Sato
- B3 Unit Frontier, Advanced Analytical and Diagnostic Imaging Center (AADIC)/Medical Engineering Unit (MEU), Nagoya University Institute for Advanced Research, Nagoya 466-8550, Japan
- FOREST-Souhatsu, CREST, JST, Tokyo 102-0076, Japan
- Graduate School of Medicine, Nagoya University, Nagoya 466-8550, Japan
- Correspondence: ; Tel.: +81-052-744-2167; Fax: +81-052-744-2176
| |
Collapse
|
8
|
Mohiuddin TM, Zhang C, Sheng W, Al-Rawe M, Zeppernick F, Meinhold-Heerlein I, Hussain AF. Near Infrared Photoimmunotherapy: A Review of Recent Progress and Their Target Molecules for Cancer Therapy. Int J Mol Sci 2023; 24:2655. [PMID: 36768976 PMCID: PMC9916513 DOI: 10.3390/ijms24032655] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 01/26/2023] [Accepted: 01/28/2023] [Indexed: 02/01/2023] Open
Abstract
Near infrared photoimmunotherapy (NIR-PIT) is a newly developed molecular targeted cancer treatment, which selectively kills cancer cells or immune-regulatory cells and induces therapeutic host immune responses by administrating a cancer targeting moiety conjugated with IRdye700. The local exposure to near-infrared (NIR) light causes a photo-induced ligand release reaction, which causes damage to the target cell, resulting in immunogenic cell death (ICD) with little or no side effect to the surrounding normal cells. Moreover, NIR-PIT can generate an immune response in distant metastases and inhibit further cancer attack by combing cancer cells targeting NIR-PIT and immune regulatory cells targeting NIR-PIT or other cancer treatment modalities. Several recent improvements in NIR-PIT have been explored such as catheter-driven NIR light delivery, real-time monitoring of cancer, and the development of new target molecule, leading to NIR-PIT being considered as a promising cancer therapy. In this review, we discuss the progress of NIR-PIT, their mechanism and design strategies for cancer treatment. Furthermore, the overall possible targeting molecules for NIR-PIT with their application for cancer treatment are briefly summarised.
Collapse
|
9
|
Nishino H, Turner MA, Amirfakhri S, Lwin TM, Hosseini M, Singer BB, Hoffman RM, Bouvet M. Proof of Principle of Combining Fluorescence-Guided Surgery with Photoimmunotherapy to Improve the Outcome of Pancreatic Cancer Therapy in an Orthotopic Mouse Model. Ann Surg Oncol 2023; 30:618-625. [PMID: 36057899 PMCID: PMC9726788 DOI: 10.1245/s10434-022-12466-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 08/08/2022] [Indexed: 12/14/2022]
Abstract
BACKGROUND Pancreatic cancer is a recalcitrant disease in which R0 resection is often not achieved owing to difficulty in visualization of the tumor margins and proximity of adjacent vessels. To improve outcomes, we have developed fluorescence-guided surgery (FGS) and photoimmunotherapy (PIT) using a fluorescent tumor-specific antibody. METHODS Nude mice received surgical orthotopic implantation (SOI) of the human pancreatic cancer cell line BxPC-3 expressing green fluorescent protein. An anti-carcinoembryonic antigen-related cell adhesion molecule (CEACAM) monoclonal antibody (6G5j) was conjugated to the 700-nm fluorescent dye IR700DyeDX (6G5j-IR700DX). Three weeks after SOI, 16 mice received 50 μg 6G5j-IR700DX via the tail vein 24 h before surgery and were randomized to two groups: FGS-only (n = 8) and FGS + PIT (n = 8). All tumors were imaged with the Pearl Trilogy imaging system and resected under the guidance of the FLARE imaging system. The FGS + PIT group received PIT of the post-surgical bed at an intensity of 150 mW/cm2 for 30 min. Mice were sacrificed 4 weeks after initial surgery, and tumors were imaged with a Dino-Lite digital microscope, excised, and weighed. RESULTS The 6G5j-IR700DX dye illuminated the orthotopic pancreatic tumors for FGS and PIT. The metastatic recurrence rate was 100.0% for FGS-only and 25.0% for FGS + PIT (p = 0.007). The average total recurrent tumor weight was 2370.3 ± 1907.8 mg for FGS-only and 705.5 ± 1200.0 mg for FGS + PIT (p = 0.039). CONCLUSIONS FGS and adjuvant PIT can be combined by using a single antibody-fluorophore conjugate to significantly reduce the frequency of pancreatic cancer recurrence.
Collapse
Affiliation(s)
- Hiroto Nishino
- Department of Surgery, UCSD Moores Cancer Center, University of California San Diego, San Diego, CA USA ,Department of Surgery, VA San Diego Healthcare System, San Diego, CA USA ,Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Michael A. Turner
- Department of Surgery, UCSD Moores Cancer Center, University of California San Diego, San Diego, CA USA ,Department of Surgery, VA San Diego Healthcare System, San Diego, CA USA
| | - Siamak Amirfakhri
- Department of Surgery, UCSD Moores Cancer Center, University of California San Diego, San Diego, CA USA ,Department of Surgery, VA San Diego Healthcare System, San Diego, CA USA
| | - Thinzar M. Lwin
- Department of Surgery, UCSD Moores Cancer Center, University of California San Diego, San Diego, CA USA ,Department of Surgical Oncology, Dana Farber Cancer Center, Boston, MA USA
| | - Mojgan Hosseini
- Department of Pathology, University of California San Diego, San Diego, CA USA
| | - Bernhard B. Singer
- Medical Faculty, Institute of Anatomy, University of Duisburg-Essen, Essen, Germany
| | - Robert M. Hoffman
- Department of Surgery, UCSD Moores Cancer Center, University of California San Diego, San Diego, CA USA ,AntiCancer, Inc., San Diego, CA USA
| | - Michael Bouvet
- Department of Surgery, UCSD Moores Cancer Center, University of California San Diego, San Diego, CA USA ,Department of Surgery, VA San Diego Healthcare System, San Diego, CA USA
| |
Collapse
|
10
|
Wei D, Qi J, Hamblin MR, Wen X, Jiang X, Yang H. Near-infrared photoimmunotherapy: design and potential applications for cancer treatment and beyond. Am J Cancer Res 2022; 12:7108-7131. [PMID: 36276636 PMCID: PMC9576624 DOI: 10.7150/thno.74820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 09/28/2022] [Indexed: 11/22/2022] Open
Abstract
Near-infrared photoimmunotherapy (NIR-PIT) is a newly developed cancer treatment modality based on a target-specific photosensitizer conjugate (TSPC) composed of an NIR phthalocyanine photosensitizer and an antigen-specific recognition system. NIR-PIT has predominantly been used for targeted therapy of tumors via local irradiation with NIR light, following binding of TSPC to antigen-expressing cells. Physical stress-induced membrane damage is thought to be a major mechanism underlying NIR-PIT-triggered photokilling. Notably, NIR-PIT can rapidly induce immunogenic cell death and activate the adaptive immune response, thereby enabling its combination with immune checkpoint inhibitors. Furthermore, NIR-PIT-triggered “super-enhanced permeability and retention” effects can enhance drug delivery into tumors. Supported by its potential efficacy and safety, NIR-PIT is a rapidly developing therapeutic option for various cancers. Hence, this review seeks to provide an update on the (i) broad range of target molecules suitable for NIR-PIT, (ii) various types of receptor-selective ligands for designing the TSPC “magic bullet,” (iii) NIR light parameters, and (iv) strategies for enhancing the efficacy of NIR-PIT. Moreover, we review the potential application of NIR-PIT, including the specific design and efficacy in 19 different cancer types, and its clinical studies. Finally, we summarize possible NIR-PIT applications in noncancerous conditions, including infection, pain, itching, metabolic disease, autoimmune disease, and tissue engineering.
Collapse
Affiliation(s)
- Danfeng Wei
- Department of Dermatology and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China.,Laboratory of Dermatology, Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease-related Molecular Network West China Hospital, Sichuan University, Chengdu 610041, China.,NHC Key Lab of Transplant Engineering and Immunology, Organ Transplant Center, West China Hospital, Sichuan University, Chengdu, Chengdu 610041, China
| | - Jinxin Qi
- Department of Dermatology and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China.,Laboratory of Dermatology, Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease-related Molecular Network West China Hospital, Sichuan University, Chengdu 610041, China
| | - Michael R Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein 2028, South Africa
| | - Xiang Wen
- Department of Dermatology and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xian Jiang
- Department of Dermatology and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China.,Laboratory of Dermatology, Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease-related Molecular Network West China Hospital, Sichuan University, Chengdu 610041, China
| | - Hao Yang
- NHC Key Lab of Transplant Engineering and Immunology, Organ Transplant Center, West China Hospital, Sichuan University, Chengdu, Chengdu 610041, China.,Sichuan Provincial Engineering Laboratory of Pathology in Clinical Application, West China Hospital, Sichuan University
| |
Collapse
|
11
|
Takahashi K, Yasui H, Taki S, Shimizu M, Koike C, Taki K, Yukawa H, Baba Y, Kobayashi H, Sato K. Near-infrared-induced drug release from antibody-drug double conjugates exerts a cytotoxic photo-bystander effect. Bioeng Transl Med 2022; 7:e10388. [PMID: 36176626 PMCID: PMC9471993 DOI: 10.1002/btm2.10388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 05/21/2022] [Accepted: 07/16/2022] [Indexed: 11/23/2022] Open
Abstract
Ideal cancer treatments specifically target and eradicate tumor cells without affecting healthy cells. Therefore, antibody-based therapies that specifically target cancer antigens can be considered ideal cancer therapies. Antibodies linked with small-molecule drugs (i.e., antibody-drug conjugates [ADCs]) are widely used in clinics as antibody-based therapeutics. However, because tumors express antigens heterogeneously, greater target specificity and stable binding of noncleavable linkers in ADCs limit their antitumor effects. To overcome this problem, strategies, including decreasing the binding strength, conjugating more drugs, and targeting tumor stroma, have been applied, albeit with limited success. Thus, further technological advancements are required to remotely control the ADCs. Here, we described a drug that is photo-releasable from an ADC created via simple double conjugation and its antitumor effects both on target and nontarget tumor cells. Specifically, noncleavable T-DM1 was conjugated with IR700DX to produce T-DM1-IR700. Although T-DM1-IR700 itself is noncleavable, with NIR-light irradiation, it can release DM1-derivatives which elicited antitumor effect in vitro mixed culture and in vivo mixed tumor model which are mimicking heterogeneous tumor-antigen expression same as real clinical tumors. This cytotoxic photo-bystander effect occurred in various types mixed cultures in vitro, and changing antibodies also exerted photo-bystander effects, suggesting that this technology can be used for targeting various specific cancer antigens. These findings can potentially aid the development of strategies to address challenges associated with tumor expression of heterogeneous antigen.
Collapse
Affiliation(s)
- Kazuomi Takahashi
- Department of Respiratory MedicineNagoya University Graduate School of MedicineShowa‐kuNagoyaJapan
| | - Hirotoshi Yasui
- Department of Respiratory MedicineNagoya University Graduate School of MedicineShowa‐kuNagoyaJapan
| | - Shunichi Taki
- Department of Respiratory MedicineNagoya University Graduate School of MedicineShowa‐kuNagoyaJapan
| | - Misae Shimizu
- Nagoya University Institute for Advanced ResearchAdvanced Analytical and Diagnostic Imaging Center (AADIC)/Medical Engineering Unit (MEU), B3 UnitShowa‐kuNagoyaJapan
| | - Chiaki Koike
- Nagoya University Institute for Advanced ResearchAdvanced Analytical and Diagnostic Imaging Center (AADIC)/Medical Engineering Unit (MEU), B3 UnitShowa‐kuNagoyaJapan
| | - Kentaro Taki
- Division for Medical Research EngineeringNagoya University Graduate School of MedicineShowa‐kuNagoyaJapan
| | - Hiroshi Yukawa
- Nagoya University Institute for Advanced ResearchAdvanced Analytical and Diagnostic Imaging Center (AADIC)/Medical Engineering Unit (MEU), B3 UnitShowa‐kuNagoyaJapan
- Institute of Nano‐Life‐Systems, Institutes of Innovation for Future SocietyNagoya UniversityNagoyaJapan
- Department of Biomolecular EngineeringNagoya University Graduate School of EngineeringNagoyaJapan
| | - Yoshinobu Baba
- Institute of Nano‐Life‐Systems, Institutes of Innovation for Future SocietyNagoya UniversityNagoyaJapan
- Department of Biomolecular EngineeringNagoya University Graduate School of EngineeringNagoyaJapan
| | - Hisataka Kobayashi
- Molecular Imaging ProgramNational Cancer Institute, National Institutes of HealthBethesdaMarylandUSA
| | - Kazuhide Sato
- Department of Respiratory MedicineNagoya University Graduate School of MedicineShowa‐kuNagoyaJapan
- Nagoya University Institute for Advanced ResearchAdvanced Analytical and Diagnostic Imaging Center (AADIC)/Medical Engineering Unit (MEU), B3 UnitShowa‐kuNagoyaJapan
- Institute of Nano‐Life‐Systems, Institutes of Innovation for Future SocietyNagoya UniversityNagoyaJapan
- FOREST‐Souhatsu, CREST, JSTChiyoda‐kuTokyoJapan
- Nagoya University Institute for Advanced Research, S‐YLCJapan
| |
Collapse
|
12
|
Near-Infrared Photoimmunotherapy for Thoracic Cancers: A Translational Perspective. Biomedicines 2022; 10:biomedicines10071662. [PMID: 35884975 PMCID: PMC9312913 DOI: 10.3390/biomedicines10071662] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 06/23/2022] [Accepted: 07/07/2022] [Indexed: 12/18/2022] Open
Abstract
The conventional treatment of thoracic tumors includes surgery, anticancer drugs, radiation, and cancer immunotherapy. Light therapy for thoracic tumors has long been used as an alternative; conventional light therapy also called photodynamic therapy (PDT) has been used mainly for early-stage lung cancer. Recently, near-infrared photoimmunotherapy (NIR-PIT), which is a completely different concept from conventional PDT, has been developed and approved in Japan for the treatment of recurrent and previously treated head and neck cancer because of its specificity and effectiveness. NIR-PIT can apply to any target by changing to different antigens. In recent years, it has become clear that various specific and promising targets are highly expressed in thoracic tumors. In combination with these various specific targets, NIR-PIT is expected to be an ideal therapeutic approach for thoracic tumors. Additionally, techniques are being developed to further develop NIR-PIT for clinical practice. In this review, NIR-PIT is introduced, and its potential therapeutic applications for thoracic cancers are described.
Collapse
|
13
|
Fukushima H, Turkbey B, Pinto PA, Furusawa A, Choyke PL, Kobayashi H. Near-Infrared Photoimmunotherapy (NIR-PIT) in Urologic Cancers. Cancers (Basel) 2022; 14:2996. [PMID: 35740662 PMCID: PMC9221010 DOI: 10.3390/cancers14122996] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 06/12/2022] [Accepted: 06/14/2022] [Indexed: 11/16/2022] Open
Abstract
Near-infrared photoimmunotherapy (NIR-PIT) is a novel molecularly-targeted therapy that selectively kills cancer cells by systemically injecting an antibody-photoabsorber conjugate (APC) that binds to cancer cells, followed by the application of NIR light that drives photochemical transformations of the APC. APCs are synthesized by selecting a monoclonal antibody that binds to a receptor on a cancer cell and conjugating it to IRDye700DX silica-phthalocyanine dye. Approximately 24 h after APC administration, NIR light is delivered to the tumor, resulting in nearly-immediate necrotic cell death of cancer cells while causing no harm to normal tissues. In addition, NIR-PIT induces a strong immunologic effect, activating anti-cancer immunity that can be further boosted when combined with either immune checkpoint inhibitors or immune suppressive cell-targeted (e.g., regulatory T cells) NIR-PIT. Currently, a global phase III study of NIR-PIT in recurrent head and neck squamous cell carcinoma is ongoing. The first APC and NIR laser systems were approved for clinical use in September 2020 in Japan. In the near future, the clinical applications of NIR-PIT will expand to other cancers, including urologic cancers. In this review, we provide an overview of NIR-PIT and its possible applications in urologic cancers.
Collapse
Affiliation(s)
- Hiroshi Fukushima
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute (NIH), Bethesda, MD 20892, USA; (H.F.); (B.T.); (A.F.); (P.L.C.)
| | - Baris Turkbey
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute (NIH), Bethesda, MD 20892, USA; (H.F.); (B.T.); (A.F.); (P.L.C.)
| | - Peter A. Pinto
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute (NIH), Bethesda, MD 20892, USA;
| | - Aki Furusawa
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute (NIH), Bethesda, MD 20892, USA; (H.F.); (B.T.); (A.F.); (P.L.C.)
| | - Peter L. Choyke
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute (NIH), Bethesda, MD 20892, USA; (H.F.); (B.T.); (A.F.); (P.L.C.)
| | - Hisataka Kobayashi
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute (NIH), Bethesda, MD 20892, USA; (H.F.); (B.T.); (A.F.); (P.L.C.)
| |
Collapse
|
14
|
Furumoto H, Kato T, Wakiyama H, Furusawa A, Choyke PL, Kobayashi H. Endoscopic Applications of Near-Infrared Photoimmunotherapy (NIR-PIT) in Cancers of the Digestive and Respiratory Tracts. Biomedicines 2022; 10:846. [PMID: 35453596 PMCID: PMC9027987 DOI: 10.3390/biomedicines10040846] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 03/24/2022] [Accepted: 03/25/2022] [Indexed: 12/11/2022] Open
Abstract
Near-infrared photoimmunotherapy (NIR-PIT) is a newly developed and promising therapy that specifically destroys target cells by irradiating antibody-photo-absorber conjugates (APCs) with NIR light. APCs bind to target molecules on the cell surface, and when exposed to NIR light, cause disruption of the cell membrane due to the ligand release reaction and dye aggregation. This leads to rapid cell swelling, blebbing, and rupture, which leads to immunogenic cell death (ICD). ICD activates host antitumor immunity, which assists in killing still viable cancer cells in the treated lesion but is also capable of producing responses in untreated lesions. In September 2020, an APC and laser system were conditionally approved for clinical use in unresectable advanced head and neck cancer in Japan, and are now routine in appropriate patients. However, most tumors have been relatively accessible in the oral cavity or neck. Endoscopes offer the opportunity to deliver light deeper within hollow organs of the body. In recent years, the application of endoscopic therapy as an alternative to surgery for the treatment of cancer has expanded, providing significant benefits to inoperable patients. In this review, we will discuss the potential applications of endoscopic NIR-PIT, especially in thoracic and gastrointestinal cancers.
Collapse
Affiliation(s)
| | | | | | | | | | - Hisataka Kobayashi
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; (H.F.); (T.K.); (H.W.); (A.F.); (P.L.C.)
| |
Collapse
|
15
|
Mussini A, Uriati E, Bianchini P, Diaspro A, Cavanna L, Abbruzzetti S, Viappiani C. Targeted photoimmunotherapy for cancer. Biomol Concepts 2022; 13:126-147. [PMID: 35304984 DOI: 10.1515/bmc-2022-0010] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 02/24/2022] [Indexed: 12/12/2022] Open
Abstract
Photodynamic therapy (PDT) is a clinically approved procedure that can exert a curative action against malignant cells. The treatment implies the administration of a photoactive molecular species that, upon absorption of visible or near infrared light, sensitizes the formation of reactive oxygen species. These species are cytotoxic and lead to tumor cell death, damage vasculature, and induce inflammation. Clinical investigations demonstrated that PDT is curative and does not compromise other treatment options. One of the major limitations of the original method was the low selectivity of the photoactive compounds for malignant over healthy tissues. The development of conjugates with antibodies has endowed photosensitizing molecules with targeting capability, so that the compounds are delivered with unprecedented precision to the site of action. Given their fluorescence emission capability, these supramolecular species are intrinsically theranostic agents.
Collapse
Affiliation(s)
- Andrea Mussini
- Dipartimento di Scienze Matematiche, Fisiche e Informatiche, Università Degli Studi di Parma, Parma, Italy
| | - Eleonora Uriati
- Dipartimento di Scienze Matematiche, Fisiche e Informatiche, Università Degli Studi di Parma, Parma, Italy.,Department of Nanophysics, Nanoscopy, Istituto Italiano di Tecnologia, Genova, Italy
| | - Paolo Bianchini
- Dipartimento di Scienze Matematiche, Fisiche e Informatiche, Università Degli Studi di Parma, Parma, Italy.,Department of Nanophysics, Nanoscopy, Istituto Italiano di Tecnologia, Genova, Italy.,DIFILAB, Dipartimento di Fisica, Università Degli Studi di Genova, Genova, Italy
| | - Alberto Diaspro
- Department of Nanophysics, Nanoscopy, Istituto Italiano di Tecnologia, Genova, Italy.,DIFILAB, Dipartimento di Fisica, Università Degli Studi di Genova, Genova, Italy
| | - Luigi Cavanna
- Dipartimento di Oncologia-Ematologia, Azienda USL di Piacenza, Piacenza, Italy
| | - Stefania Abbruzzetti
- Dipartimento di Scienze Matematiche, Fisiche e Informatiche, Università Degli Studi di Parma, Parma, Italy
| | - Cristiano Viappiani
- Dipartimento di Scienze Matematiche, Fisiche e Informatiche, Università Degli Studi di Parma, Parma, Italy
| |
Collapse
|
16
|
Zhang X, Nakajima T, Mizoi K, Tsushima Y, Ogihara T. Imaging modalities for monitoring acute therapeutic effects after near-infrared photoimmunotherapy in vivo. JOURNAL OF BIOPHOTONICS 2022; 15:e202100266. [PMID: 34783185 DOI: 10.1002/jbio.202100266] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 10/11/2021] [Accepted: 10/17/2021] [Indexed: 06/13/2023]
Abstract
Near-infrared photoimmunotherapy (NIR-PIT) induces immediate cell death after irradiation with near-infrared (NIR) light. Acute therapeutic effects caused by NIR-PIT before the change of tumor size is essential to be monitored by imaging modalities. We summarized and compared the imaging modalities for evaluating acute therapeutic effects after NIR-PIT, and aimed to provide a better understanding of advantages and disadvantages of each modality for evaluation in clinical applications. Fluorescence imaging and fluorescence lifetime, with high resolution, remains high accumulation of fluorescence dyes in the normal organs. High resolution and noninvasiveness are the major advantages of magnetic resonance imaging, while 18 F-fluorodeoxyglucose positron emission tomography provides information about the glucose metabolism. Optical coherence tomography provided more information about the blood vessels. Thus, all of the imaging modalities play an important role in evaluating acute therapeutic effects after NIR-PIT. Clinicians should choose suitable modality according to specific purpose and conditions in clinical application.
Collapse
Affiliation(s)
- Xieyi Zhang
- Laboratory of Biopharmaceutics, Department of Pharmacology, Faculty of Pharmacy, Takasaki University of Health and Welfare, Takasaki, Gunma, Japan
| | - Takahito Nakajima
- Department of Diagnostic and Interventional Radiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
- Department of Diagnostic Radiology and Nuclear Medicine, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Kenta Mizoi
- Laboratory of Biopharmaceutics, Department of Pharmacology, Faculty of Pharmacy, Takasaki University of Health and Welfare, Takasaki, Gunma, Japan
| | - Yoshito Tsushima
- Department of Diagnostic Radiology and Nuclear Medicine, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
- Research Program for Diagnostic and Molecular Imaging, Division of Integrated Oncology Research, Gunma University Initiative for Advanced Research (GIAR), Maebashi, Gunma, Japan
| | - Takuo Ogihara
- Laboratory of Biopharmaceutics, Department of Pharmacology, Faculty of Pharmacy, Takasaki University of Health and Welfare, Takasaki, Gunma, Japan
- Laboratory of Clinical Pharmacokinetics, Graduate School of Pharmaceutical Sciences, Takasaki University of Health and Welfare, Takasaki, Gunma, Japan
| |
Collapse
|
17
|
Sato K. Bioluminescence Imaging for Evaluation of Antitumor Effect In Vitro and In Vivo in Mice Xenografted Tumor Models. Methods Mol Biol 2022; 2524:307-315. [PMID: 35821482 DOI: 10.1007/978-1-0716-2453-1_24] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Bioluminescence imaging (BLI) enables real-time imaging in vitro and in vivo; it is widely used in laboratories. In vitro, the bioluminescence is commonly used as a reporter for the transfection. In vivo, BLI is employed to evaluate cell expression, migration, and proliferation inside animal bodies and visualize specific cells in various fields. Here, this chapter introduces BLI protocols for assaying the efficacy of in vivo BLI in monitoring cancer treatment using mice orthotopic models.
Collapse
Affiliation(s)
- Kazuhide Sato
- Respiratory Medicine, Nagoya University Graduate School of Medicine, Nagoya, Japan.
- Nagoya University Institute for Advanced Research, B3-Unit, Advanced Analytical and Diagnostic Imaging Center (AADIC)/Medical Engineering Unit (MEU), S-YLC, Nagoya, Japan.
- FOREST-Souhatsu, CREST, JST, Nagoya, Japan.
| |
Collapse
|
18
|
Privitera L, Paraboschi I, Cross K, Giuliani S. Above and Beyond Robotic Surgery and 3D Modelling in Paediatric Cancer Surgery. Front Pediatr 2021; 9:777840. [PMID: 34988038 PMCID: PMC8721224 DOI: 10.3389/fped.2021.777840] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 11/17/2021] [Indexed: 12/15/2022] Open
Abstract
Although the survival rates for children's cancers have more than doubled in the last few decades, the surgical practise has not significantly changed. Among the most recent innovations introduced in the clinic, robotic surgery and augmented reality are two of the most promising, even if they are not widespread. The increased flexibility of the motion, the magnification of the surgical field and the tremor reduction provided by robotic surgery have been beneficial to perform complex oncological procedures in children. Besides, augmented reality has been proven helpful in planning for tumour removal, facilitating early discrimination between cancer and healthy organs. Nowadays, research in the field of surgical oncology is moving fast, and new technologies and innovations wich will help to shape a new way to perform cancer surgery. Paediatric surgeons need to be ready to adopt these novel devices and intraoperative techniques to allow more radical tumour resections with fewer complications. This review aims to present the mechanism of action and indications of several novel technologies such as optical imaging surgery, high definition cameras, and intraoperative loco-regional treatments. We hope this will enhance early adoption and more research on how to employ technology for the benefit of children.
Collapse
Affiliation(s)
- Laura Privitera
- Wellcome/Engineering and Physical Sciences Research Council Centre for Interventional & Surgical Sciences, University College London, London, United Kingdom
- Developmental Biology and Cancer Department, University College London Great Ormond Street Institute of Child Health, London, United Kingdom
| | - Irene Paraboschi
- Wellcome/Engineering and Physical Sciences Research Council Centre for Interventional & Surgical Sciences, University College London, London, United Kingdom
- Developmental Biology and Cancer Department, University College London Great Ormond Street Institute of Child Health, London, United Kingdom
| | - Kate Cross
- Department of Specialist Neonatal and Paediatric Surgery, Great Ormond Street Hospital for Children NHS Foundation Trust, London, United Kingdom
| | - Stefano Giuliani
- Wellcome/Engineering and Physical Sciences Research Council Centre for Interventional & Surgical Sciences, University College London, London, United Kingdom
- Developmental Biology and Cancer Department, University College London Great Ormond Street Institute of Child Health, London, United Kingdom
- Department of Specialist Neonatal and Paediatric Surgery, Great Ormond Street Hospital for Children NHS Foundation Trust, London, United Kingdom
| |
Collapse
|
19
|
Russo I, Fagotto L, Colombo A, Sartor E, Luisetto R, Alaibac M. Near-infrared photoimmunotherapy for the treatment of skin disorders. Expert Opin Biol Ther 2021; 22:509-517. [PMID: 34860146 DOI: 10.1080/14712598.2022.2012147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
INTRODUCTION Near-Infrared Photoimmunotherapy (NIR-PIT) is a novel molecularly targeted phototherapy. This technique is based on a conjugate of a near-infrared photo-inducible molecule (antibody-photon absorber conjugate, APC) and a monoclonal antibody that targets a tumor-specific antigen. To date, this novel approach has been successfully applied to several types of cancer. AREAS COVERED The authors discuss the possible use of NIR-PIT for the management of skin diseases, with special attention given to squamous cell carcinomas, advanced melanomas, and primary cutaneous lymphomas. EXPERT OPINION NIR-PIT may be an attractive strategy for the treatment of skin disorders. The main advantage of NIR-PIT therapy is its low toxicity to healthy tissues. Cutaneous lymphocyte antigen is a potential molecular target for NIR-PIT for both cutaneous T-cell lymphomas and inflammatory skin disorders.
Collapse
Affiliation(s)
- Irene Russo
- Unit of Dermatology, University of Padua, Padova, Italy
| | - Laura Fagotto
- Unit of Dermatology, University of Padua, Padova, Italy
| | - Anna Colombo
- Unit of Dermatology, University of Padua, Padova, Italy
| | - Emma Sartor
- Unit of Dermatology, University of Padua, Padova, Italy
| | - Roberto Luisetto
- DISCOG-Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
| | - Mauro Alaibac
- Unit of Dermatology, University of Padua, Padova, Italy
| |
Collapse
|
20
|
Xue G, Wang Z, Zheng N, Fang J, Mao C, Li X, Jin G, Ming X, Lu Y. Elimination of acquired resistance to PD-1 blockade via the concurrent depletion of tumour cells and immunosuppressive cells. Nat Biomed Eng 2021; 5:1306-1319. [PMID: 34725506 PMCID: PMC8595849 DOI: 10.1038/s41551-021-00799-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 08/26/2021] [Indexed: 12/12/2022]
Abstract
Antigen release resulting from the death of tumour cells induced by chemotherapies and targeted therapies can augment the antitumour responses induced by immune checkpoint blockade (ICB). However, tumours responding to ICB therapies often become resistant to them. Here we show that the specific targeting of tumour cells promotes the growth of tumour-cell variants that are resistant to ICB, and that the acquired resistance can be overcome via the concurrent depletion of tumour cells and of major types of immunosuppressive cell via a monoclonal antibody binding the enzyme CD73, which we identified as highly expressed on tumour cells and on regulatory T cells, myeloid-derived suppressor cells and tumour-associated macrophages, but not on cytolytic T lymphocytes, natural killer cells and dendritic cells. In mice with murine tumours, the systemic administration of anti-PD1 antibodies and anti-CD73 antibodies conjugated to a near-infrared dye prevented near-infrared-irradiated tumours from acquiring resistance to ICB and resulted in the eradication of advanced tumours. The elimination of immunosuppressive cells may overcome acquired resistance to ICB across a range of tumour types and combination therapies.
Collapse
Affiliation(s)
- Gang Xue
- Comprehensive Cancer Center, Wake Forest Baptist Health, Winston-Salem, NC, USA, 27157; Department of Microbiology & Immunology, Wake Forest School of Medicine, Winston-Salem, NC, USA, 27101
| | - Ziyu Wang
- Comprehensive Cancer Center, Wake Forest Baptist Health, Winston-Salem, NC, USA, 27157; Department of Microbiology & Immunology, Wake Forest School of Medicine, Winston-Salem, NC, USA, 27101
| | - Ningbo Zheng
- Comprehensive Cancer Center, Wake Forest Baptist Health, Winston-Salem, NC, USA, 27157; Department of Microbiology & Immunology, Wake Forest School of Medicine, Winston-Salem, NC, USA, 27101
| | - Jing Fang
- Comprehensive Cancer Center, Wake Forest Baptist Health, Winston-Salem, NC, USA, 27157; Department of Microbiology & Immunology, Wake Forest School of Medicine, Winston-Salem, NC, USA, 27101
| | - Chengqiong Mao
- Comprehensive Cancer Center, Wake Forest Baptist Health, Winston-Salem, NC, USA, 27157; Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC, USA, 27157
| | - Xiaoyin Li
- Department of Mathematics and Statistics, St. Cloud State University, St Cloud, MN, USA, 56301
| | - Guangxu Jin
- Comprehensive Cancer Center, Wake Forest Baptist Health, Winston-Salem, NC, USA, 27157; Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC, USA, 27157
| | - Xin Ming
- Comprehensive Cancer Center, Wake Forest Baptist Health, Winston-Salem, NC, USA, 27157; Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC, USA, 27157
| | - Yong Lu
- Comprehensive Cancer Center, Wake Forest Baptist Health, Winston-Salem, NC, USA, 27157; Department of Microbiology & Immunology, Wake Forest School of Medicine, Winston-Salem, NC, USA, 27101,Corresponding authors, , Correspondence and requests for materials should be addressed to Y.L
| |
Collapse
|
21
|
Matsuoka K, Sato M, Sato K. Hurdles for the wide implementation of photoimmunotherapy. Immunotherapy 2021; 13:1427-1438. [PMID: 34693721 DOI: 10.2217/imt-2021-0241] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Near infrared photoimmunotherapy (NIR-PIT) is a molecularly targeted treatment for cancers achieved by injecting a conjugate of IRDye700DX® (IR700), a water-soluble silicon phthalocyanine derivative in the near infrared, and a monoclonal antibody that targets cancer cell antigens. NIR-PIT is a highly specific treatment with few side effects that results in rapid immunogenic cell death. Despite it being a very effective and innovative therapy, there are a few challenges preventing full implementation in clinical practice. These include the limits of near infrared light penetration, selection of targets, concerns about tumor lysis syndrome and drug costs. However, NIR-PIT has been approved by the regulatory authorities in Japan, allowing for exploration of how to mitigate challenges while maximizing the benefits of this treatment modality.
Collapse
Affiliation(s)
- Kohei Matsuoka
- Department of Integrated Health Sciences, Nagoya University Graduate School of Medicine, Nagoya, 461-8673, Japan
| | - Mitsuo Sato
- Department of Integrated Health Sciences, Nagoya University Graduate School of Medicine, Nagoya, 461-8673, Japan
| | - Kazuhide Sato
- Respiratory Medicine, Nagoya University Graduate School of Medicine, Nagoya, 464-0814, Japan.,Nagoya University Institute for Advanced Research, Advanced Analytical & Diagnostic Imaging Center (AADIC)/Medical Engineering Unit (MEU), B3 Unit, Nagoya, Japan.,FOREST-Souhatsu, CREST, JST, Tokyo, 102-8666, Japan.,Nagoya University Institute for Advanced Research, S-YLC, Nagoya, 464-8601, Japan
| |
Collapse
|
22
|
Kobayashi H, Choyke PL. Future applications of and prospects for near-IR photoimmunotherapy: benefits and differences compared with photodynamic and photothermal therapy. Immunotherapy 2021; 13:1305-1307. [PMID: 34514846 DOI: 10.2217/imt-2021-0216] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Affiliation(s)
- Hisataka Kobayashi
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Peter L Choyke
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| |
Collapse
|
23
|
Simultaneous exposure to intracellular and extracellular photosensitizers for the treatment of Staphylococcus aureus infections. Antimicrob Agents Chemother 2021; 65:e0091921. [PMID: 34516248 DOI: 10.1128/aac.00919-21] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Staphylococcus aureus is a serious threat to public health due to the rise of antibiotic resistance in this organism, which can prolong or exacerbate skin and soft tissue infections (SSTIs). Methicillin-resistant S. aureus is a Gram-positive bacterium and a leading cause of SSTIs. As such, many efforts are underway to develop therapies that target essential biological processes in S. aureus. Antimicrobial photodynamic therapy is effective alternative to antibiotics, therefore we developed an approach to simultaneously expose S. aureus to intracellular and extracellular photoactivators. A near infrared photosensitizer was conjugated to human monoclonal antibodies (mAbs) that target the S. aureus Isd heme acquisition proteins. Additionally, the compound VU0038882 was developed to increase photoactivatable porphyrins within the cell. Combinatorial PDT treatment of drug-resistant S. aureus exposed to VU0038882 and conjugated anti-Isd mAbs proved to be an effective antibacterial strategy in vitro and in a murine model of SSTIs.
Collapse
|
24
|
Wakiyama H, Kato T, Furusawa A, Choyke PL, Kobayashi H. Near infrared photoimmunotherapy of cancer; possible clinical applications. NANOPHOTONICS 2021; 10:3135-3151. [PMID: 36405499 PMCID: PMC9646249 DOI: 10.1515/nanoph-2021-0119] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Accepted: 04/15/2021] [Indexed: 05/07/2023]
Abstract
Near-infrared photoimmunotherapy (NIR-PIT) is a new cancer treatment that uses an antibody-photo-absorber conjugate (APC) composed of a targeting monoclonal antibody conjugated with a photoactivatable phthalocyanine-derivative dye, IRDye700DX (IR700). APCs injected into the body can bind to cancer cells where they are activated by local exposure to NIR light typically delivered by a NIR laser. NIR light alters the APC chemical conformation inducing damage to cancer cell membranes, resulting in necrotic cell death within minutes of light exposure. NIR-PIT selectivity kills cancer cells by immunogenic cell death (ICD) with minimal damage to adjacent normal cells thus, leading to rapid recovery by the patient. Moreover, since NIR-PIT induces ICD only on cancer cells, NIR-PIT initiates and activates antitumor host immunity that could be further enhanced when combined with immune checkpoint inhibition. NIR-PIT induces dramatic changes in the tumor vascularity causing the super-enhanced permeability and retention (SUPR) effect that dramatically enhances nanodrug delivery to the tumor bed. Currently, a worldwide Phase 3 study of NIR-PIT for recurrent or inoperable head and neck cancer patients is underway. In September 2020, the first APC and accompanying laser system were conditionally approved for clinical use in Japan. In this review, we introduce NIR-PIT and the SUPR effect and summarize possible applications of NIR-PIT in a variety of cancers.
Collapse
Affiliation(s)
- Hiroaki Wakiyama
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, 20892, USA
| | - Takuya Kato
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, 20892, USA
| | - Aki Furusawa
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, 20892, USA
| | - Peter L. Choyke
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, 20892, USA
| | - Hisataka Kobayashi
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, 20892, USA
| |
Collapse
|
25
|
Maruoka Y, Wakiyama H, Choyke PL, Kobayashi H. Near infrared photoimmunotherapy for cancers: A translational perspective. EBioMedicine 2021; 70:103501. [PMID: 34332294 PMCID: PMC8340111 DOI: 10.1016/j.ebiom.2021.103501] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Revised: 06/20/2021] [Accepted: 07/12/2021] [Indexed: 12/11/2022] Open
Abstract
Near-infrared photoimmunotherapy (NIR-PIT) is a newly-developed, highly-selective cancer treatment, which utilizes a monoclonal antibody conjugated to a photoabsorbing dye, IRDye700DX (IR700). The antibody conjugate is injected into the patient and accumulates in the tumour. Within 24 h of injection the tumour is exposed to NIR light which activates the conjugate and causes rapid, selective cancer cell death. A global phase III clinical trial of NIR-PIT in recurrent head and neck squamous cell cancer (HNSCC) patients is currently underway. Conditional clinical approval for NIR-PIT in recurrent HNSCC has been granted in Japan as of September 2020. Not only does NIR-PIT induce highly selective and immediate cancer cell killing, but it also stimulates highly active anti-tumour immunity. While monotherapy with NIR-PIT has proven effective it is likely that combinations with immune-checkpoint inhibitors or additional NIR-PIT targeting immune suppressive cells in the tumour microenvironment will further improve results. In this review, we discuss the translational aspects of NIR-PIT especially in HNSCC, and potential future applications.
Collapse
Affiliation(s)
- Yasuhiro Maruoka
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; Departments of Clinical Radiology, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Hiroaki Wakiyama
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Peter L Choyke
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Hisataka Kobayashi
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
26
|
Li F, Mao C, Yeh S, Sun Y, Xin J, Shi Q, Ming X. MRP1-targeted near infrared photoimmunotherapy for drug resistant small cell lung cancer. Int J Pharm 2021; 604:120760. [PMID: 34077781 DOI: 10.1016/j.ijpharm.2021.120760] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 05/07/2021] [Accepted: 05/27/2021] [Indexed: 12/11/2022]
Abstract
Small cell lung cancer (SCLC), one of the most aggressive cancers, has a high mortality rate and poor prognosis, and the clinical therapeutic outcomes of multidrug resistant SCLC are even worse. Multidrug resistance protein 1 (MRP1), one of the ATP-binding cassette (ABC) transporter proteins that cause decreased drug accumulation in cancer cells, is overexpressed in drug resistant SCLC cells and could be a promising target for treating the patients suffering from this illness. Near infrared photoimmunotherapy (NIR-PIT) is a newly developed approach for targeted cancer treatment which uses a conjugate of a monoclonal antibody and photoabosorber IR700 followed by NIR light irradiation to induce rapid cancer cell death. In the present study, an anti-MRP1 antibody (Mab) -IR700 conjugate (Mab-IR700) was synthesized, purified and used to treat chemoresistant SCLC H69AR cells that overexpressed MRP1, while non-MRP1-expressing H69 cells were used as a control. Then, the photokilling and tumor suppression effect were separately evaluated in H69AR cells both in vitro and in vivo. Higher cellular delivery of Mab-IR700 was detected in H69AR cells, whereas there was little uptake of IgG-IR700 in both H69 and H69AR cells. Due to the targeting activity of Mab, stronger photokilling effect was found both in H69AR cells and spheroids treated with Mab-IR700, while superior tumor suppression effect was also observed in the mice treated with Mab-IR700 and light illumination. Photoacoustic imaging results proved that oxygen was involved in NIR-PIT treatment, and TUNEL staining images showed the occurrence of cell apoptosis, which was also testified by HE staining. This research provides MRP1 as a novel target for PIT and presents a prospective way for treating drug resistant SCLC and, thus, should be further studied.
Collapse
Affiliation(s)
- Fang Li
- School of Pharmacy, Jiangsu Vocational College of Medicine, Yancheng 224005, China; Department of Cancer Biology and Comprehensive Cancer Center, Wake Forest University School of Medicine, Winston-Salem 27157, USA.
| | - Chengqiong Mao
- Department of Cancer Biology and Comprehensive Cancer Center, Wake Forest University School of Medicine, Winston-Salem 27157, USA
| | - Stacy Yeh
- Department of Cancer Biology and Comprehensive Cancer Center, Wake Forest University School of Medicine, Winston-Salem 27157, USA
| | - Yao Sun
- Department of Cancer Biology and Comprehensive Cancer Center, Wake Forest University School of Medicine, Winston-Salem 27157, USA
| | - Junbo Xin
- School of Pharmacy, Jiangsu Vocational College of Medicine, Yancheng 224005, China
| | - Qin Shi
- School of Pharmacy, Jiangsu Vocational College of Medicine, Yancheng 224005, China
| | - Xin Ming
- Department of Cancer Biology and Comprehensive Cancer Center, Wake Forest University School of Medicine, Winston-Salem 27157, USA.
| |
Collapse
|
27
|
Yasui H, Nishinaga Y, Taki S, Takahashi K, Isobe Y, Shimizu M, Koike C, Taki T, Sakamoto A, Katsumi K, Ishii K, Sato K. Near-infrared photoimmunotherapy targeting GPR87: Development of a humanised anti-GPR87 mAb and therapeutic efficacy on a lung cancer mouse model. EBioMedicine 2021; 67:103372. [PMID: 33993055 PMCID: PMC8138482 DOI: 10.1016/j.ebiom.2021.103372] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Revised: 03/25/2021] [Accepted: 04/16/2021] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND GPR87 is a G-protein receptor that is specifically expressed in tumour cells, such as lung cancer, and rarely expressed in normal cells. GPR87 is a promising target for cancer therapy, but its ligand is controversial. Near-infrared photoimmunotherapy (NIR-PIT) is a novel cancer therapy in which a photosensitiser, IRDye700DX (IR700), binds to antibodies and specifically destroys target cells by irradiating them with near-infrared-light. Here, we aimed to develop a NIR-PIT targeting GPR87. METHODS We evaluated the expression of GPR87 in resected specimens of lung cancer and malignant pleural mesothelioma (MPM) resected at Nagoya University Hospital using immunostaining. Humanised anti-GPR87 antibody (huGPR87) was generated by introducing CDRs from mouse anti-GPR87 antibody generated by standard hybridoma method. HuGPR87 was conjugated with IR700 and the therapeutic effect of NIR-PIT was evaluated in vitro and in vivo using lung cancer or MPM cell lines. FINDINGS Among the surgical specimens, 54% of lung cancer and 100% of MPM showed high expression of GPR87. It showed therapeutic effects on lung cancer and MPM cell lines in vitro, and showed therapeutic effects in multiple models in vivo. INTERPRETATION These results suggest that NIR-PIT targeting GPR87 is a promising therapeutic approach for the treatment of thoracic cancer. FUNDING This research was supported by the Program for Developing Next-generation Researchers (Japan Science and Technology Agency), KAKEN (18K15923, 21K07217, JSPS), FOREST-Souhatsu, CREST (JST).
Collapse
Affiliation(s)
- Hirotoshi Yasui
- Respiratory Medicine, Nagoya University Graduate School of Medicine, 65, Tsurumai-cho, Showa-ku, Nagoya 466-8560, Aichi, Japan
| | - Yuko Nishinaga
- Respiratory Medicine, Nagoya University Graduate School of Medicine, 65, Tsurumai-cho, Showa-ku, Nagoya 466-8560, Aichi, Japan
| | - Shunichi Taki
- Respiratory Medicine, Nagoya University Graduate School of Medicine, 65, Tsurumai-cho, Showa-ku, Nagoya 466-8560, Aichi, Japan
| | - Kazuomi Takahashi
- Respiratory Medicine, Nagoya University Graduate School of Medicine, 65, Tsurumai-cho, Showa-ku, Nagoya 466-8560, Aichi, Japan
| | - Yoshitaka Isobe
- Respiratory Medicine, Nagoya University Graduate School of Medicine, 65, Tsurumai-cho, Showa-ku, Nagoya 466-8560, Aichi, Japan
| | - Misae Shimizu
- Nagoya University Institute for Advanced Research, Advanced Analytical and Diagnostic Imaging Center (AADIC) / Medical Engineering Unit (MEU), B3 Unit, 65, Tsurumai-cho, Showa-ku, Nagoya 466-8560, Aichi, Japan
| | - Chiaki Koike
- Nagoya University Institute for Advanced Research, Advanced Analytical and Diagnostic Imaging Center (AADIC) / Medical Engineering Unit (MEU), B3 Unit, 65, Tsurumai-cho, Showa-ku, Nagoya 466-8560, Aichi, Japan
| | - Tetsuro Taki
- Department of Pathology, Nagoya University Graduate School of Medicine, 65, Tsurumai-cho, Showa-ku, Nagoya 466-8560, Aichi, Japan
| | - Aya Sakamoto
- Perseus Proteomics, Inc., 4-7-6, Komaba 153-0041, Meguro-ku, Tokyo, Japan
| | - Keiko Katsumi
- Perseus Proteomics, Inc., 4-7-6, Komaba 153-0041, Meguro-ku, Tokyo, Japan
| | - Keisuke Ishii
- Perseus Proteomics, Inc., 4-7-6, Komaba 153-0041, Meguro-ku, Tokyo, Japan
| | - Kazuhide Sato
- Respiratory Medicine, Nagoya University Graduate School of Medicine, 65, Tsurumai-cho, Showa-ku, Nagoya 466-8560, Aichi, Japan; Nagoya University Institute for Advanced Research, Advanced Analytical and Diagnostic Imaging Center (AADIC) / Medical Engineering Unit (MEU), B3 Unit, 65, Tsurumai-cho, Showa-ku, Nagoya 466-8560, Aichi, Japan; FOREST- Souhatsu, CREST, JST; Nagoya University Institute for Advanced Research, S-YLC, Furo-cho, Chikusa-ku, Nagoya 464-8601, Aichi,, Japan.
| |
Collapse
|
28
|
Paraboschi I, Turnock S, Kramer-Marek G, Musleh L, Barisa M, Anderson J, Giuliani S. Near-InfraRed PhotoImmunoTherapy (NIR-PIT) for the local control of solid cancers: Challenges and potentials for human applications. Crit Rev Oncol Hematol 2021; 161:103325. [PMID: 33836238 PMCID: PMC8177002 DOI: 10.1016/j.critrevonc.2021.103325] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 01/29/2021] [Accepted: 03/26/2021] [Indexed: 01/10/2023] Open
Abstract
Near-InfraRed PhotoImmunoTherapy (NIR-PIT) is a novel cancer-targeted treatment effected by a chemical conjugation between a photosensitiser (e.g. the NIR phthalocyanine dye IRDye700DX) and a cancer-targeting moiety (e.g. a monoclonal antibody, moAb). Delivery of a conjugate in vivo leads to accumulation at the tumour cell surface by binding to cell surface receptors or antigens. Upon deployment of focal NIR-light, irradiation of the conjugate results in a rapid, targeted cell death. However, the mechanisms of action to produce the cytotoxic effects have yet to be fully understood. Herein, we bring together the current knowledge of NIR-PIT from preclinical and clinical studies in a variety of cancers highlighting the key unanswered research questions. Furthermore, we discuss how to enhance the local control of solid cancers using this novel treatment regimen.
Collapse
Affiliation(s)
- Irene Paraboschi
- Wellcome/EPSRC Centre for Interventional & Surgical Sciences, University College London, London, UK
| | - Stephen Turnock
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, UK
| | | | - Layla Musleh
- Department of Specialist Neonatal and Pediatric Surgery, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Marta Barisa
- Cancer Section, Developmental Biology and Cancer Programme, UCL Great Ormond Street Institute of Child Health, London, UK
| | - John Anderson
- Cancer Section, Developmental Biology and Cancer Programme, UCL Great Ormond Street Institute of Child Health, London, UK; Department of Oncology, Great Ormond Street Hospital for Children NHS Foundation Trust, London, England, UK
| | - Stefano Giuliani
- Wellcome/EPSRC Centre for Interventional & Surgical Sciences, University College London, London, UK; Department of Specialist Neonatal and Pediatric Surgery, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK.
| |
Collapse
|
29
|
Akopov A, Papayan G. Photodynamic theranostics of central lung cancer: Present state and future prospects. Photodiagnosis Photodyn Ther 2021; 33:102203. [PMID: 33529744 DOI: 10.1016/j.pdpdt.2021.102203] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 12/13/2020] [Accepted: 01/25/2021] [Indexed: 12/12/2022]
Abstract
Endobronchial photodynamic therapy (PDT) in central lung cancer (CLC) shows feasibility even in late stage disease. Our experience with chlorin e6 based photosensitizers (PS), including in combination with medical cancer treatment, demonstrated regression of tumor lesions of the trachea and bronchi in 94 % of patients with central NSCLC. It is possible to increase the efficiency of the treatment and achieve its personalization by using fluorescent bronchoscopes, which provide fluorescence guided PDT - photodynamic theranostics (PT). PT allows to clarify localization the area of treatment due to visualization of tumor foci which are invisible in white light, to carry out targeted irradiation and at the same time to monitor its effectiveness using the effects of bleaching/flare-up of PS. PT prospects are associated with the transition to the near-infrared (NIR) region, which makes possible to increase the depth of light penetration. The first experiments using the combined NIR/visible PT system showed the possibility of detecting tumor sites using the OS-BPT method (On-Site Bronchoscopic Photodynamic Theranostics), which consists in NIR visualization of tumor when indocyanine green (ICG) is injected directly during examination in a minimal dose. This allows the technology to be used for CLC screening in the future. Further progress of endobronchial PT will be determined by the development of clinically available devices and new NIR PSs with targeted properties, high singlet oxygen yield and fluorescence.
Collapse
Affiliation(s)
- Andrey Akopov
- Department of Thoracic Oncology, Institute of Surgery and Emergency Medicine, Pavlov First State Medical University, Saint-Petersburg, Russia.
| | - Garry Papayan
- Department of Thoracic Oncology, Institute of Surgery and Emergency Medicine, Pavlov First State Medical University, Saint-Petersburg, Russia; Institute of Experimental Medicine, Almazov Federal Medical Research Center, Saint-Petersburg, Russia
| |
Collapse
|
30
|
Hirata H, Kuwatani M, Nakajima K, Kodama Y, Yoshikawa Y, Ogawa M, Sakamoto N. Near-infrared photoimmunotherapy (NIR-PIT) on cholangiocarcinoma using a novel catheter device with light emitting diodes. Cancer Sci 2021; 112:828-838. [PMID: 33345417 PMCID: PMC7894014 DOI: 10.1111/cas.14780] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 12/14/2020] [Accepted: 12/17/2020] [Indexed: 12/11/2022] Open
Abstract
Near-infrared photoimmunotherapy (NIR-PIT) is a novel therapy for cancers that uses NIR light and antibody-photosensitizer (IR700) conjugates. However, it is difficult to deliver NIR light into the bile duct for cholangiocarcinoma (CCA) from the conventional extracorporeal apparatus. Thus, in this study, we developed a dedicated catheter with light emitting diodes (LEDs) that supersedes conventional external irradiation devices; we investigated the therapeutic effect of NIR-PIT for CCA using the novel catheter. The new catheter was designed to be placed in the bile duct and a temperature sensor was attached to the tip of the catheter to avoid thermal burn. An anti-epidermal growth factor receptor (EGFR) antibody, Panitumumab-IR700 conjugate or anti-human epidermal growth factor receptor type 2 (HER2) antibody, Trastuzumab-IR700 conjugate, was used with EGFR- or HER2-expressing cell lines, respectively. The in vitro efficacy of NIR-PIT was confirmed in cultured cells; the capability of the new catheter for NIR-PIT was then tested in a mouse tumor model. NIR-PIT via the developed catheter treated CCA xenografts in mice. NIR-PIT had an effect in Panitumumab-IR700 conjugate- and Trastuzumab-IR700 conjugate-treated CCA cells that depended on the receptor expression level. Tumor growth was significantly suppressed in mice treated with NIR-PIT using the novel catheter compared with controls (P < .01). NIR-PIT was an effective treatment for EGFR- and HER2-expressing CCA cells, and the novel catheter with mounted LEDs was useful for NIR-PIT of CCA.
Collapse
Affiliation(s)
- Hajime Hirata
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Masaki Kuwatani
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Kohei Nakajima
- Laboratory of Bioanalysis and Molecular Imaging, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | | | | | - Mikako Ogawa
- Laboratory of Bioanalysis and Molecular Imaging, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Naoya Sakamoto
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| |
Collapse
|
31
|
Yasui H, Takahashi K, Taki S, Shimizu M, Koike C, Umeda K, Rahman S, Akashi T, Nguyen VS, Nakagawa Y, Sato K. Near Infrared Photo‐Antimicrobial Targeting Therapy for
Candida albicans. ADVANCED THERAPEUTICS 2021. [DOI: 10.1002/adtp.202000221] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Hirotoshi Yasui
- Respiratory Medicine Nagoya University Graduate School of Medicine 65 Tsuumai‐cho, Showa‐ku Nagoya Aichi 466‐8550 Japan
| | - Kazuomi Takahashi
- Respiratory Medicine Nagoya University Graduate School of Medicine 65 Tsuumai‐cho, Showa‐ku Nagoya Aichi 466‐8550 Japan
| | - Shunichi Taki
- Respiratory Medicine Nagoya University Graduate School of Medicine 65 Tsuumai‐cho, Showa‐ku Nagoya Aichi 466‐8550 Japan
| | - Misae Shimizu
- Advanced Analytical and Diagnostic Imaging Center (AADIC)/Medical Engineering Unit (MEU), B3 Unit Nagoya University Institute for Advanced Research 65 Tsuumai‐cho, Showa‐ku Nagoya Aichi 466‐8550 Japan
| | - Chiaki Koike
- Advanced Analytical and Diagnostic Imaging Center (AADIC)/Medical Engineering Unit (MEU), B3 Unit Nagoya University Institute for Advanced Research 65 Tsuumai‐cho, Showa‐ku Nagoya Aichi 466‐8550 Japan
| | - Koji Umeda
- EW Nutrition Japan Immunology Research Institute in Gifu 839‐7, Gifu‐City Sano Gifu 501‐1101 Japan
| | - Shofiqur Rahman
- EW Nutrition Japan Immunology Research Institute in Gifu 839‐7, Gifu‐City Sano Gifu 501‐1101 Japan
| | - Tomohiro Akashi
- Division of OMICS Analysis Nagoya University Graduate School of Medicine 65 Tsuumai‐cho, Showa‐ku Nagoya Aichi 466‐8550 Japan
- Division of Systems Biology Nagoya University Graduate School of Medicine 65 Tsuumai‐cho, Showa‐ku Nagoya Aichi 466‐8550 Japan
- S‐YLC Nagoya University Institute for Advanced Research Furo‐cho, Chikusa‐ku Nagoya Aichi 464‐8601 Japan
| | - Van Sa Nguyen
- EW Nutrition Japan Immunology Research Institute in Gifu 839‐7, Gifu‐City Sano Gifu 501‐1101 Japan
| | - Yoshiyuki Nakagawa
- Division of OMICS Analysis Nagoya University Graduate School of Medicine 65 Tsuumai‐cho, Showa‐ku Nagoya Aichi 466‐8550 Japan
| | - Kazuhide Sato
- Respiratory Medicine Nagoya University Graduate School of Medicine 65 Tsuumai‐cho, Showa‐ku Nagoya Aichi 466‐8550 Japan
- CREST, JST Honcho Kawaguchi Saitama 332‐0012 Japan
| |
Collapse
|
32
|
Kobayashi H, Furusawa A, Rosenberg A, Choyke PL. Near-infrared photoimmunotherapy of cancer: a new approach that kills cancer cells and enhances anti-cancer host immunity. Int Immunol 2021; 33:7-15. [PMID: 32496557 PMCID: PMC7771006 DOI: 10.1093/intimm/dxaa037] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 05/27/2020] [Indexed: 12/16/2022] Open
Abstract
Near-infrared photoimmunotherapy (NIR-PIT) is a recently developed hybrid cancer therapy that directly kills cancer cells as well as producing a therapeutic host immune response. Conventional immunotherapies, such as immune-activating cytokine therapy, checkpoint inhibition, engineered T cells and suppressor cell depletion, do not directly destroy cancer cells, but rely exclusively on activating the immune system. NIR-PIT selectively destroys cancer cells, leading to immunogenic cell death that initiates local immune reactions to released cancer antigens from dying cancer cells. These are characterized by rapid maturation of dendritic cells and priming of multi-clonal cancer-specific cytotoxic T cells that kill cells that escaped the initial direct effects of NIR-PIT. The NIR-PIT can be applied to a wide variety of cancers either as monotherapy or in combination with conventional immune therapies to further activate anti-cancer immunity. A global Phase 3 clinical trial (https://clinicaltrials.gov/ct2/show/NCT03769506) of NIR-PIT targeting the epidermal growth factor receptor (EGFR) in patients with recurrent head and neck cancer is underway, employing RM1929/ASP1929, a conjugate of anti-EGFR antibody (cetuximab) plus the photo-absorber IRDye700DX (IR700). NIR-PIT has been given fast-track recognition by regulators in the USA and Japan. A variety of imaging methods, including direct IR700 fluorescence imaging, can be used to monitor NIR-PIT. As experience with NIR-PIT grows, additional antibodies will be employed to target additional antigens on other cancers or to target immune-suppressor cells to enhance host immunity. NIR-PIT will be particularly important in patients with localized and locally advanced cancers and may help such patients avoid side-effects associated with surgery, radiation and chemotherapy.
Collapse
Affiliation(s)
- Hisataka Kobayashi
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Aki Furusawa
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Adrian Rosenberg
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Peter L Choyke
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
33
|
Wang M, Rao J, Wang M, Li X, Liu K, Naylor MF, Nordquist RE, Chen WR, Zhou F. Cancer photo-immunotherapy: from bench to bedside. Theranostics 2021; 11:2218-2231. [PMID: 33500721 PMCID: PMC7797676 DOI: 10.7150/thno.53056] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 11/18/2020] [Indexed: 02/06/2023] Open
Abstract
Targeted therapy and immunotherapy in combination is considered the ideal strategy for treating metastatic cancer, as it can eliminate the primary tumors and induce host immunity to control distant metastases. Phototherapy, a promising targeted therapy, eradicates primary tumors using an appropriate dosage of focal light irradiation, while initiating antitumor immune responses through induced immunogenic tumor cell death. Recently, phototherapy has been employed to improve the efficacy of immunotherapies such as chimeric antigen receptor T-cell therapy and immune checkpoint inhibitors. Phototherapy and immunoadjuvant therapy have been used in combination clinically, wherein the induced immunogenic cell death and enhanced antigen presentation synergy, inducing a systemic antitumor immune response to control residual tumor cells at the treatment site and distant metastases. This review summarizes studies on photo-immunotherapy, the combination of phototherapy and immunotherapy, especially focusing on the development and progress of this unique combination from a benchtop project to a promising clinical therapy for metastatic cancer.
Collapse
Affiliation(s)
- Miao Wang
- School of Biomedical Engineering, Hainan University, Haikou, 570228, China
| | - Jie Rao
- School of Biomedical Engineering, Hainan University, Haikou, 570228, China
| | - Meng Wang
- College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen, 518060, China
| | - Xiaosong Li
- Department of Oncology, the First Affiliated Hospital of Chinese PLA General Hospital, Beijing 100048, China
| | - Kaili Liu
- Stephenson School of Biomedical Engineering, The University of Oklahoma, Norman, OK 73019, USA
| | | | - Robert E. Nordquist
- Immunophotonics, Inc., 4320 Forest Park Ave., #303 (BAL), St. Louis, MO 63108, USA
| | - Wei R. Chen
- Stephenson School of Biomedical Engineering, The University of Oklahoma, Norman, OK 73019, USA
| | - Feifan Zhou
- School of Biomedical Engineering, Hainan University, Haikou, 570228, China
| |
Collapse
|
34
|
Nagaya T, Choyke PL, Kobayashi H. Near-Infrared Photoimmunotherapy for Cancers of the Gastrointestinal Tract. Digestion 2020; 102:1-8. [PMID: 33316807 PMCID: PMC8200364 DOI: 10.1159/000513216] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 11/20/2020] [Indexed: 02/04/2023]
Abstract
BACKGROUND Cancers of the gastrointestinal (GI) tract are the common leading cause of cancer-related death in the world. Recent advances in cancer therapies such as intensive multidrug chemotherapy and molecular targeted treatment have improved therapeutic efficacy; however, the outcomes are not satisfied. Moreover, these therapies also cause severe side effects. New type of cancer therapies is urgently needed to improve the outcomes and to reduce side effects of GI tract cancers. SUMMARY This account is a comprehensive review article on the newly developed, photochemistry-based cancer therapy named as near-infrared photoimmunotherapy (NIR-PIT). NIR-PIT is a highly selective tumor treatment that employs an antibody-photoabsorber conjugate, which is activated by near-infrared light. A world-wide phase 3 clinical trial of NIR-PIT against recurrent head and neck cancer patients is currently underway. NIR-PIT differs from conventional cancer therapies such as surgery, chemotherapy, and radiation in its selectivity for killing cancer cells and cells treated with NIR-PIT leading to immunogenic cell death. Preclinical research in animals with combining cancer-targeting NIR-PIT and other cancer immunotherapies could lead to responses not only in local tumor but also in distant metastases. NIR-PIT also leads to an immediate and dramatic increase in vascular permeability after therapy. From these aspects, NIR-PIT appears to be a promising new form of cancer therapy. NIR-PIT could be readily translated into clinical use for virtually any cancers in the near future provided suitable humanized antibodies are available. Here, we describe the specific advantages and applications of NIR-PIT in the GI tract. Key Messages: We believe that NIR-PIT with NIR excitation light, which can be delivered via a fiber optic diffuser through endoscopes, is a promising method for a new treatment of GI cancers.
Collapse
Affiliation(s)
- Tadanobu Nagaya
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA,
- Department of Gastroenterology, Shinshu University Hospital, Matsumoto, Japan,
| | - Peter L Choyke
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Hisataka Kobayashi
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
35
|
Polikarpov DM, Campbell DH, Lund ME, Lu Y, Lu Y, Wu J, Walsh BJ, Zvyagin AV, Gillatt DA. The feasibility of Miltuximab®-IRDye700DX-mediated photoimmunotherapy of solid tumors. Photodiagnosis Photodyn Ther 2020; 32:102064. [PMID: 33069874 DOI: 10.1016/j.pdpdt.2020.102064] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 09/28/2020] [Accepted: 10/02/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND Photoimmunotherapy (PIT) is an emerging method of cancer treatment based on the use of a photosensitizer near-infrared dye IRDye700DX (IR700) conjugated to a monoclonal antibody. The antibody selectively delivers IR700 to cancer cells, which can then be killed after photoexcitation. Glypican-1 (GPC-1) is a novel target expressed specifically in malignant tumors. We aimed to investigate whether anti-GPC-1 antibody Miltuximab® (Glytherix Ltd., Sydney, Australia) can be conjugated with IR700 for PIT of solid tumors. METHODS The dye IR700 was conjugated with Miltuximab® and characterized by spectrophotometry and flow cytometry. Miltuximab®-IR700-mediated PIT was tested in prostate (DU-145), bladder (C3 and T-24), brain (U-87 and U-251) and ovarian (SKOV-3) cancer cell lines. After 1 h incubation with Miltuximab®-IR700, the cells were washed by PBS and illuminated using a 690-nm light-emitting diode. The viability of the cells was assessed by a CCK-8 viability kit 24 h later. RESULTS Miltuximab®-IR700-mediated PIT caused 67.3-92.3% reduction in viability of cells with medium-high GPC-1 expression and did not affect the viability of GPC-1-low cells. Cytotoxicity was attributed to the targeted binding of the conjugate with subsequent photoactivation, as the conjugate or light exposure alone had no effect on the cell viability. Miltuximab®-IR700 did not induce cytotoxicity in cells blocked by unconjugated Miltuximab®. CONCLUSIONS PIT with Miltuximab®-IR700 appears to be highly specific and effective against GPC-1-expressing cancer cells, indicating that it holds promise for an effective and safe treatment of early stage solid tumors or as adjuvant therapy following surgical resection. These findings necessitate further investigation of PIT with Miltuximab®-IR700 in other GPC-1-expressing cancer cell lines in vitro and in vivo in xenograft tumor models.
Collapse
Affiliation(s)
- Dmitry M Polikarpov
- Department of Clinical Medicine, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW, 2109, Australia.
| | | | | | - Yanling Lu
- Glytherix Ltd., Sydney, NSW, 2113, Australia
| | - Yiqing Lu
- ARC Centre of Excellence for Nanoscale BioPhotonics, Macquarie University, Sydney, NSW, 2109, Australia
| | - Jiehua Wu
- Glytherix Ltd., Sydney, NSW, 2113, Australia
| | | | - Andrei V Zvyagin
- ARC Centre of Excellence for Nanoscale BioPhotonics, Macquarie University, Sydney, NSW, 2109, Australia; Institute of Molecular Medicine, Sechenov University, 119991, Moscow, Russia.
| | - David A Gillatt
- Department of Clinical Medicine, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW, 2109, Australia
| |
Collapse
|
36
|
Targeted Phototherapy for Malignant Pleural Mesothelioma: Near-Infrared Photoimmunotherapy Targeting Podoplanin. Cells 2020; 9:cells9041019. [PMID: 32326079 PMCID: PMC7225918 DOI: 10.3390/cells9041019] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 04/15/2020] [Accepted: 04/16/2020] [Indexed: 12/22/2022] Open
Abstract
Malignant pleural mesothelioma (MPM) has extremely limited treatment despite a poor prognosis. Moreover, molecular targeted therapy for MPM has not yet been implemented; thus, a new targeted therapy is highly desirable. Near-infrared photoimmunotherapy (NIR-PIT) is a recently developed cancer therapy that combines the specificity of antibodies for targeting tumors with toxicity induced by the photoabsorber after exposure to NIR-light. In this study, we developed a new phototherapy targeting podoplanin (PDPN) for MPM with the use of both NIR-PIT and an anti-PDPN antibody, NZ-1. An antibody–photosensitizer conjugate consisting of NZ-1 and phthalocyanine dye was synthesized. In vitro NIR-PIT-induced cytotoxicity was measured with both dead cell staining and luciferase activity on various MPM cell lines. In vivo NIR-PIT was examined in both the flank tumor and orthotopic mouse model with in vivo real-time imaging. In vitro NIR-PIT-induced cytotoxicity was NIR-light dose dependent. In vivo NIR-PIT led to significant reduction in both tumor volume and luciferase activity in a flank model (p < 0.05, NIR-PIT group versus NZ-1-IR700 group). The PDPN-targeted NIR-PIT resulted in a significant antitumor effect in an MPM orthotopic mouse model (p < 0.05, NIR-PIT group versus NZ-1-IR700 group). This study suggests that PDPN-targeted NIR-PIT could be a new promising treatment for MPM.
Collapse
|
37
|
Isobe Y, Sato K, Nishinaga Y, Takahashi K, Taki S, Yasui H, Shimizu M, Endo R, Koike C, Kuramoto N, Yukawa H, Nakamura S, Fukui T, Kawaguchi K, Chen-Yoshikawa TF, Baba Y, Hasegawa Y. Near infrared photoimmunotherapy targeting DLL3 for small cell lung cancer. EBioMedicine 2020; 52:102632. [PMID: 31981983 PMCID: PMC6992936 DOI: 10.1016/j.ebiom.2020.102632] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Revised: 12/25/2019] [Accepted: 01/06/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Small cell lung cancer (SCLC) has a poor prognosis, and its treatment options are limited. Delta-like protein 3 (DLL3) is expressed specifically in SCLC and is considered a promising therapeutic target for patients with this disease. Rovalpituzumab tesirine (Rova-T) was the first antibody-drug conjugate targeting DLL3. Although Rova-T development was unfortunately terminated, DLL3 remains an ideal target for SCLC. Near infrared photoimmunotherapy (NIR-PIT) is a new form of cancer treatment that employs an antibody-photosensitiser conjugate followed by NIR light exposure and damage target cells specifically. In this study, we demonstrate DLL3-targeted NIR-PIT to develop a novel molecularly targeted treatment for SCLC. METHODS The anti-DLL3 monoclonal antibody rovalpituzumab was conjugated to an IR700 photosensitiser (termed 'rova-IR700'). SCLC cells overexpressing DLL3 as well as non-DLL3-expressing controls were incubated with rova-IR700 and then exposed to NIR-light. Next, mice with SCLC xenografts were injected with rova-IR700 and irradiated with NIR-light. FINDINGS DLL3-overexpressing cells underwent immediate destruction upon NIR-light exposure, whereas the control cells remained intact. The xenograft in mice treated with rova-IR700 and NIR-light shrank markedly, whereas neither rova-IR700 injection nor NIR-light irradiation alone affected tumour size. INTERPRETATION Our data suggest that targeting of DLL3 using NIR-PIT could be a novel and promising treatment for SCLC. FUNDING Research supported by grants from the Program for Developing Next-generation Researchers (Japan Science and Technology Agency), KAKEN (18K15923, JSPS), Medical Research Encouragement Prize of The Japan Medical Association, The Nitto Foundation, Kanae Foundation for the Promotion of Medical Science.
Collapse
Affiliation(s)
- Yoshitaka Isobe
- Department of Respiratory Medicine, Nagoya University Graduate School of Medicine, Nagoya, Aichi 466-8550, Japan
| | - Kazuhide Sato
- Department of Respiratory Medicine, Nagoya University Graduate School of Medicine, Nagoya, Aichi 466-8550, Japan; S-YLC, Nagoya University Institute for Advanced Research, Japan; B3-Unit, Advanced Analytical and Diagnostic Imaging Center (AADIC)/Medical Engineering Unit (MEU), Nagoya University Institute for Advanced Research, Japan.
| | - Yuko Nishinaga
- Department of Respiratory Medicine, Nagoya University Graduate School of Medicine, Nagoya, Aichi 466-8550, Japan
| | - Kazuomi Takahashi
- Department of Respiratory Medicine, Nagoya University Graduate School of Medicine, Nagoya, Aichi 466-8550, Japan
| | - Shunichi Taki
- Department of Respiratory Medicine, Nagoya University Graduate School of Medicine, Nagoya, Aichi 466-8550, Japan
| | - Hirotoshi Yasui
- Department of Respiratory Medicine, Nagoya University Graduate School of Medicine, Nagoya, Aichi 466-8550, Japan
| | - Misae Shimizu
- Department of Respiratory Medicine, Nagoya University Graduate School of Medicine, Nagoya, Aichi 466-8550, Japan; B3-Unit, Advanced Analytical and Diagnostic Imaging Center (AADIC)/Medical Engineering Unit (MEU), Nagoya University Institute for Advanced Research, Japan
| | - Rena Endo
- Department of Respiratory Medicine, Nagoya University Graduate School of Medicine, Nagoya, Aichi 466-8550, Japan; B3-Unit, Advanced Analytical and Diagnostic Imaging Center (AADIC)/Medical Engineering Unit (MEU), Nagoya University Institute for Advanced Research, Japan
| | - Chiaki Koike
- Department of Respiratory Medicine, Nagoya University Graduate School of Medicine, Nagoya, Aichi 466-8550, Japan; B3-Unit, Advanced Analytical and Diagnostic Imaging Center (AADIC)/Medical Engineering Unit (MEU), Nagoya University Institute for Advanced Research, Japan
| | - Noriko Kuramoto
- B3-Unit, Advanced Analytical and Diagnostic Imaging Center (AADIC)/Medical Engineering Unit (MEU), Nagoya University Institute for Advanced Research, Japan
| | - Hiroshi Yukawa
- B3-Unit, Advanced Analytical and Diagnostic Imaging Center (AADIC)/Medical Engineering Unit (MEU), Nagoya University Institute for Advanced Research, Japan; Nagoya University Institute of Nano-Life-Systems, Institutes of Innovation for Future Society, Japan; Department of Biomolecular Engineering, Nagoya University Graduate School of Engineering, Japan
| | - Shota Nakamura
- Department of Thoracic Surgery, Nagoya University Graduate School of Medicine, Japan
| | - Takayuki Fukui
- Department of Thoracic Surgery, Nagoya University Graduate School of Medicine, Japan
| | - Koji Kawaguchi
- Department of Thoracic Surgery, Nagoya University Graduate School of Medicine, Japan
| | | | - Yoshinobu Baba
- Nagoya University Institute of Nano-Life-Systems, Institutes of Innovation for Future Society, Japan; Department of Biomolecular Engineering, Nagoya University Graduate School of Engineering, Japan
| | | |
Collapse
|
38
|
Abstract
This Account is the first comprehensive review article on the newly developed, photochemistry-based cancer therapy near-infrared (NIR) photoimmunotherapy (PIT). NIR-PIT is a molecularly targeted phototherapy for cancer that is based on injecting a conjugate of a near-infrared, water-soluble, silicon-phthalocyanine derivative, IRdye700DX (IR700), and a monoclonal antibody (mAb) that targets an expressed antigen on the cancer cell surface. Subsequent local exposure to NIR light turns on this photochemical "death" switch, resulting in the rapid and highly selective immunogenic cell death (ICD) of targeted cancer cells. ICD occurs as early as 1 min after exposure to NIR light and results in irreversible morphologic changes only in target-expressing cells based on the newly discovered photoinduced ligand release reaction that induces physical changes on conjugated antibody/antigen complex resulting in functional damage on cell membrane. Meanwhile, immediately adjacent receptor-negative cells are totally unharmed. Because of its highly targeted nature, NIR-PIT carries few side effects and healing is rapid. Evaluation of the tumor microenvironment reveals that ICD induced by NIR-PIT results in rapid maturation of immature dendritic cells adjacent to dying cancer cells initiating a host anticancer immune response, resulting in repriming of polyclonal CD8+T cells against various released cancer antigens, which amplifies the therapeutic effect of NIR-PIT. NIR-PIT can target and treat virtually any cell surface antigens including cancer stem cell markers, that is, CD44 and CD133. A first-in-human phase 1/2 clinical trial of NIR-PIT using cetuximab-IR700 (RM1929) targeting EGFR in inoperable recurrent head and neck cancer patients successfully concluded in 2017 and led to "fast tracking" by the FDA and a phase 3 trial ( https://clinicaltrials.gov/ct2/show/NCT03769506 ) that is currently underway in 3 countries in Asia, US/Canada, and 4 countries in EU. The next step for NIR-PIT is to further exploit the immune response. Preclinical research in animals with intact immune systems has shown that NIT-PIT targeting of immunosuppressor cells within the tumor, such as regulatory T-cells, can further enhance tumor-cell-selective systemic host-immunity leading to significant responses in distant metastatic tumors, which are not treated with light. By combining cancer-targeting NIR-PIT and immune-activating NIR-PIT or other cancer immunotherapies, NIR-PIT of a local tumor, could lead to responses in distant metastases and may also inhibit recurrences due to activation of systemic anticancer immunity and long-term immune memory without the systemic autoimmune adverse effects often associated with immune checkpoint inhibitors. Furthermore, NIR-PIT also enhances nanodrug delivery into tumors up to 24-fold superior to untreated tumors with conventional EPR effects by intensively damaging cancer cells behind tumor vessels. We conclude by describing future advances in this novel photochemical cancer therapy that are likely to further enhance the efficacy of NIR-PIT.
Collapse
Affiliation(s)
- Hisataka Kobayashi
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Building 10, Room B3B69, MSC1088, Bethesda, Maryland 20892-1088, United States
| | - Peter L. Choyke
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Building 10, Room B3B69, MSC1088, Bethesda, Maryland 20892-1088, United States
| |
Collapse
|
39
|
FA-NBs-IR780: Novel multifunctional nanobubbles as molecule-targeted ultrasound contrast agents for accurate diagnosis and photothermal therapy of cancer. Cancer Lett 2019; 455:14-25. [DOI: 10.1016/j.canlet.2019.04.023] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 03/30/2019] [Accepted: 04/17/2019] [Indexed: 12/22/2022]
|
40
|
Poiroux G, Barre A, Rougé P, Benoist H. Targeting Glycosylation Aberrations to Improve the Efficiency of Cancer Phototherapy. Curr Cancer Drug Targets 2019; 19:349-359. [DOI: 10.2174/1568009618666180628101059] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 05/12/2018] [Accepted: 06/01/2018] [Indexed: 12/13/2022]
Abstract
The use of photodynamic therapy in cancer still remains limited, partly because of the lack of photosensitizer (PS) specificity for the cancerous tissues. Various molecular tools are available to increase PS efficiency by targeting the cancer cell molecular alterations. Most strategies use the protein-protein interactions, e.g. monoclonal antibodies directed toward tumor antigens, such as HER2 or EGFR. An alternative could be the targeting of the tumor glycosylation aberrations, e.g. T/Tn antigens that are truncated O-glycans over-expressed in numerous tumors. Thus, to achieve an effective targeting, PS can be conjugated to molecules that specifically recognize the Oglycosylation aberrations at the cancer cell surface.
Collapse
Affiliation(s)
- Guillaume Poiroux
- Universite de Toulouse, CRCT, INSERM UMR 1037, 2 Avenue Hubert Curien, 31037 Toulouse, France
| | - Annick Barre
- Universite de Toulouse, Pharma-Dev, Institut de Recherche pour le Developpement (IRD) UMR 152, Faculte des Sciences Pharmaceutiques, F-31062 Toulouse, Cedex 09, France
| | - Pierre Rougé
- Universite de Toulouse, Pharma-Dev, Institut de Recherche pour le Developpement (IRD) UMR 152, Faculte des Sciences Pharmaceutiques, F-31062 Toulouse, Cedex 09, France
| | - Hervé Benoist
- Universite de Toulouse, Pharma-Dev, Institut de Recherche pour le Developpement (IRD) UMR 152, Faculte des Sciences Pharmaceutiques, F-31062 Toulouse, Cedex 09, France
| |
Collapse
|
41
|
Sandland J, Boyle RW. Photosensitizer Antibody–Drug Conjugates: Past, Present, and Future. Bioconjug Chem 2019; 30:975-993. [DOI: 10.1021/acs.bioconjchem.9b00055] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
42
|
Sato K, Ando K, Okuyama S, Moriguchi S, Ogura T, Totoki S, Hanaoka H, Nagaya T, Kokawa R, Takakura H, Nishimura M, Hasegawa Y, Choyke PL, Ogawa M, Kobayashi H. Photoinduced Ligand Release from a Silicon Phthalocyanine Dye Conjugated with Monoclonal Antibodies: A Mechanism of Cancer Cell Cytotoxicity after Near-Infrared Photoimmunotherapy. ACS CENTRAL SCIENCE 2018; 4:1559-1569. [PMID: 30555909 PMCID: PMC6276043 DOI: 10.1021/acscentsci.8b00565] [Citation(s) in RCA: 162] [Impact Index Per Article: 23.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Indexed: 05/23/2023]
Abstract
Photochemical reactions can dramatically alter physical characteristics of reacted molecules. In this study, we demonstrate that near-infrared (NIR) light induces an axial ligand-releasing reaction, which dramatically alters hydrophilicity of a silicon phthalocyanine derivative (IR700) dye leading to a change in the shape of the conjugate and its propensity to aggregate in aqueous solution. This photochemical reaction is proposed as a major mechanism of cell death induced by NIR photoimmunotherapy (NIR-PIT), which was recently developed as a molecularly targeted cancer therapy. Once the antibody-IR700 conjugate is bound to its target, activation by NIR light causes physical changes in the shape of antibody antigen complexes that are thought to induce physical stress within the cellular membrane leading to increases in transmembrane water flow that eventually lead to cell bursting and necrotic cell death.
Collapse
Affiliation(s)
- Kazuhide Sato
- Molecular
Imaging Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892-1088, United States
- Institute
for Advanced Research, Nagoya University, Nagoya, Aichi 464-0814, Japan
- Department
of Respiratory Medicine, Nagoya University
Graduate School of Medicine, Nagoya, Aichi 466-8550, Japan
| | - Kanta Ando
- Laboratory
for Bioanalysis and Molecular Imaging, Graduate School of Pharmaceutical
Sciences, Hokkaido University, Sapporo, Hokkaido 060-0812, Japan
| | - Shuhei Okuyama
- Molecular
Imaging Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892-1088, United States
- Shimadzu
Corporation, Kyoto 604-8511, Japan
| | | | | | | | - Hirofumi Hanaoka
- Molecular
Imaging Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892-1088, United States
| | - Tadanobu Nagaya
- Molecular
Imaging Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892-1088, United States
| | | | - Hideo Takakura
- Laboratory
for Bioanalysis and Molecular Imaging, Graduate School of Pharmaceutical
Sciences, Hokkaido University, Sapporo, Hokkaido 060-0812, Japan
| | | | - Yoshinori Hasegawa
- Department
of Respiratory Medicine, Nagoya University
Graduate School of Medicine, Nagoya, Aichi 466-8550, Japan
| | - Peter L. Choyke
- Molecular
Imaging Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892-1088, United States
| | - Mikako Ogawa
- Laboratory
for Bioanalysis and Molecular Imaging, Graduate School of Pharmaceutical
Sciences, Hokkaido University, Sapporo, Hokkaido 060-0812, Japan
| | - Hisataka Kobayashi
- Molecular
Imaging Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892-1088, United States
| |
Collapse
|
43
|
Evans ER, Bugga P, Asthana V, Drezek R. Metallic Nanoparticles for Cancer Immunotherapy. MATERIALS TODAY (KIDLINGTON, ENGLAND) 2018; 21:673-685. [PMID: 30197553 PMCID: PMC6124314 DOI: 10.1016/j.mattod.2017.11.022] [Citation(s) in RCA: 118] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
Cancer immunotherapy, or the utilization of the body's immune system to attack tumor cells, has gained prominence over the past few decades as a viable cancer treatment strategy. Recently approved immunotherapeutics have conferred remission upon patients with previously bleak outcomes and have expanded the number of tools available to treat cancer. Nanoparticles -including polymeric, liposomal, and metallic formulations - naturally traffic to the spleen and lymph organs and the relevant immune cells therein, making them good candidates for delivery of immunotherapeutic agents. Metallic nanoparticle formulations in particular are advantageous because of their potential for dense surface functionalization and their capability for optical or heat based therapeutic methods. Many research groups have investigated the potential of nanoparticle-mediated delivery platforms to improve the efficacy of immunotherapies. Despite the significant preclinical successes demonstrated by many of these platforms over the last twenty years, few metallic nanoparticles have successfully entered clinical trials with none achieving FDA approval for cancer therapy. In this review, we will discuss preclinical research and clinical trials involving metallic nanoparticles (MNPs) for cancer immunotherapy applications and discuss the potential for clinical translation of MNPs.
Collapse
Affiliation(s)
- Emily Reiser Evans
- Department of Bioengineering, Rice University, Houston, TX 77005, United States
| | - Pallavi Bugga
- Department of Bioengineering, Rice University, Houston, TX 77005, United States
| | - Vishwaratn Asthana
- Department of Bioengineering, Rice University, Houston, TX 77005, United States
| | - Rebekah Drezek
- Department of Bioengineering, Rice University, Houston, TX 77005, United States. Department of Electrical and Computer Engineering, Rice University, Houston, TX 77005, United States
| |
Collapse
|
44
|
Burley TA, Mączyńska J, Shah A, Szopa W, Harrington KJ, Boult JK, Mrozek‐Wilczkiewicz A, Vinci M, Bamber JC, Kaspera W, Kramer‐Marek G. Near-infrared photoimmunotherapy targeting EGFR-Shedding new light on glioblastoma treatment. Int J Cancer 2018; 142:2363-2374. [PMID: 29313975 PMCID: PMC6016485 DOI: 10.1002/ijc.31246] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Revised: 11/30/2017] [Accepted: 12/19/2017] [Indexed: 01/01/2023]
Abstract
Glioblastomas (GBMs) are high-grade brain tumors, differentially driven by alterations (amplification, deletion or missense mutations) in the epidermal growth factor receptor (EGFR), that carry a poor prognosis of just 12-15 months following standard therapy. A combination of interventions targeting tumor-specific cell surface regulators along with convergent downstream signaling pathways may enhance treatment efficacy. Against this background, we investigated a novel photoimmunotherapy approach combining the cytotoxicity of photodynamic therapy with the specificity of immunotherapy. An EGFR-specific affibody (ZEGFR:03115 ) was conjugated to the phthalocyanine dye, IR700DX, which when excited with near-infrared light produces a cytotoxic response. ZEGFR:03115 -IR700DX EGFR-specific binding was confirmed by flow cytometry and confocal microscopy. The conjugate showed effective targeting of EGFR positive GBM cells in the brain. The therapeutic potential of the conjugate was assessed both in vitro, in GBM cell lines and spheroids by the CellTiter-Glo® assay, and in vivo using subcutaneous U87-MGvIII xenografts. In addition, mice were imaged pre- and post-PIT using the IVIS/Spectrum/CT to monitor treatment response. Binding of the conjugate correlated to the level of EGFR expression in GBM cell lines. The cell proliferation assay revealed a receptor-dependent response between the tested cell lines. Inhibition of EGFRvIII+ve tumor growth was observed following administration of the immunoconjugate and irradiation. Importantly, this response was not seen in control tumors. In conclusion, the ZEGFR:03115 -IR700DX showed specific uptake in vitro and enabled imaging of EGFR expression in the orthotopic brain tumor model. Moreover, the proof-of-concept in vivo PIT study demonstrated therapeutic efficacy of the conjugate in subcutaneous glioma xenografts.
Collapse
Affiliation(s)
- Thomas A. Burley
- Division of Radiotherapy and ImagingThe Institute of Cancer ResearchLondonUnited Kingdom
| | - Justyna Mączyńska
- Division of Radiotherapy and ImagingThe Institute of Cancer ResearchLondonUnited Kingdom
| | - Anant Shah
- Division of Radiotherapy and ImagingThe Institute of Cancer ResearchLondonUnited Kingdom
| | - Wojciech Szopa
- Department of NeurosurgeryMedical University of Silesia, Regional HospitalSosnowiecPoland
| | - Kevin J. Harrington
- Division of Radiotherapy and ImagingThe Institute of Cancer ResearchLondonUnited Kingdom
| | - Jessica K.R. Boult
- Division of Radiotherapy and ImagingThe Institute of Cancer ResearchLondonUnited Kingdom
| | | | - Maria Vinci
- Department of Onco‐HematologyBambino Gesù Children's HospitalRomeItaly
| | - Jeffrey C. Bamber
- Division of Radiotherapy and ImagingThe Institute of Cancer ResearchLondonUnited Kingdom
| | - Wojciech Kaspera
- Department of NeurosurgeryMedical University of Silesia, Regional HospitalSosnowiecPoland
| | - Gabriela Kramer‐Marek
- Division of Radiotherapy and ImagingThe Institute of Cancer ResearchLondonUnited Kingdom
| |
Collapse
|
45
|
Effect of multiple cyclic RGD peptides on tumor accumulation and intratumoral distribution of IRDye 700DX-conjugated polymers. Sci Rep 2018; 8:8126. [PMID: 29802410 PMCID: PMC5970177 DOI: 10.1038/s41598-018-26593-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 05/10/2018] [Indexed: 12/20/2022] Open
Abstract
Strategic delivery of IRDye 700DX (photosensitizer) is a key for improving its effect in photodynamic therapy. In this study, we have synthesized IRDye 700DX-conjugated polymers containing multiple cyclic RGD peptides to deliver IRDye 700DX selectively to tumor cells and tumor-associated blood vessels overexpressing αvβ3 integrin. Our polymer has a backbone of hydrophilic poly(ethylene glycol)-poly(L-glutamic acid) block copolymer, and cyclic RGD peptides are conjugated to side chains of the poly(L-glutamic acid) while IRDye 700DX is conjugated to the terminal of poly(ethylene glycol). The polymers exhibited selective accumulation to the target sites in a subcutaneous solid tumor, and the accumulation was augmented with the increased number of cyclic RGD peptides. More importantly, the polymer containing 15 cyclic RGD peptides in one construct revealed preferential accumulation on the tumor-associated blood vessels without compromising penetration to deep portions of the tumor, thereby drastically inhibiting tumor growth upon photoirradiation, while the polymer containing 5 cyclic RGD peptides showed moderate antitumor activity despite efficient accumulation in the tumor with almost homogenous intratumoral distribution. These results suggest that controlling the intratumoral distribution of IRDye 700DX is critical for successful PDT, and our polymer containing multiple cyclic RGD peptides may be a promising carrier for this spatial control.
Collapse
|
46
|
Nakajima K, Kimura T, Takakura H, Yoshikawa Y, Kameda A, Shindo T, Sato K, Kobayashi H, Ogawa M. Implantable wireless powered light emitting diode (LED) for near-infrared photoimmunotherapy: device development and experimental assessment in vitro and in vivo. Oncotarget 2018; 9:20048-20057. [PMID: 29732002 PMCID: PMC5929445 DOI: 10.18632/oncotarget.25068] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Accepted: 03/21/2018] [Indexed: 01/17/2023] Open
Abstract
Purpose The aim of this study was to develop and assess a novel implantable, wireless-powered, light-emitting diode (LED) for near-infrared photoimmunotherapy (NIR-PIT). NIR-PIT is a recently developed cancer therapy that uses NIR light and antibody-photosensitizer conjugates and is able to induce cancer-specific cell death. Due to limited light penetration depth it is currently unable to treat tumors in deep tissues. Use of implanted LED might potentially overcome this limitation. Results The wireless LED system was able to emit NIR light up to a distance of 20 cm from the transmitter coil by using low magnetic fields as compliant with limits for use in humans. Results indicated that the LED system was able to kill tumor cells in vitro and to suppress tumor growth in implanted tumor-bearing mice. Conclusions Results indicated that the proposed implantable wireless LED system was able to suppress tumor growth in vivo. These results are encouraging as wireless LED systems such as the one here developed might be a possible solution to treat tumors in deep regions in humans. Further research in this area would be important. Materials and Methods An implantable LED system was developed. It consisted of a LED capsule including two LED sources and a receiver coil coupled with an external coil and power source. Wireless power transmission was guaranteed by using electromagnetic induction. The system was tested in vitro by using EGFR-expressing cells and HER2-expressing cells. The system was also tested in vivo in tumor-bearing mice.
Collapse
Affiliation(s)
- Kohei Nakajima
- Laboratory of Bioanalysis and Molecular Imaging, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Toshihiro Kimura
- Laboratory of Bioanalysis and Molecular Imaging, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, Hokkaido, Japan.,Savior, Inc., Yokohama, Kanagawa, Japan.,B and Plus K.K., Ogawamachi, Saitama, Japan
| | - Hideo Takakura
- Laboratory of Bioanalysis and Molecular Imaging, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, Hokkaido, Japan
| | | | | | | | - Kazuhide Sato
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Hisataka Kobayashi
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Mikako Ogawa
- Laboratory of Bioanalysis and Molecular Imaging, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, Hokkaido, Japan.,Presto, Japan Science and Technology Agency, Kawaguchi, Saitama, Japan
| |
Collapse
|
47
|
Chow SYS, Wong RCH, Zhao S, Lo PC, Ng DKP. Disulfide-Linked Dendritic Oligomeric Phthalocyanines as Glutathione-Responsive Photosensitizers for Photodynamic Therapy. Chemistry 2018; 24:5779-5789. [DOI: 10.1002/chem.201706128] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2017] [Indexed: 02/04/2023]
Affiliation(s)
- Sun Y. S. Chow
- Department of Chemistry; The Chinese University of Hong Kong; Shatin N.T. Hong Kong China
| | - Roy C. H. Wong
- Department of Chemistry; The Chinese University of Hong Kong; Shatin N.T. Hong Kong China
| | - Shirui Zhao
- Department of Chemistry; The Chinese University of Hong Kong; Shatin N.T. Hong Kong China
| | - Pui-Chi Lo
- Department of Biomedical Sciences; City University of Hong Kong; Tat Chee Avenue Kowloon Hong Kong China
| | - Dennis K. P. Ng
- Department of Chemistry; The Chinese University of Hong Kong; Shatin N.T. Hong Kong China
| |
Collapse
|
48
|
Interstitial near-infrared photoimmunotherapy: effective treatment areas and light doses needed for use with fiber optic diffusers. Oncotarget 2018. [PMID: 29541404 PMCID: PMC5834257 DOI: 10.18632/oncotarget.24329] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Near-infrared photoimmunotherapy (NIR-PIT), a promising cancer therapy utilizing an antibody-photoabsorber conjugate (APC) and NIR light, which induces rapid necrotic cell death only in APC-bound cells. Effective NIR-PIT in mouse models has been achieved using superficial light illumination (SLI) with light emitting diodes (LEDs) or lasers, but in the clinical setting, fiber optic diffusers have been employed to deliver light to deeper tumors. However, the performance of NIR light in tissue delivered by fiber optic diffusers is poorly understood. Here, we investigated NIR-PIT using a cylindrical fiber optic diffuser in a mouse model of A431 tumors. NIR-PIT with 100 J/cm, the same light dose used in clinical trials of NIR-PIT, was applied after insertion of the diffuser within the tumor bed, and then both bioluminescence and fluorescence imaging were analyzed to assess the therapeutic efficacy. The diffuser can deliver adequate NIR light dose for effective NIR-PIT to the A431 tumor at a distance of approximately 1 cm around the light source at 100 J/cm. At 50 J/cm NIR light effective NIR-PIT was reduced to a distance of 5 – 7 mm diameter around the light source. These results indicate that the energy of interstitial light (measured in Joules/cm) administered via a fiber diffuser determines the depth of effective NIR-PIT around the diffuser and determines the spacing at which such diffusers should be placed to entirely cover the tumor. Thermal measurements demonstrate that interstitial light for NIR-PIT does not cause damage to the skin overlying the diffuser.
Collapse
|
49
|
Chen G, Lin S, Huang D, Zhang Y, Li C, Wang M, Wang Q. Revealing the Fate of Transplanted Stem Cells In Vivo with a Novel Optical Imaging Strategy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2018; 14. [PMID: 29171718 DOI: 10.1002/smll.201702679] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 09/30/2017] [Indexed: 05/17/2023]
Abstract
Stem-cell-based regenerative medicine holds great promise in clinical practices. However, the fate of stem cells after transplantation, including the distribution, viability, and the cell clearance, is not fully understood, which is critical to understand the process and the underlying mechanism of regeneration for better therapeutic effects. Herein, we develop a dual-labeling strategy to in situ visualize the fate of transplanted stem cells in vivo by combining the exogenous near-infrared fluorescence imaging in the second window (NIR-II) and endogenous red bioluminescence imaging (BLI). The NIR-II fluorescence of Ag2 S quantum dots is employed to dynamically monitor the trafficking and distribution of all transplanted stem cells in vivo due to its deep tissue penetration and high spatiotemporal resolution, while BLI of red-emitting firefly luciferase (RfLuc) identifies the living stem cells after transplantation in vivo because only the living stem cells express RfLuc. This facile strategy allows for in situ visualization of the dynamic trafficking of stem cells in vivo and the quantitative evaluation of cell translocation and viability with high temporal and spatial resolution, and thus reports the fate of transplanted stem cells and how the living stem cells help, regeneration, for an instance, of a mouse with acute liver failure.
Collapse
Affiliation(s)
- Guangcun Chen
- CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine and i-Lab, CAS Center for Excellence in Brain Science, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
| | - Suying Lin
- CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine and i-Lab, CAS Center for Excellence in Brain Science, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
| | - Dehua Huang
- CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine and i-Lab, CAS Center for Excellence in Brain Science, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
| | - Yejun Zhang
- CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine and i-Lab, CAS Center for Excellence in Brain Science, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
- Suzhou NIR-Optics Technology Co., Ltd., Suzhou, 215124, China
| | - Chunyan Li
- CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine and i-Lab, CAS Center for Excellence in Brain Science, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
| | - Mao Wang
- CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine and i-Lab, CAS Center for Excellence in Brain Science, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
| | - Qiangbin Wang
- CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine and i-Lab, CAS Center for Excellence in Brain Science, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
- College of Materials Sciences and Opto-Electronic Technology, University of Chinese Academy of Sciences, Beijing, 100049, China
| |
Collapse
|
50
|
Almeida-Marrero V, van de Winckel E, Anaya-Plaza E, Torres T, de la Escosura A. Porphyrinoid biohybrid materials as an emerging toolbox for biomedical light management. Chem Soc Rev 2018; 47:7369-7400. [DOI: 10.1039/c7cs00554g] [Citation(s) in RCA: 136] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The present article reviews the most important developing strategies in light-induced nanomedicine, based on the combination of porphyrinoid photosensitizers with a wide variety of biomolecules and biomolecular assemblies.
Collapse
Affiliation(s)
| | | | - Eduardo Anaya-Plaza
- Departamento de Química Orgánica
- Universidad Autónoma de Madrid
- Cantoblanco 28049
- Spain
| | - Tomás Torres
- Departamento de Química Orgánica
- Universidad Autónoma de Madrid
- Cantoblanco 28049
- Spain
- Institute for Advanced Research in Chemistry (IAdChem)
| | - Andrés de la Escosura
- Departamento de Química Orgánica
- Universidad Autónoma de Madrid
- Cantoblanco 28049
- Spain
- Institute for Advanced Research in Chemistry (IAdChem)
| |
Collapse
|