1
|
Okada R, Asakage T. Near-infrared photoimmunotherapy: basics and clinical application. Jpn J Clin Oncol 2025:hyaf069. [PMID: 40319478 DOI: 10.1093/jjco/hyaf069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2025] [Accepted: 04/14/2025] [Indexed: 05/07/2025] Open
Abstract
Use of antibody-drug conjugates (ADCs) is rapidly increasing in the field of oncology. While ADCs exhibit strong and cell-selective cytotoxicity, they do not show spatial selectivity. Near-infrared photoimmunotherapy (NIR-PIT, Alluminox™) utilizes photoactivatable ADCs, that is, antibody-photoabsorber conjugates (APCs). The photoabsorber used in NIR-PIT, IRDye700DX (IR700), is activated by light of ~690 nm wavelength. APCs, usually administered by intravenous injection, bind to the target cell surface, and subsequent excitation-light illumination dramatically changes the status of IR700 from hydrophilic to hydrophobic, inducing aggregation of the APC-target molecule complex and cell burst. Dying cells release neoantigens as well as damage-associated molecular patterns, resulting in immunogenic cell death (ICD). Based on the favorable results of clinical trials, epidermal growth factor-targeted NIR-PIT has been performed in Japan since 2021 for patients with unresectable head and neck cancers (HNCs). Since pain and local edema are frequent adverse events (AEs), various measures have been taken against these AEs. Because NIR-PIT induces ICD, combining NIR-PIT with immune checkpoint inhibitor (ICI) therapy is thought to be a rather effective strategy. NIR-PIT could also locally destroy immune suppressor cells, such as regulatory T cells, in the tumor microenvironment. Currently, numerous clinical trials are under way to evaluate the efficacy of NIR-PIT as well as of combined NIR-PIT plus ICI therapy. In this review article, we describe the basics of NIT-PIT, results of translational experiments, current clinical application of NIT-PIT in HNCs, and relevant ongoing clinical trials.
Collapse
Affiliation(s)
- Ryuhei Okada
- Department of Head and Neck Surgery, Institute of Science Tokyo, Tokyo, Japan
| | - Takahiro Asakage
- Department of Head and Neck Surgery, Institute of Science Tokyo, Tokyo, Japan
| |
Collapse
|
2
|
Sato K, Okada T, Okada R, Yasui H, Yamada M, Isobe Y, Nishinaga Y, Shimizu M, Koike C, Fukushima R, Takahashi K, Taki S, Kato A, Sato M, Ogura T. Photoinduced Actin Aggregation Involves Cell Death: A Mechanism of Cancer Cell Cytotoxicity after Near-Infrared Photoimmunotherapy. ACS NANO 2025; 19:8338-8356. [PMID: 39964399 PMCID: PMC11887486 DOI: 10.1021/acsnano.5c00104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 01/28/2025] [Accepted: 01/30/2025] [Indexed: 03/05/2025]
Abstract
Near-infrared photoimmunotherapy (NIR-PIT) is a cancer treatment modality that uses antibody-photoabsorber (IR700) conjugates to destroy specific cells. The reaction between the antibody and photoabsorber is triggered by NIR-light, and this alters the shape and hydrophilicity of the conjugate. This photochemical reaction is responsible for NIR-PIT-induced cell death; however, the detailed mechanism underlying this effect remains unknown. In this study, we demonstrated that actin filaments underneath the cell membrane play an important role in NIR-PIT-induced cell death and that IR700 mediates the photochemical reaction of the conjugates, leading to actin filament aggregation upon NIR-light irradiation. The destruction of cortical actin beneath the cell plasma membrane allows water to flow into the cell based on osmotic conditions, resulting in cell rupture. This sequence of events may constitute the mechanism of NIR-PIT-induced cell death, making NIR-PIT a promising cancer treatment modality.
Collapse
Affiliation(s)
- Kazuhide Sato
- Nagoya
University Graduate School of Medicine, Nagoya 466-8550, Japan
- Nagoya
University Institute for Advanced Research, Advanced Analytical and Diagnostic Imaging Center (AADIC)/Medical
Engineering Unit (MEU), B3 Unit Frontier, Nagoya 466-8550, Japan
- CREST,
JST, Tokyo 102-8666, Japan
- Nagoya
University Institute for Advanced Research, Nagoya 464-8601, Japan
- FOREST-Souhatsu,
JST, Tokyo 102-8666, Japan
| | - Tomoko Okada
- CREST,
JST, Tokyo 102-8666, Japan
- Health and
Medical Research Institute, National Institute of Advanced Industrial
Science and Technology (AIST), Tsukuba 305-8566, Japan
| | - Ryu Okada
- Nagoya
University Graduate School of Medicine, Nagoya 466-8550, Japan
- Nagoya
University Institute for Advanced Research, Advanced Analytical and Diagnostic Imaging Center (AADIC)/Medical
Engineering Unit (MEU), B3 Unit Frontier, Nagoya 466-8550, Japan
| | - Hirotoshi Yasui
- Nagoya
University Institute for Advanced Research, Advanced Analytical and Diagnostic Imaging Center (AADIC)/Medical
Engineering Unit (MEU), B3 Unit Frontier, Nagoya 466-8550, Japan
| | - Mizuki Yamada
- Nagoya
University Institute for Advanced Research, Advanced Analytical and Diagnostic Imaging Center (AADIC)/Medical
Engineering Unit (MEU), B3 Unit Frontier, Nagoya 466-8550, Japan
- Division
of Host Defense Sciences, Dept. of Integrated Health Sciences, Nagoya University Graduate School of Medicine, Nagoya 461-8673, Japan
| | - Yoshitaka Isobe
- Nagoya
University Institute for Advanced Research, Advanced Analytical and Diagnostic Imaging Center (AADIC)/Medical
Engineering Unit (MEU), B3 Unit Frontier, Nagoya 466-8550, Japan
| | - Yuko Nishinaga
- Nagoya
University Institute for Advanced Research, Advanced Analytical and Diagnostic Imaging Center (AADIC)/Medical
Engineering Unit (MEU), B3 Unit Frontier, Nagoya 466-8550, Japan
| | - Misae Shimizu
- Nagoya
University Institute for Advanced Research, Advanced Analytical and Diagnostic Imaging Center (AADIC)/Medical
Engineering Unit (MEU), B3 Unit Frontier, Nagoya 466-8550, Japan
| | - Chiaki Koike
- Nagoya
University Institute for Advanced Research, Advanced Analytical and Diagnostic Imaging Center (AADIC)/Medical
Engineering Unit (MEU), B3 Unit Frontier, Nagoya 466-8550, Japan
| | - Rika Fukushima
- Nagoya
University Institute for Advanced Research, Advanced Analytical and Diagnostic Imaging Center (AADIC)/Medical
Engineering Unit (MEU), B3 Unit Frontier, Nagoya 466-8550, Japan
- Division
of Host Defense Sciences, Dept. of Integrated Health Sciences, Nagoya University Graduate School of Medicine, Nagoya 461-8673, Japan
| | - Kazuomi Takahashi
- Nagoya
University Institute for Advanced Research, Advanced Analytical and Diagnostic Imaging Center (AADIC)/Medical
Engineering Unit (MEU), B3 Unit Frontier, Nagoya 466-8550, Japan
| | - Shunichi Taki
- Nagoya
University Institute for Advanced Research, Advanced Analytical and Diagnostic Imaging Center (AADIC)/Medical
Engineering Unit (MEU), B3 Unit Frontier, Nagoya 466-8550, Japan
| | - Ayako Kato
- Nagoya
University Institute for Advanced Research, Advanced Analytical and Diagnostic Imaging Center (AADIC)/Medical
Engineering Unit (MEU), B3 Unit Frontier, Nagoya 466-8550, Japan
| | - Mitsuo Sato
- Division
of Host Defense Sciences, Dept. of Integrated Health Sciences, Nagoya University Graduate School of Medicine, Nagoya 461-8673, Japan
| | - Toshihiko Ogura
- CREST,
JST, Tokyo 102-8666, Japan
- Health and
Medical Research Institute, National Institute of Advanced Industrial
Science and Technology (AIST), Tsukuba 305-8566, Japan
| |
Collapse
|
3
|
Wu T, Wu S, Gao H, Liu H, Feng J, Yin G. Astragaloside IV augments anti-PD-1 therapy to suppress tumor growth in lung cancer by remodeling the tumor microenvironment. Eur J Histochem 2024; 68:4098. [PMID: 39440587 PMCID: PMC11558310 DOI: 10.4081/ejh.2024.4098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Accepted: 08/22/2024] [Indexed: 10/25/2024] Open
Abstract
Programmed cell death protein-1 (PD-1) inhibitors are increasingly utilized in the treatment of lung cancer (LC). Combination therapy has recently gained popularity in treating LC. This study aimed to assess the efficacy of combining Astragaloside IV (AS-IV) and anti-PD-1 in LC. C57BL/6J mice were subcutaneously injected with Lewis lung carcinoma (LLC) cells. After 3 weeks, the animals were sacrificed, and the tumors were harvested for analysis. Ki-67 immuno-labeling and TUNEL assay were used for evaluating cell proliferation and apoptosis in tumor tissues. In addition, anti-cleaved caspase 3 was used for immunolabelling of apoptotic cells. Immune cell infiltration (macrophages and T cells) and gene expression in tumor tissues were also investigated by using immunofluorescence staining. Compared to treatment with anti-PD-1 or AS-IV, the combination of AS-IV and anti-PD-1 notably reduced tumor volume and weight of LLC-bearing mice. Additionally, the combination treatment strongly induced the apoptosis and suppressed the proliferation in tumor tissues through inactivating PI3K/Akt and ERK signaling pathways, compared to single treatment group. Moreover, the combination treatment elevated levels of the M1 macrophage marker mCD86, reduced levels of the M2 macrophage marker mCD206, as well as upregulated levels of the T cell activation marker mCD69 in tumor tissues. Collectively, the combination treatment effectively inhibited tumor growth in LLC mice through promoting M1 macrophage polarization and T cell activation. These findings showed that combining AS-IV with anti-PD-1 therapy could be a promising therapeutic approach for LC.
Collapse
Affiliation(s)
- Tao Wu
- Department of Oncology, The First Affiliated Hospital of Hunan University of Traditional Chinese Medicine, Zhuzhou.
| | - Shikui Wu
- Department of Oncology, The First Affiliated Hospital of Hunan University of Traditional Chinese Medicine, Zhuzhou.
| | - Hui Gao
- Department of Oncology, The First Affiliated Hospital of Hunan University of Traditional Chinese Medicine, Zhuzhou.
| | - Haolei Liu
- Department of Oncology, The First Affiliated Hospital of Hunan University of Traditional Chinese Medicine, Zhuzhou.
| | - Jun Feng
- Department of Oncology, The First Affiliated Hospital of Hunan University of Traditional Chinese Medicine, Zhuzhou.
| | - Ge Yin
- Department of Oncology, The First Affiliated Hospital of Hunan University of Traditional Chinese Medicine, Zhuzhou.
| |
Collapse
|
4
|
Sari G, Dhatchinamoorthy K, Orellano-Ariza L, Ferreira LM, Brehm MA, Rock K. IRF2 loss is associated with reduced MHC I pathway transcripts in subsets of most human cancers and causes resistance to checkpoint immunotherapy in human and mouse melanomas. J Exp Clin Cancer Res 2024; 43:276. [PMID: 39354629 PMCID: PMC11446056 DOI: 10.1186/s13046-024-03187-5] [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: 06/25/2024] [Accepted: 09/07/2024] [Indexed: 10/03/2024] Open
Abstract
BACKGROUND In order for cancers to progress, they must evade elimination by CD8 T cells or other immune mechanisms. CD8 T cells recognize and kill tumor cells that display immunogenic tumor peptides bound to MHC I molecules. One of the ways that cancers can escape such killing is by reducing expression of MHC I molecules, and loss of MHC I is frequently observed in tumors. There are multiple different mechanisms that can underly the loss of MHC I complexes on tumor and it is currently unclear whether there are particular mechanisms that occur frequently and, if so, in what types of cancers. Also of importance to know is whether the loss of MHC I is reversible and how such loss and/or its restoration would impact responses to immunotherapy. Here, we investigate these issues for loss of IRF1 and IRF2, which are transcription factors that drive expression of MHC I pathway genes and some killing mechanisms. METHODS Bioinformatics analyses of IRF2 and IRF2-dependent gene transcripts were performed for all human cancers in the TCGA RNAseq database. IRF2 protein-DNA-binding was analyzed in ChIPseq databases. CRISRPcas9 was used to knock out IRF1 and IRF2 genes in human and mouse melanoma cells and the resulting phenotypes were analyzed in vitro and in vivo. RESULTS Transcriptomic analysis revealed that IRF2 expression was reduced in a substantial subset of cases in almost all types of human cancers. When this occurred there was a corresponding reduction in the expression of IRF2-regulated genes that were needed for CD8 T cell recognition. To test cause and effect for these IRF2 correlations and the consequences of IRF2 loss, we gene-edited IRF2 in a patient-derived melanoma and a mouse melanoma. The IRF2 gene-edited melanomas had reduced expression of transcripts for genes in the MHC I pathway and decreased levels of MHC I complexes on the cell surface. Levels of Caspase 7, an IRF2 target gene involved in CD8 T cell killing of tumors, were also reduced. This loss of IRF2 caused both human and mouse melanomas to become resistant to immunotherapy with a checkpoint inhibitor. Importantly, these effects were reversible. Stimulation of the IRF2-deficient melanomas with interferon induced the expression of a functionally homologous transcription factor, IRF1, which then restored the MHC I pathway and responsiveness to CPI. CONCLUSIONS Our study shows that a subset of cases within most types of cancers downregulates IRF2 and that this can allow cancers to escape immune control. This can cause resistance to checkpoint blockade immunotherapy and is reversible with currently available biologics.
Collapse
Affiliation(s)
- G Sari
- Department of Pathology, UMass Chan Medical School, 55 Lake Avenue North, Worcester, MA, 01655, USA
| | - K Dhatchinamoorthy
- Department of Pathology, UMass Chan Medical School, 55 Lake Avenue North, Worcester, MA, 01655, USA
| | - L Orellano-Ariza
- Department of Pathology, UMass Chan Medical School, 55 Lake Avenue North, Worcester, MA, 01655, USA
| | - L M Ferreira
- Program in Molecular Medicine, Diabetes Center of Excellence, UMass Chan Medical School, Worcester, MA, USA
| | - M A Brehm
- Program in Molecular Medicine, Diabetes Center of Excellence, UMass Chan Medical School, Worcester, MA, USA
| | - K Rock
- Department of Pathology, UMass Chan Medical School, 55 Lake Avenue North, Worcester, MA, 01655, USA.
| |
Collapse
|
5
|
Inagaki FF, Kano M, Furusawa A, Kato T, Okada R, Fukushima H, Takao S, Okuyama S, Choyke PL, Kobayashi H. Near-infrared photoimmunotherapy targeting PD-L1: Improved efficacy by preconditioning the tumor microenvironment. Cancer Sci 2024; 115:2396-2409. [PMID: 38671582 PMCID: PMC11247602 DOI: 10.1111/cas.16195] [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: 12/21/2023] [Revised: 04/04/2024] [Accepted: 04/09/2024] [Indexed: 04/28/2024] Open
Abstract
Near-infrared photoimmunotherapy (NIR-PIT) is a new type of cancer therapy that employs antibody-IRDye700DX conjugates (AbPCs) and near-infrared (NIR) light at a wavelength of 689 nm, the excitation wavelength of IR700. Administered intravenously, injected AbPCs bind specifically to cells expressing the target antigen, whereupon NIR light exposure causes rapid, selective killing. This process induces an anticancer T cell response, leading to sustained anticancer host immune response. Programmed cell death ligand-1 (PD-L1) is a major inhibitory immune checkpoint molecule expressed in various cancers. In this study, we first assessed the efficacy of PD-L1-targeted NIR-PIT (αPD-L1-PIT) in immune-competent tumor mouse models. αPD-L1-PIT showed a significant therapeutic effect on the tumor models with high PD-L1 expression. Furthermore, αPD-L1-PIT induced an abscopal effect on distant tumors and long-term immunological memory. In contrast, αPD-L1-PIT was not as effective for tumor models with low PD-L1 expression. To improve the efficacy of PD-L1-targeted NIR-PIT, PEGylated interferon-gamma (IFNγ) was administered with αPD-L1-PIT. The combination therapy improved the treatment efficacy by increasing PD-L1 expression leading to more efficient cell killing by αPD-L1-PIT. Furthermore, the PEGylated IFNγ led to a CD8+ T cell-dominant tumor microenvironment (TME) with an enhanced anticancer T cell response after αPD-L1-PIT. As a result, even so-called cold tumors exhibited complete responses after αPD-L1-PIT. Thus, combination therapy of PEGylated IFNγ and PD-L1-targeted NIR-PIT has the potential to be an important future strategy for cancer immunotherapy.
Collapse
Affiliation(s)
- Fuyuki F Inagaki
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Makoto Kano
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Aki Furusawa
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Takuya Kato
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Ryuhei Okada
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Hiroshi Fukushima
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Seiichiro Takao
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Shuhei Okuyama
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Peter L Choyke
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Hisataka Kobayashi
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
6
|
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] [Grants] [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
|
7
|
Kumai T, Shinomiya H, Shibata H, Takahashi H, Kishikawa T, Okada R, Fujieda S, Sakashita M. Translational research in head and neck cancer: Molecular and immunological updates. Auris Nasus Larynx 2024; 51:391-400. [PMID: 37640594 DOI: 10.1016/j.anl.2023.08.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 08/17/2023] [Accepted: 08/21/2023] [Indexed: 08/31/2023]
Abstract
Head and neck squamous cell carcinoma (HNSCC) has a poor prognosis. Each year, approximately 880,000 patients are newly diagnosed with HNSCC worldwide, and 450,000 patients with HNSCC die. Risk factors for developing HNSCC have been identified, with cigarette smoking, alcohol consumption, and viral infections being the major factors. Owing to the prevalence of human papillomavirus infection, the number of HNSCC cases is increasing considerably. Surgery and chemoradiotherapy are the primary treatments for HNSCC. With advancements in tumor biology, patients are eligible for novel treatment modalities, namely targeted therapies, immunotherapy, and photoimmunotherapy. Because this area of research has rapidly progressed, clinicians should understand the basic biology of HNSCC to choose an appropriate therapy in the upcoming era of personalized medicine. This review summarized recent developments in tumor biology, focusing on epidemiology, genetic/epigenetic factors, the tumor microenvironment, microbiota, immunity, and photoimmunotherapy in HNSCC, as well as how these findings can be translated into clinical settings.
Collapse
Affiliation(s)
- Takumi Kumai
- Department of Otolaryngology-Head and Neck Surgery, Asahikawa Medical University, Midorigaoka-Higashi 2-1-1-1, Asahikawa 078-8510, Japan.
| | - Hirotaka Shinomiya
- Department of Otolaryngology-Head and Neck Surgery, Kobe University Graduate School of Medicine, Kobe, Japan.
| | - Hirofumi Shibata
- Department of Otolaryngology-Head and Neck Surgery, Gifu University Graduate School of Medicine, Gifu, Japan.
| | - Hideaki Takahashi
- Department of Otorhinolaryngology, Head and Neck Surgery, School of Medicine, Yokohama City University, Yokohama, Japan.
| | - Toshihiro Kishikawa
- Department of Head and Neck Surgery, Aichi Cancer Center Hospital, Nagoya, Japan.
| | - Ryuhei Okada
- Department of Head and Neck Surgery, Tokyo Medical and Dental University, Tokyo, Japan.
| | - Shigeharu Fujieda
- Department of Otorhinolaryngology-Head and Neck Surgery, Faculty of Medical Sciences, University of Fukui, Fukui, Japan.
| | - Masafumi Sakashita
- Department of Otorhinolaryngology-Head and Neck Surgery, Faculty of Medical Sciences, University of Fukui, Fukui, Japan.
| |
Collapse
|
8
|
Delcanale P, Alampi MM, Mussini A, Fumarola C, Galetti M, Petronini PG, Viappiani C, Bruno S, Abbruzzetti S. A Photoactive Supramolecular Complex Targeting PD-L1 Reveals a Weak Correlation between Photoactivation Efficiency and Receptor Expression Levels in Non-Small-Cell Lung Cancer Tumor Models. Pharmaceutics 2023; 15:2776. [PMID: 38140116 PMCID: PMC10747218 DOI: 10.3390/pharmaceutics15122776] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 12/04/2023] [Accepted: 12/06/2023] [Indexed: 12/24/2023] Open
Abstract
Photo-immunotherapy uses antibodies conjugated to photosensitizers to produce nanostructured constructs endowed with targeting properties and photo-inactivation capabilities towards tumor cells. The superficial receptor density on cancer cells is considered a determining factor for the efficacy of the photodynamic treatment. In this work, we propose the use of a photoactive conjugate that consists of the clinical grade PD-L1-binding monoclonal antibody Atezolizumab, covalently linked to either the well-known photosensitizer eosin or the fluorescent probe Alexa647. Using single-molecule localization microscopy (direct stochastic optical reconstruction microscopy, dSTORM), and an anti-PD-L1 monoclonal antibody labelled with Alexa647, we quantified the density of PD-L1 receptors exposed on the cell surface in two human non-small-cell lung cancer lines (H322 and A549) expressing PD-L1 to a different level. We then investigated if this value correlates with the effectiveness of the photodynamic treatment. The photodynamic treatment of H322 and A549 with the photo-immunoconjugate demonstrated its potential for PDT treatments, but the efficacy did not correlate with the PD-L1 expression levels. Our results provide additional evidence that receptor density does not determine a priori the level of photo-induced cell death.
Collapse
Affiliation(s)
- Pietro Delcanale
- Department of Mathematical, Physical and Computer Sciences, University of Parma, 43124 Parma, Italy; (P.D.); (M.M.A.); (A.M.); (C.V.)
| | - Manuela Maria Alampi
- Department of Mathematical, Physical and Computer Sciences, University of Parma, 43124 Parma, Italy; (P.D.); (M.M.A.); (A.M.); (C.V.)
| | - Andrea Mussini
- Department of Mathematical, Physical and Computer Sciences, University of Parma, 43124 Parma, Italy; (P.D.); (M.M.A.); (A.M.); (C.V.)
| | - Claudia Fumarola
- Department of Medicine and Surgery, University of Parma, 43125 Parma, Italy; (C.F.); (P.G.P.)
| | - Maricla Galetti
- Department of Occupational and Environmental Medicine, Epidemiology and Hygiene, INAIL-Italian Workers’ Compensation Authority, 00078 Rome, Italy;
| | - Pier Giorgio Petronini
- Department of Medicine and Surgery, University of Parma, 43125 Parma, Italy; (C.F.); (P.G.P.)
| | - Cristiano Viappiani
- Department of Mathematical, Physical and Computer Sciences, University of Parma, 43124 Parma, Italy; (P.D.); (M.M.A.); (A.M.); (C.V.)
| | - Stefano Bruno
- Department of Food and Drug, University of Parma, 43124 Parma, Italy;
| | - Stefania Abbruzzetti
- Department of Mathematical, Physical and Computer Sciences, University of Parma, 43124 Parma, Italy; (P.D.); (M.M.A.); (A.M.); (C.V.)
| |
Collapse
|
9
|
Li K, Yang D, Liu D. Targeted Nanophotoimmunotherapy Potentiates Cancer Treatment by Enhancing Tumor Immunogenicity and Improving the Immunosuppressive Tumor Microenvironment. Bioconjug Chem 2023; 34:283-301. [PMID: 36648963 DOI: 10.1021/acs.bioconjchem.2c00593] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Cancer immunotherapy, such as immune checkpoint blockade, chimeric antigen receptor, and cytokine therapy, has emerged as a robust therapeutic strategy activating the host immune system to inhibit primary and metastatic lesions. However, low tumor immunogenicity (LTI) and immunosuppressive tumor microenvironment (ITM) severely compromise the killing effect of immune cells on tumor cells, which fail to evoke a strong and effective immune response. As an exogenous stimulation therapy, phototherapy can induce immunogenic cell death (ICD), enhancing the therapeutic effect of tumor immunotherapy. However, the lack of tumor targeting and the occurrence of immune escape significantly reduce its efficacy in vivo, thus limiting its clinical application. Nanophotoimmunotherapy (nano-PIT) is a precision-targeted tumor treatment that co-loaded phototherapeutic agents and various immunotherapeutic agents by specifically targeted nanoparticles (NPs) to improve the effectiveness of phototherapy, reduce its phototoxicity, enhance tumor immunogenicity, and reverse the ITM. This review will focus on the theme of nano-PIT, introduce the current research status of nano-PIT on converting "cold" tumors to "hot" tumors to improve immune efficacy according to the classification of immunotherapy targets, and discuss the challenges, opportunities, and prospects.
Collapse
Affiliation(s)
- Kunwei Li
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, 127 West Youyi Road, Xi'an, Shaanxi 710072, China
| | - Dan Yang
- Department of Pharmaceutical Sciences, School of Biological and Pharmaceutical Sciences, Shaanxi University of Science and Technology, Weiyang University Park, Xi'an 710021, China
| | - Dechun Liu
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, 127 West Youyi Road, Xi'an, Shaanxi 710072, China
| |
Collapse
|
10
|
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
|
11
|
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: 23] [Impact Index Per Article: 11.5] [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
|
12
|
Yin X, Cheng Y, Feng Y, Stiles WR, Park SH, Kang H, Choi HS. Phototheranostics for multifunctional treatment of cancer with fluorescence imaging. Adv Drug Deliv Rev 2022; 189:114483. [PMID: 35944585 PMCID: PMC9860309 DOI: 10.1016/j.addr.2022.114483] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 07/06/2022] [Accepted: 08/03/2022] [Indexed: 01/25/2023]
Abstract
Phototheranostics stem from the recent advances in nanomedicines and bioimaging to diagnose and treat human diseases. Since tumors' diversity, heterogeneity, and instability limit the clinical application of traditional diagnostics and therapeutics, phototheranostics, which combine light-induced therapeutic and diagnostic modalities in a single platform, have been widely investigated. Numerous efforts have been made to develop phototheranostics for efficient light-induced antitumor therapeutics with minimal side effects. Herein, we review the fundamentals of phototheranostic nanomedicines with their biomedical applications. Furthermore, the progress of near-infrared fluorescence imaging and cancer treatments, including photodynamic therapy and photothermal therapy, along with chemotherapy, immunotherapy, and gene therapy, are summarized. This review also discusses the opportunities and challenges associated with the clinical translation of phototheranostics in pan-cancer research. Phototheranostics can pave the way for future research, improve the quality of life, and prolong cancer patients' survival times.
Collapse
Affiliation(s)
- Xiaoran Yin
- Department of Oncology, The Second Affiliate Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi 710004, China,Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Yifan Cheng
- Department of Oncology, The Second Affiliate Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi 710004, China
| | - Yan Feng
- Department of Oncology, The Second Affiliate Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi 710004, China
| | - Wesley R. Stiles
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Seung Hun Park
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Homan Kang
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA,Corresponding authors at: 149 13th Street, Boston, MA 02129, USA., (H. Kang), (H.S. Choi)
| | - Hak Soo Choi
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA,Corresponding authors at: 149 13th Street, Boston, MA 02129, USA., (H. Kang), (H.S. Choi)
| |
Collapse
|
13
|
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
|
14
|
Zanello A, Bortolotti M, Maiello S, Bolognesi A, Polito L. Anti-PD-L1 immunoconjugates for cancer therapy: Are available antibodies good carriers for toxic payload delivering? Front Pharmacol 2022; 13:972046. [PMID: 36052121 PMCID: PMC9424723 DOI: 10.3389/fphar.2022.972046] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 07/19/2022] [Indexed: 12/15/2022] Open
Abstract
Immune checkpoint mechanisms are important molecular cell systems that maintain tolerance toward autoantigens in order to prevent immunity-mediated accidental damage. It is well known that cancer cells may exploit these molecular and cellular mechanisms to escape recognition and elimination by immune cells. Programmed cell death protein-1 (PD-1) and its natural ligand programmed cell death ligand-1 (PD-L1) form the PD-L1/PD-1 axis, a well-known immune checkpoint mechanism, which is considered an interesting target in cancer immunotherapy. In fact, the expression of PD-L1 was found in various solid malignancies and the overactivation of PD-L1/PD-1 axis results in a poor patient survival rate. Breaking PD-L1/PD-1 axis, by blocking either the cancer side or the immune side of the axis, is currently used as anti-cancer strategy to re-establish a tumor-specific immune response. For this purpose, several blocking antibodies are now available. To date, three anti-PD-L1 antibodies have been approved by the FDA, namely atezolizumab, durvalumab and avelumab. The main advantages of anti-PD-L1 antibodies arise from the overexpression of PD-L1 antigen by a high number of tumor cells, also deriving from different tissues; this makes anti-PD-L1 antibodies potential pan-specific anti-cancer molecules. Despite the good results reported in clinical trials with anti-PD-L1 antibodies, there is a significant number of patients that do not respond to the therapy. In fact, it should be considered that, in some neoplastic patients, reduced or absent infiltration of cytotoxic T cells and natural killer cells in the tumor microenvironment or presence of other immunosuppressive molecules make immunotherapy with anti-PD-L1 blocking antibodies less effective. A strategy to improve the efficacy of antibodies is to use them as carriers for toxic payloads (toxins, drugs, enzymes, radionuclides, etc.) to form immunoconjugates. Several immunoconjugates have been already approved by FDA for treatment of malignancies. In this review, we focused on PD-L1 targeting antibodies utilized as carrier to construct immunoconjugates for the potential elimination of neoplastic cells, expressing PD-L1. A complete examination of the literature regarding anti-PD-L1 immunoconjugates is here reported, describing the results obtained in vitro and in vivo. The real potential of anti-PD-L1 antibodies as carriers for toxic payload delivery is considered and extensively discussed.
Collapse
|
15
|
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
|
16
|
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
|
17
|
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
|