1
|
Prichard K, Chau N, Xue J, Krauss M, Sakoff JA, Gilbert J, Bahnik C, Muehlbauer M, Radetzki S, Robinson PJ, Haucke V, McCluskey A. Inhibition Clathrin Mediated Endocytosis: Pitstop 1 and Pitstop 2 Chimeras. ChemMedChem 2024; 19:e202400253. [PMID: 38894585 DOI: 10.1002/cmdc.202400253] [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: 04/08/2024] [Revised: 06/18/2024] [Accepted: 06/18/2024] [Indexed: 06/21/2024]
Abstract
Twenty-five chimera compounds of Pitstop 1 and 2 were synthesised and screened for their ability to block the clathrin terminal domain-amphiphysin protein-protein interaction (NTD-PPI using an ELISA) and clathrin mediated endocytosis (CME) in cells. Library 1 was based on Pitstop 2, but no notable clathrin PPI or in-cell activity was observed. With the Pitstop 1, 16 analogues were produced with 1,8-naphthalic imide core as a foundation. Analogues with methylene spaced linkers and simple amides showed a modest to good range of PPI inhibition (7.6-42.5 μM, naphthyl 39 and 4-nitrophenyl 40 respectively) activity. These data reveal the importance of the naphthalene sulfonate moiety, with no des-SO3 analogue displaying PPI inhibition. This was consistent with the observed analogue docked poses within the clathrin terminal domain Site 1 binding pocket. Further modifications targeted the naphthalene imide moiety, with the installation of 5-Br (45 a), 5-OH (45 c) and 5-propyl ether (45 d) moieties. Among them, the OH 45 c and propyl ether 45 d retained PPI inhibition, with propyl ether 45 d being the most active with a PPI inhibition IC50=7.3 μM. This is 2x more potent than Pitstop 2 and 3x more potent than Pitstop 1.
Collapse
Affiliation(s)
- Kate Prichard
- Chemistry, School of Environmental & Life Sciences, The University of Newcastle, University Drive, Callaghan, NSW, 2308, Australia
| | - Ngoc Chau
- Cell Signalling Unit, Children's Medical Research Institute, The University of Sydney, Hawkesbury Road, Westmead, Sydney, Australia
| | - Jing Xue
- Cell Signalling Unit, Children's Medical Research Institute, The University of Sydney, Hawkesbury Road, Westmead, Sydney, Australia
| | - Michael Krauss
- Leibniz Institute fur Molecular Pharmacologie, Department of Biology, Chemistry, Pharmacy, Robert-Roessle-Strasse 10, Berlin, 13125, Germany
| | - Jennette A Sakoff
- Experimental Therapeutics Group, Medical Oncology, Calvary Mater Newcastle Hospital, Edith Street, Waratah, NSW, 2298, Australia
| | - Jayne Gilbert
- Experimental Therapeutics Group, Medical Oncology, Calvary Mater Newcastle Hospital, Edith Street, Waratah, NSW, 2298, Australia
| | - Claudia Bahnik
- Leibniz Institute fur Molecular Pharmacologie, Department of Biology, Chemistry, Pharmacy, Robert-Roessle-Strasse 10, Berlin, 13125, Germany
| | - Maria Muehlbauer
- Leibniz Institute fur Molecular Pharmacologie, Department of Biology, Chemistry, Pharmacy, Robert-Roessle-Strasse 10, Berlin, 13125, Germany
| | - Silke Radetzki
- Leibniz Institute fur Molecular Pharmacologie, Department of Biology, Chemistry, Pharmacy, Robert-Roessle-Strasse 10, Berlin, 13125, Germany
| | - Phillip J Robinson
- Cell Signalling Unit, Children's Medical Research Institute, The University of Sydney, Hawkesbury Road, Westmead, Sydney, Australia
| | - Volker Haucke
- Leibniz Institute fur Molecular Pharmacologie, Department of Biology, Chemistry, Pharmacy, Robert-Roessle-Strasse 10, Berlin, 13125, Germany
| | - Adam McCluskey
- Chemistry, School of Environmental & Life Sciences, The University of Newcastle, University Drive, Callaghan, NSW, 2308, Australia
| |
Collapse
|
2
|
Liu S, Lan B, Wang Y, Yang T, Li L, Ge H, Zeng C, Xu B, Qian H, Ma F. Pyrotinib and trastuzumab combination treatment synergistically overcomes HER2 dependency in HER2-positive breast cancer: insights from the PHILA trial. EBioMedicine 2024; 109:105379. [PMID: 39368454 PMCID: PMC11489075 DOI: 10.1016/j.ebiom.2024.105379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 09/19/2024] [Accepted: 09/19/2024] [Indexed: 10/07/2024] Open
Abstract
BACKGROUND The PHILA study suggests that pyrotinib, trastuzumab, and docetaxel significantly improved progression-free survival (PFS) compared with placebo, trastuzumab, and docetaxel in patients with untreated HER2-positive metastatic breast cancer. In this study, we aimed to investigate the synergistic mechanisms of pyrotinib plus trastuzumab and provide further insights for the PHILA trial. METHODS The in vitro activity of combination treatments was assessed through cell biological and biochemical experiments. The in vivo efficacy was evaluated in cell-derived xenografts, a TUBO tumour model, and one clinical case. Next-generation sequencing was performed on circulating tumour DNA (ctDNA) from patients in the PHILA trial. FINDINGS The combination of pyrotinib and trastuzumab more effectively inhibited cell growth than pyrotinib or trastuzumab alone in models of HER2-dependent breast cancer. It potentiated membrane HER2 ubiquitination and downregulation, which resulted in a comprehensive blockade of the HER2 signalling pathway. The pyrotinib-altered membrane HER2 levels had no significant effect on trastuzumab-mediated antibody-dependent cell-mediated cytotoxicity (ADCC). We further validated the synergistic mechanisms in TUBO tumours and one clinical case, rather than models of HCC1954 cells harbouring the PIK3CA H1047R mutation. Similarly, in our centre cohort of the PHILA study, patients with genetic alterations in the HER2 signalling cascade had significantly shorter median PFS than individuals with the wild-type pathway. INTERPRETATION Our findings underscore the robust synergy between pyrotinib and trastuzumab in overcoming HER2 dependency and provide a rationale for pyrotinib, trastuzumab, and docetaxel as one of the optimal choices for patients with untreated HER2-positive metastatic breast cancer, who are dependent on the HER2 signalling cascade. FUNDING This work was supported by the National Key Research and Development Program of China (2021YFF1201300), the National Natural Science Foundation of China (82172875), the CAMS Innovation Fund for Medical Sciences (CIFMS) (2022-I2M-2-001), and the Joint Innovative Fund of Beijing Natural Science Foundation and Changping District (L234004).
Collapse
Affiliation(s)
- Shuning Liu
- Department of Medical Oncology, National Cancer Centre/National Clinical Research Centre for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China; State Key Laboratory of Molecular Oncology, National Cancer Centre/National Clinical Research Centre for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Bo Lan
- Department of Medical Oncology, National Cancer Centre/National Clinical Research Centre for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Yuanyi Wang
- Department of Medical Oncology, National Cancer Centre/National Clinical Research Centre for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China; State Key Laboratory of Molecular Oncology, National Cancer Centre/National Clinical Research Centre for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Tao Yang
- Department of Medical Oncology, National Cancer Centre/National Clinical Research Centre for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China; State Key Laboratory of Molecular Oncology, National Cancer Centre/National Clinical Research Centre for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Lixi Li
- Department of Medical Oncology, National Cancer Centre/National Clinical Research Centre for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China; State Key Laboratory of Molecular Oncology, National Cancer Centre/National Clinical Research Centre for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Hewei Ge
- Department of Medical Oncology, National Cancer Centre/National Clinical Research Centre for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China; State Key Laboratory of Molecular Oncology, National Cancer Centre/National Clinical Research Centre for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Cheng Zeng
- Department of Medical Oncology, National Cancer Centre/National Clinical Research Centre for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China; State Key Laboratory of Molecular Oncology, National Cancer Centre/National Clinical Research Centre for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Binghe Xu
- Department of Medical Oncology, National Cancer Centre/National Clinical Research Centre for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China; State Key Laboratory of Molecular Oncology, National Cancer Centre/National Clinical Research Centre for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China.
| | - Haili Qian
- State Key Laboratory of Molecular Oncology, National Cancer Centre/National Clinical Research Centre for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China.
| | - Fei Ma
- Department of Medical Oncology, National Cancer Centre/National Clinical Research Centre for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China; State Key Laboratory of Molecular Oncology, National Cancer Centre/National Clinical Research Centre for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China.
| |
Collapse
|
3
|
Simoni-Nieves A, Lindzen M, Giri S, Gupta N, Chatterjee R, Selvadurai BR, Van Daele M, Love D, Haga Y, Romaniello D, Salame TM, Zerbib M, Oren R, Tsutsumi Y, Lauriola M, Marrocco I, Yarden Y. A bispecific antibody targeting EGFR and AXL delays resistance to osimertinib. Cell Rep Med 2024; 5:101703. [PMID: 39216477 PMCID: PMC11528239 DOI: 10.1016/j.xcrm.2024.101703] [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: 12/18/2023] [Revised: 06/19/2024] [Accepted: 08/07/2024] [Indexed: 09/04/2024]
Abstract
Activating EGFR (epidermal growth factor receptor) mutations can be inhibited by specific tyrosine kinase inhibitors (TKIs), which have changed the landscape of lung cancer therapy. However, due to secondary mutations and bypass receptors, such as AXL (AXL receptor tyrosine kinase), drug resistance eventually emerges in most patients treated with the first-, second-, or third-generation TKIs (e.g., osimertinib). To inhibit AXL and resistance to osimertinib, we compare two anti-AXL drugs, an antibody (mAb654) and a TKI (bemcentinib). While no pair of osimertinib and an anti-AXL drug is able to prevent relapses, triplets combining osimertinib, cetuximab (an anti-EGFR antibody), and either anti-AXL drug are initially effective. However, longer monitoring uncovers superiority of the mAb654-containing triplet, possibly due to induction of receptor endocytosis, activation of immune mechanisms, or disabling intrinsic mutators. Hence, we constructed a bispecific antibody that engages both AXL and EGFR. When combined with osimertinib, the bispecific antibody consistently inhibits tumor relapses, which warrants clinical trials.
Collapse
Affiliation(s)
- Arturo Simoni-Nieves
- Departments of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Moshit Lindzen
- Departments of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Suvendu Giri
- Departments of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Nitin Gupta
- Departments of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Rishita Chatterjee
- Departments of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Boobash-Raj Selvadurai
- Departments of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Marieke Van Daele
- Departments of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Danielle Love
- Departments of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Yuya Haga
- Graduate School of Pharmaceutical Sciences, Osaka University, Osaka 565-0871, Japan
| | - Donatella Romaniello
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40126 Bologna, Italy
| | - Tomer-Meir Salame
- Flow Cytometry Unit, Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Mirie Zerbib
- Department of Veterinary Resources, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Roni Oren
- Department of Veterinary Resources, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Yasuo Tsutsumi
- Graduate School of Pharmaceutical Sciences, Osaka University, Osaka 565-0871, Japan; Global Center for Medical Engineering and Informatics, Osaka University, Osaka 565-0871, Japan; Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Osaka 565-0871, Japan
| | - Mattia Lauriola
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40126 Bologna, Italy
| | - Ilaria Marrocco
- Department of Life Sciences and Public Health, Università Cattolica del Sacro Cuore, 00168 Rome, Italy.
| | - Yosef Yarden
- Departments of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot 76100, Israel.
| |
Collapse
|
4
|
Chan ET, Kural C. Targeting endocytosis to sensitize cancer cells to programmed cell death. Biochem Soc Trans 2024; 52:1703-1713. [PMID: 39092762 PMCID: PMC11519968 DOI: 10.1042/bst20231332] [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: 04/24/2024] [Revised: 07/01/2024] [Accepted: 07/18/2024] [Indexed: 08/04/2024]
Abstract
Evading programmed cell death (PCD) is a hallmark of cancer that allows tumor cells to survive and proliferate unchecked. Endocytosis, the process by which cells internalize extracellular materials, has emerged as a key regulator of cell death pathways in cancer. Many tumor types exhibit dysregulated endocytic dynamics that fuel their metabolic demands, promote resistance to cytotoxic therapies, and facilitate immune evasion. This review examines the roles of endocytosis in apoptotic resistance and immune escape mechanisms utilized by cancer cells. We highlight how inhibiting endocytosis can sensitize malignant cells to therapeutic agents and restore susceptibility to PCD. Strategies to modulate endocytosis for enhanced cancer treatment are discussed, including targeting endocytic regulatory proteins, altering membrane biophysical properties, and inhibiting Rho-associated kinases. While promising, challenges remain regarding the specificity and selectivity of endocytosis-targeting agents. Nonetheless, harnessing endocytic pathways represents an attractive approach to overcome apoptotic resistance and could yield more effective therapies by rendering cancer cells vulnerable to PCD. Understanding the interplay between endocytosis and PCD regulation is crucial for developing novel anticancer strategies that selectively induce tumor cell death.
Collapse
Affiliation(s)
- Emily T. Chan
- Interdisciplinary Biophysics Graduate Program, The Ohio State University, Columbus, OH 43210, U.S.A
| | - Cömert Kural
- Interdisciplinary Biophysics Graduate Program, The Ohio State University, Columbus, OH 43210, U.S.A
- Department of Physics, The Ohio State University, Columbus, OH 43210, U.S.A
| |
Collapse
|
5
|
Wofford W, Kim J, Kim D, Janneh AH, Lee HG, Atilgan FC, Oleinik N, Kassir MF, Saatci O, Chakraborty P, Tokat UM, Gencer S, Howley B, Howe P, Mehrotra S, Sahin O, Ogretmen B. Alterations of ceramide synthesis induce PD-L1 internalization and signaling to regulate tumor metastasis and immunotherapy response. Cell Rep 2024; 43:114532. [PMID: 39046874 PMCID: PMC11404065 DOI: 10.1016/j.celrep.2024.114532] [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: 01/09/2024] [Revised: 05/17/2024] [Accepted: 07/09/2024] [Indexed: 07/27/2024] Open
Abstract
Programmed death ligand 1, PD-L1 (CD274), facilitates immune evasion and exerts pro-survival functions in cancer cells. Here, we report a mechanism whereby internalization of PD-L1 in response to alterations of bioactive lipid/ceramide metabolism by ceramide synthase 4 (CerS4) induces sonic hedgehog (Shh) and transforming growth factor β receptor signaling to enhance tumor metastasis in triple-negative breast cancers (TNBCs), exhibiting immunotherapy resistance. Mechanistically, data showed that internalized PD-L1 interacts with an RNA-binding protein, caprin-1, to stabilize Shh/TGFBR1/Wnt mRNAs to induce β-catenin signaling and TNBC growth/metastasis, consistent with increased infiltration of FoxP3+ regulatory T cells and resistance to immunotherapy. While mammary tumors developed in MMTV-PyMT/CerS4-/- were highly metastatic, targeting the Shh/PD-L1 axis using sonidegib and anti-PD-L1 antibody vastly decreased tumor growth and metastasis, consistent with the inhibition of PD-L1 internalization and Shh/Wnt signaling, restoring anti-tumor immune response. These data, validated in clinical samples and databases, provide a mechanism-based therapeutic strategy to improve immunotherapy responses in metastatic TNBCs.
Collapse
Affiliation(s)
- Wyatt Wofford
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, 86 Jonathan Lucas Street, Charleston, SC 29425, USA; Hollings Cancer Center, Medical University of South Carolina, 86 Jonathan Lucas Street, Charleston, SC 29425, USA
| | - Jisun Kim
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, 86 Jonathan Lucas Street, Charleston, SC 29425, USA; Hollings Cancer Center, Medical University of South Carolina, 86 Jonathan Lucas Street, Charleston, SC 29425, USA
| | - Dosung Kim
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, 86 Jonathan Lucas Street, Charleston, SC 29425, USA; Hollings Cancer Center, Medical University of South Carolina, 86 Jonathan Lucas Street, Charleston, SC 29425, USA
| | - Alhaji H Janneh
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, 86 Jonathan Lucas Street, Charleston, SC 29425, USA; Hollings Cancer Center, Medical University of South Carolina, 86 Jonathan Lucas Street, Charleston, SC 29425, USA
| | - Han Gyul Lee
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, 86 Jonathan Lucas Street, Charleston, SC 29425, USA; Hollings Cancer Center, Medical University of South Carolina, 86 Jonathan Lucas Street, Charleston, SC 29425, USA
| | - F Cansu Atilgan
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, 86 Jonathan Lucas Street, Charleston, SC 29425, USA; Hollings Cancer Center, Medical University of South Carolina, 86 Jonathan Lucas Street, Charleston, SC 29425, USA
| | - Natalia Oleinik
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, 86 Jonathan Lucas Street, Charleston, SC 29425, USA; Hollings Cancer Center, Medical University of South Carolina, 86 Jonathan Lucas Street, Charleston, SC 29425, USA
| | - Mohamed Faisal Kassir
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, 86 Jonathan Lucas Street, Charleston, SC 29425, USA; Hollings Cancer Center, Medical University of South Carolina, 86 Jonathan Lucas Street, Charleston, SC 29425, USA
| | - Ozge Saatci
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, 86 Jonathan Lucas Street, Charleston, SC 29425, USA; Hollings Cancer Center, Medical University of South Carolina, 86 Jonathan Lucas Street, Charleston, SC 29425, USA
| | - Paramita Chakraborty
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, 86 Jonathan Lucas Street, Charleston, SC 29425, USA; Hollings Cancer Center, Medical University of South Carolina, 86 Jonathan Lucas Street, Charleston, SC 29425, USA; Department of Surgery, Medical University of South Carolina, 86 Jonathan Lucas Street, Charleston, SC 29425, USA
| | - Unal Metin Tokat
- Department of Molecular Biology and Genetics, Bilkent University, Ankara, Turkey
| | - Salih Gencer
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, 86 Jonathan Lucas Street, Charleston, SC 29425, USA; Hollings Cancer Center, Medical University of South Carolina, 86 Jonathan Lucas Street, Charleston, SC 29425, USA; Istanbul Medipol University, Health Science and Technologies Research Institute (SABİTA), Cancer Research Center, Istanbul, Turkey
| | - Breege Howley
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, 86 Jonathan Lucas Street, Charleston, SC 29425, USA; Hollings Cancer Center, Medical University of South Carolina, 86 Jonathan Lucas Street, Charleston, SC 29425, USA
| | - Philip Howe
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, 86 Jonathan Lucas Street, Charleston, SC 29425, USA; Hollings Cancer Center, Medical University of South Carolina, 86 Jonathan Lucas Street, Charleston, SC 29425, USA
| | - Shikhar Mehrotra
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, 86 Jonathan Lucas Street, Charleston, SC 29425, USA; Hollings Cancer Center, Medical University of South Carolina, 86 Jonathan Lucas Street, Charleston, SC 29425, USA; Department of Surgery, Medical University of South Carolina, 86 Jonathan Lucas Street, Charleston, SC 29425, USA
| | - Ozgur Sahin
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, 86 Jonathan Lucas Street, Charleston, SC 29425, USA; Hollings Cancer Center, Medical University of South Carolina, 86 Jonathan Lucas Street, Charleston, SC 29425, USA
| | - Besim Ogretmen
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, 86 Jonathan Lucas Street, Charleston, SC 29425, USA; Hollings Cancer Center, Medical University of South Carolina, 86 Jonathan Lucas Street, Charleston, SC 29425, USA.
| |
Collapse
|
6
|
He YR, Xia H, Yun P, Xu Y, Ma WMJ, Xiao ZX, Zha GF. α-Lactalbumin mRNA-LNP Evokes an Anti-Tumor Effect Combined with Surgery in Triple-Negative Breast Cancer. Pharmaceutics 2024; 16:940. [PMID: 39065637 PMCID: PMC11279974 DOI: 10.3390/pharmaceutics16070940] [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: 06/04/2024] [Revised: 07/04/2024] [Accepted: 07/04/2024] [Indexed: 07/28/2024] Open
Abstract
Triple-negative breast cancer (TNBC) has been considered a huge clinical unmet need due to its aggressive progression and highly frequent metastasis. mRNA therapeutics supply a potential and versatile immunotherapy of oncology treatment. Here, we developed α-lactalbumin mRNA-lipid nanoparticles (α-LNP) as a potential therapeutical strategy for TNBC. The α-LNP induced the specific IgG antibodies and activated IFN γ-secreting-T cells in vivo. Additionally, the safety of α-LNP also had been demonstrated in vivo. When vaccinated prior to tumor implantation, α-LNP showed a preventive effect against 4T1 tumor growth and extended the survival of the tumor model by activating the memory immune responses. Furthermore, α-LNP administration in combination with surgical removal of neoplasm effectively inhibited the progression and metastasis in the TNBC model. Taken together, our results indicate that the α-LNP vaccine is a promising novel treatment for both therapeutics and prophylactics in TNBC.
Collapse
Affiliation(s)
- Yun-Ru He
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital, Sun Yat-sen University, No. 628, Zhenyuan Road, Guangming District, Shenzhen 518107, China; (Y.-R.H.); (H.X.); (Y.X.)
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, No. 628, Zhenyuan Road, Guangming District, Shenzhen 518107, China
| | - Heng Xia
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital, Sun Yat-sen University, No. 628, Zhenyuan Road, Guangming District, Shenzhen 518107, China; (Y.-R.H.); (H.X.); (Y.X.)
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, No. 628, Zhenyuan Road, Guangming District, Shenzhen 518107, China
| | - Peng Yun
- Department of Endocrinology, The Seventh Affiliated Hospital, Sun Yat-sen University, No. 628, Zhenyuan Road, Guangming District, Shenzhen 518107, China;
| | - Yuandong Xu
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital, Sun Yat-sen University, No. 628, Zhenyuan Road, Guangming District, Shenzhen 518107, China; (Y.-R.H.); (H.X.); (Y.X.)
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, No. 628, Zhenyuan Road, Guangming District, Shenzhen 518107, China
| | - Winson M. J. Ma
- Shenzhen Institute of Advanced Technology, 1068 Xueyuan Avenue, Shenzhen University Town, Shenzhen 518055, China;
| | - Ze-Xiu Xiao
- Drug Discovery Center, Shenzhen MagicRNA Biotech, Shenzhen 518107, China
| | - Gao-Feng Zha
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital, Sun Yat-sen University, No. 628, Zhenyuan Road, Guangming District, Shenzhen 518107, China; (Y.-R.H.); (H.X.); (Y.X.)
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, No. 628, Zhenyuan Road, Guangming District, Shenzhen 518107, China
| |
Collapse
|
7
|
Han ZG, He K, Zheng Y, Qian L. Visualizing the cellular internalization of therapeutic antibodies via pH-sensitive release of AIEgen. Org Biomol Chem 2024; 22:4950-4957. [PMID: 38817202 DOI: 10.1039/d4ob00512k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/01/2024]
Abstract
Among the fastest-growing bio-pharmaceuticals, therapeutic antibodies have achieved unprecedented success in treating various diseases. Though powerful, issues such as inefficacy or acquired resistance are waiting to be addressed to benefit more patients with improved therapeutic outcomes. In addition to in vivo distribution, the cellular spatiotemporal information including the antibody-antigen interaction and subsequent internalization is found to be important for the therapeutic effects. To better understand the cellular fate of therapeutic antibodies, especially the cellular internalization process, we employed a pH-sensitive linker to attach a red-emissive AIEgen onto the antibody. The resulting antibody conjugate will undergo AIEgen release to liberate brilliant fluorescence inside acidic endo/lysosomes, allowing wash-free visualization of the internalization process and facilitating the evaluation of antibody-drug efficacy.
Collapse
Affiliation(s)
- Zai-Gang Han
- Department of Pharmacy, Affiliated Hospital of Beihua University, Jilin 132011, China
| | - Kaifeng He
- Department of Pharmacy, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou 310052, China
- Institute of Drug Metabolism and Pharmaceutical Analysis, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China.
| | - Yi Zheng
- Institute of Drug Metabolism and Pharmaceutical Analysis, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China.
| | - Linghui Qian
- Institute of Drug Metabolism and Pharmaceutical Analysis, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
8
|
Baker AG, Hartono M, Ou H, Popov AB, Brown EL, Joseph J, Golinska M, González‐Gualda E, Macias D, Ge J, Denholm M, Morsli S, Sanghera C, Else TR, Greer HF, Vernet A, Bohndiek SE, Muñoz‐Espín D, Fruk L. An Indocyanine Green-Based Nanoprobe for In Vivo Detection of Cellular Senescence. Angew Chem Int Ed Engl 2024; 63:e202404885. [PMID: 38622059 PMCID: PMC11497227 DOI: 10.1002/anie.202404885] [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: 03/12/2024] [Revised: 04/12/2024] [Accepted: 04/15/2024] [Indexed: 04/17/2024]
Abstract
There is an urgent need to improve conventional cancer-treatments by preventing detrimental side effects, cancer recurrence and metastases. Recent studies have shown that presence of senescent cells in tissues treated with chemo- or radiotherapy can be used to predict the effectiveness of cancer treatment. However, although the accumulation of senescent cells is one of the hallmarks of cancer, surprisingly little progress has been made in development of strategies for their detection in vivo. To address a lack of detection tools, we developed a biocompatible, injectable organic nanoprobe (NanoJagg), which is selectively taken up by senescent cells and accumulates in the lysosomes. The NanoJagg probe is obtained by self-assembly of indocyanine green (ICG) dimers using a scalable manufacturing process and characterized by a unique spectral signature suitable for both photoacoustic tomography (PAT) and fluorescence imaging. In vitro, ex vivo and in vivo studies all indicate that NanoJaggs are a clinically translatable probe for detection of senescence and their PAT signal makes them suitable for longitudinal monitoring of the senescence burden in solid tumors after chemotherapy or radiotherapy.
Collapse
Affiliation(s)
- Andrew G. Baker
- Early Cancer instituteDepartment of OncologyUniversity of CambridgeHills RoadCambridgeCB2 0XZUK
- Department of Chemical Engineering and BiotechnologyUniversity of CambridgePhilippa Fawcett DriveCambridgeCB3 0ASUK
| | - Muhamad Hartono
- Early Cancer instituteDepartment of OncologyUniversity of CambridgeHills RoadCambridgeCB2 0XZUK
- Department of Chemical Engineering and BiotechnologyUniversity of CambridgePhilippa Fawcett DriveCambridgeCB3 0ASUK
| | - Hui‐Ling Ou
- Early Cancer instituteDepartment of OncologyUniversity of CambridgeHills RoadCambridgeCB2 0XZUK
| | - Andrea Bistrović Popov
- Department of Chemical Engineering and BiotechnologyUniversity of CambridgePhilippa Fawcett DriveCambridgeCB3 0ASUK
| | - Emma L. Brown
- Department of PhysicsUniversity ofCambridgeJJ Thomson AvenueCB3 0HEUnited Kingdom
- Cancer Research UK Cambridge InstituteRobinson WayCambridgeCB2 0REUK
| | - James Joseph
- Department of PhysicsUniversity ofCambridgeJJ Thomson AvenueCB3 0HEUnited Kingdom
- Cancer Research UK Cambridge InstituteRobinson WayCambridgeCB2 0REUK
- School of Science and EngineeringUniversity of DundeeDundeeDD1 4HNScotlandUK
| | - Monika Golinska
- Department of PhysicsUniversity ofCambridgeJJ Thomson AvenueCB3 0HEUnited Kingdom
- Cancer Research UK Cambridge InstituteRobinson WayCambridgeCB2 0REUK
| | - Estela González‐Gualda
- Early Cancer instituteDepartment of OncologyUniversity of CambridgeHills RoadCambridgeCB2 0XZUK
| | - David Macias
- Instituto de Biomedicina de Sevilla, IBIS/Hospital Universitario Virgen del RocioUniversidad de SevillaAvda.Dr. Fedriani/>Sevilla41009Spain
| | - Jianfeng Ge
- Early Cancer instituteDepartment of OncologyUniversity of CambridgeHills RoadCambridgeCB2 0XZUK
| | - Mary Denholm
- Early Cancer instituteDepartment of OncologyUniversity of CambridgeHills RoadCambridgeCB2 0XZUK
| | - Samir Morsli
- Early Cancer instituteDepartment of OncologyUniversity of CambridgeHills RoadCambridgeCB2 0XZUK
| | - Chandan Sanghera
- Department of Chemical Engineering and BiotechnologyUniversity of CambridgePhilippa Fawcett DriveCambridgeCB3 0ASUK
| | - Thomas R. Else
- Department of PhysicsUniversity ofCambridgeJJ Thomson AvenueCB3 0HEUnited Kingdom
- Cancer Research UK Cambridge InstituteRobinson WayCambridgeCB2 0REUK
| | - Heather F. Greer
- Yusuf Hamied Department of ChemistryUniversity of CambridgeLensfield RoadCambridgeCB2 1EWUK
| | - Aude Vernet
- Cancer Research UK Cambridge InstituteRobinson WayCambridgeCB2 0REUK
| | - Sarah E. Bohndiek
- Department of PhysicsUniversity ofCambridgeJJ Thomson AvenueCB3 0HEUnited Kingdom
- Cancer Research UK Cambridge InstituteRobinson WayCambridgeCB2 0REUK
| | - Daniel Muñoz‐Espín
- Early Cancer instituteDepartment of OncologyUniversity of CambridgeHills RoadCambridgeCB2 0XZUK
- Yusuf Hamied Department of ChemistryUniversity of CambridgeLensfield RoadCambridgeCB2 1EWUK
| | - Ljiljana Fruk
- Department of Chemical Engineering and BiotechnologyUniversity of CambridgePhilippa Fawcett DriveCambridgeCB3 0ASUK
- Yusuf Hamied Department of ChemistryUniversity of CambridgeLensfield RoadCambridgeCB2 1EWUK
| |
Collapse
|
9
|
Chen L, Patil S, Barbon J, Waire J, Laroux S, McCarthy D, Pratibha M, Zhong S, Dong F, Orsi K, Nguyen G, Yang Y, Crosbie N, Dominguez E, Deora A, Veldman G, Westmoreland S, Jin L, Radstake T, White K, Wei HJ. Agonistic anti-DCIR antibody inhibits ITAM-mediated inflammatory signaling and promotes immune resolution. JCI Insight 2024; 9:e176064. [PMID: 38781017 PMCID: PMC11383175 DOI: 10.1172/jci.insight.176064] [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: 10/05/2023] [Accepted: 05/15/2024] [Indexed: 05/25/2024] Open
Abstract
DC inhibitory receptor (DCIR) is a C-type lectin receptor selectively expressed on myeloid cells, including monocytes, macrophages, DCs, and neutrophils. Its role in immune regulation has been implicated in murine models and human genome-wide association studies, suggesting defective DCIR function associates with increased susceptibility to autoimmune diseases such as rheumatoid arthritis, lupus, and Sjögren's syndrome. However, little is known about the mechanisms underlying DCIR activation to dampen inflammation. Here, we developed anti-DCIR agonistic antibodies that promote phosphorylation on DCIR's immunoreceptor tyrosine-based inhibitory motifs and recruitment of SH2 containing protein tyrosine phosphatase-2 for reducing inflammation. We also explored the inflammation resolution by depleting DCIR+ cells with antibodies. Utilizing a human DCIR-knock-in mouse model, we validated the antiinflammatory properties of the agonistic anti-DCIR antibody in experimental peritonitis and colitis. These findings provide critical evidence for targeting DCIR to develop transformative therapies for inflammatory diseases.
Collapse
Affiliation(s)
- Liang Chen
- AbbVie, Cambridge Research Center, Cambridge, Massachusetts, USA
| | - Suresh Patil
- AbbVie, Cambridge Research Center, Cambridge, Massachusetts, USA
| | - Jeffrey Barbon
- AbbVie Bioresearch Center, Worcester, Massachusetts, USA
| | - James Waire
- AbbVie Bioresearch Center, Worcester, Massachusetts, USA
| | - Stephen Laroux
- AbbVie Bioresearch Center, Worcester, Massachusetts, USA
| | - Donna McCarthy
- AbbVie Bioresearch Center, Worcester, Massachusetts, USA
| | - Mishra Pratibha
- AbbVie, Cambridge Research Center, Cambridge, Massachusetts, USA
| | - Suju Zhong
- AbbVie, Cambridge Research Center, Cambridge, Massachusetts, USA
| | - Feng Dong
- AbbVie, Cambridge Research Center, Cambridge, Massachusetts, USA
| | - Karin Orsi
- AbbVie Bioresearch Center, Worcester, Massachusetts, USA
| | - Gunarso Nguyen
- AbbVie Bioresearch Center, Worcester, Massachusetts, USA
| | - Yingli Yang
- AbbVie Bioresearch Center, Worcester, Massachusetts, USA
| | - Nancy Crosbie
- AbbVie Bioresearch Center, Worcester, Massachusetts, USA
| | - Eric Dominguez
- AbbVie Bioresearch Center, Worcester, Massachusetts, USA
| | - Arun Deora
- AbbVie, Cambridge Research Center, Cambridge, Massachusetts, USA
| | | | | | - Liang Jin
- AbbVie Bioresearch Center, Worcester, Massachusetts, USA
| | - Timothy Radstake
- AbbVie, Cambridge Research Center, Cambridge, Massachusetts, USA
| | - Kevin White
- AbbVie, Cambridge Research Center, Cambridge, Massachusetts, USA
| | - Hsi-Ju Wei
- AbbVie Bay Area, South San Francisco, California, USA
| |
Collapse
|
10
|
Huang Y, Zhao Y, Huang Y, Yang Y, Zhang Y, Hong S, Zhao H, Zhao S, Zhou T, Chen G, Zhou H, Ma Y, Zhou N, Zhang L, Fang W. Phase 1b trial of anti-HER2 antibody inetetamab and pan-HER inhibitor pyrotinib in HER2-positive advanced lung cancer. MedComm (Beijing) 2024; 5:e536. [PMID: 38685972 PMCID: PMC11057420 DOI: 10.1002/mco2.536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 03/16/2024] [Accepted: 03/18/2024] [Indexed: 05/02/2024] Open
Abstract
There remains an unmet need for targeted therapies against advanced non-small-cell lung cancer (NSCLC) with HER2 mutations. To improve the antitumor activity of single anti-HER2 agent, this prospective, single-arm clinical trial (NCT05016544) examined the safety profile and efficacy of anti-HER2 antibody inetetamab and pan-HER TKI pyrotinib in HER2-posivite advanced NSCLC patients. Enrolled patients received inetetamab every 3 weeks and pyrotinib once per day (pyrotinib, dose-escalation part, 240 mg, 320 mg; dose-expansion part, 320 mg). Primary endpoints were dose-limiting toxicity (DLT) dosage and safety. Secondary endpoints included progression-free survival (PFS), objective response rate (ORR), and disease control rate (DCR). A total of 48 patients were enrolled. During the dose-escalation period, no DLT occurred. Diarrhea was the most commonly reported treatment-related adverse event (TRAE). Grade 3 TRAEs occurred in seven patients. The median PFS (mPFS) was 5.5 months [95% confidence interval (CI): 4.4-8.6 months]. The confirmed ORR and DCR reached 25% (11/44) and 84.1% (37/44), respectively. Responses were shown in patients with distinct HER2 aberrations. In summary, inetetamab in combination with pyrotinib demonstrated acceptable safety and antitumor activity among patients with advanced HER2-mutant NSCLC.
Collapse
Affiliation(s)
- Yihua Huang
- Department of Medical OncologyState Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerCollaborative Innovation Center for Cancer MedicineSun Yat‐Sen University Cancer CenterGuangzhouPR China
| | - Yuanyuan Zhao
- Department of Medical OncologyState Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerCollaborative Innovation Center for Cancer MedicineSun Yat‐Sen University Cancer CenterGuangzhouPR China
| | - Yan Huang
- Department of Medical OncologyState Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerCollaborative Innovation Center for Cancer MedicineSun Yat‐Sen University Cancer CenterGuangzhouPR China
| | - Yunpeng Yang
- Department of Medical OncologyState Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerCollaborative Innovation Center for Cancer MedicineSun Yat‐Sen University Cancer CenterGuangzhouPR China
| | - Yaxiong Zhang
- Department of Medical OncologyState Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerCollaborative Innovation Center for Cancer MedicineSun Yat‐Sen University Cancer CenterGuangzhouPR China
| | - Shaodong Hong
- Department of Medical OncologyState Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerCollaborative Innovation Center for Cancer MedicineSun Yat‐Sen University Cancer CenterGuangzhouPR China
| | - Hongyun Zhao
- Department of Clinical ResearchState Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerCollaborative Innovation Center for Cancer MedicineSun Yat‐sen University Cancer CenterGuangzhouPR China
| | - Shen Zhao
- Department of Medical OncologyState Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerCollaborative Innovation Center for Cancer MedicineSun Yat‐Sen University Cancer CenterGuangzhouPR China
| | - Ting Zhou
- Department of Medical OncologyState Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerCollaborative Innovation Center for Cancer MedicineSun Yat‐Sen University Cancer CenterGuangzhouPR China
| | - Gang Chen
- Department of Medical OncologyState Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerCollaborative Innovation Center for Cancer MedicineSun Yat‐Sen University Cancer CenterGuangzhouPR China
| | - Huaqiang Zhou
- Department of Medical OncologyState Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerCollaborative Innovation Center for Cancer MedicineSun Yat‐Sen University Cancer CenterGuangzhouPR China
| | - Yuxiang Ma
- Department of Clinical ResearchState Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerCollaborative Innovation Center for Cancer MedicineSun Yat‐sen University Cancer CenterGuangzhouPR China
| | - Ningning Zhou
- Department of Medical OncologyState Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerCollaborative Innovation Center for Cancer MedicineSun Yat‐Sen University Cancer CenterGuangzhouPR China
| | - Li Zhang
- Department of Medical OncologyState Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerCollaborative Innovation Center for Cancer MedicineSun Yat‐Sen University Cancer CenterGuangzhouPR China
| | - Wenfeng Fang
- Department of Medical OncologyState Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerCollaborative Innovation Center for Cancer MedicineSun Yat‐Sen University Cancer CenterGuangzhouPR China
| |
Collapse
|
11
|
Fernandez-Rojo MA, Pearen MA, Burgess AG, Ikonomopoulou MP, Hoang-Le D, Genz B, Saggiomo SL, Nawaratna SSK, Poli M, Reissmann R, Gobert GN, Deutsch U, Engelhardt B, Brooks AJ, Jones A, Arosio P, Ramm GA. The heavy subunit of ferritin stimulates NLRP3 inflammasomes in hepatic stellate cells through ICAM-1 to drive hepatic inflammation. Sci Signal 2024; 17:eade4335. [PMID: 38564492 DOI: 10.1126/scisignal.ade4335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 03/11/2024] [Indexed: 04/04/2024]
Abstract
Serum ferritin concentrations increase during hepatic inflammation and correlate with the severity of chronic liver disease. Here, we report a molecular mechanism whereby the heavy subunit of ferritin (FTH) contributes to hepatic inflammation. We found that FTH induced activation of the NLRP3 inflammasome and secretion of the proinflammatory cytokine interleukin-1β (IL-1β) in primary rat hepatic stellate cells (HSCs) through intercellular adhesion molecule-1 (ICAM-1). FTH-ICAM-1 stimulated the expression of Il1b, NLRP3 inflammasome activation, and the processing and secretion of IL-1β in a manner that depended on plasma membrane remodeling, clathrin-mediated endocytosis, and lysosomal destabilization. FTH-ICAM-1 signaling at early endosomes stimulated Il1b expression, implying that this endosomal signaling primed inflammasome activation in HSCs. In contrast, lysosomal destabilization was required for FTH-induced IL-1β secretion, suggesting that lysosomal damage activated inflammasomes. FTH induced IL-1β production in liver slices from wild-type mice but not in those from Icam1-/- or Nlrp3-/- mice. Thus, FTH signals through its receptor ICAM-1 on HSCs to activate the NLRP3 inflammasome. We speculate that this pathway contributes to hepatic inflammation, a key process that stimulates hepatic fibrogenesis associated with chronic liver disease.
Collapse
Affiliation(s)
- Manuel A Fernandez-Rojo
- QIMR Berghofer Medical Research Institute, Brisbane, Herston, QLD 4006, Australia
- School of Medicine, University of Queensland, Brisbane, Herston, QLD 4006, Australia
- Hepatic Regenerative Medicine Laboratory, Madrid Institute for Advanced Studies in Food, Madrid 28049, Spain
- University of Queensland Diamantina Institute, University of Queensland, Translational Research Institute, Brisbane, QLD 4102, Australia
| | - Michael A Pearen
- QIMR Berghofer Medical Research Institute, Brisbane, Herston, QLD 4006, Australia
| | - Anita G Burgess
- QIMR Berghofer Medical Research Institute, Brisbane, Herston, QLD 4006, Australia
| | - Maria P Ikonomopoulou
- QIMR Berghofer Medical Research Institute, Brisbane, Herston, QLD 4006, Australia
- School of Medicine, University of Queensland, Brisbane, Herston, QLD 4006, Australia
- Translational Venomics Laboratory, Madrid Institute for Advanced Studies in Food, Madrid 28049, Spain
- Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD 4072, Australia
| | - Diem Hoang-Le
- QIMR Berghofer Medical Research Institute, Brisbane, Herston, QLD 4006, Australia
| | - Berit Genz
- QIMR Berghofer Medical Research Institute, Brisbane, Herston, QLD 4006, Australia
| | - Silvia L Saggiomo
- QIMR Berghofer Medical Research Institute, Brisbane, Herston, QLD 4006, Australia
| | | | - Maura Poli
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Regina Reissmann
- Department for BioMedical Research (DBMR), University of Bern, Freiestrasse 1, CH-3012 Bern, Switzerland
| | - Geoffrey N Gobert
- QIMR Berghofer Medical Research Institute, Brisbane, Herston, QLD 4006, Australia
- School of Biological Sciences, Queen's University Belfast, Belfast, UK
| | - Urban Deutsch
- Theodor Kocher Institute, University of Bern, Freiestrasse 1, CH-3012 Bern, Switzerland
| | - Britta Engelhardt
- Theodor Kocher Institute, University of Bern, Freiestrasse 1, CH-3012 Bern, Switzerland
| | - Andrew J Brooks
- University of Queensland Diamantina Institute, University of Queensland, Translational Research Institute, Brisbane, QLD 4102, Australia
| | - Alun Jones
- Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD 4072, Australia
| | - Paolo Arosio
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Grant A Ramm
- QIMR Berghofer Medical Research Institute, Brisbane, Herston, QLD 4006, Australia
- School of Medicine, University of Queensland, Brisbane, Herston, QLD 4006, Australia
| |
Collapse
|
12
|
Lopes RM, Souza ACS, Otręba M, Rzepecka-Stojko A, Tersariol ILS, Rodrigues T. Targeting autophagy by antipsychotic phenothiazines: potential drug repurposing for cancer therapy. Biochem Pharmacol 2024; 222:116075. [PMID: 38395266 DOI: 10.1016/j.bcp.2024.116075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Revised: 01/14/2024] [Accepted: 02/20/2024] [Indexed: 02/25/2024]
Abstract
Cancer is recognized as the major cause of death worldwide and the most challenging public health issues. Tumor cells exhibit molecular adaptations and metabolic reprograming to sustain their high proliferative rate and autophagy plays a pivotal role to supply the high demand for metabolic substrates and for recycling cellular components, which has attracted the attention of the researchers. The modulation of the autophagic process sensitizes tumor cells to chemotherapy-induced cell death and reverts drug resistance. In this regard, many in vitro and in vivo studies having shown the anticancer activity of phenothiazine (PTZ) derivatives due to their potent cytotoxicity in tumor cells. Interestingly, PTZ have been used as antiemetics in antitumor chemotherapy-induced vomiting, maybe exerting a combined antitumor effect. Among the mechanisms of cytotoxicity, the modulation of autophagy by these drugs has been highlighted. Therefore, the use of PTZ derivatives can be considered as a repurposing strategy in antitumor chemotherapy. Here, we provided an overview of the effects of antipsychotic PTZ on autophagy in tumor cells, evidencing the molecular targets and discussing the underlying mechanisms. The modulation of autophagy by PTZ in tumor cells have been consistently related to their cytotoxic action. These effects depend on the derivative, their concentration, and also the type of cancer. Most data have shown the impairment of autophagic flux by PTZ, probably due to the blockade of lysosome-autophagosome fusion, but some studies have also suggested the induction of autophagy. These data highlight the therapeutic potential of targeting autophagy by PTZ in cancer chemotherapy.
Collapse
Affiliation(s)
- Rayssa M Lopes
- Center for Natural and Human Sciences (CCNH), Federal University of ABC (UFABC), Santo Andre, SP, Brazil.
| | - Ana Carolina S Souza
- Center for Natural and Human Sciences (CCNH), Federal University of ABC (UFABC), Santo Andre, SP, Brazil.
| | - Michał Otręba
- Department of Drug and Cosmetics Technology, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia in Katowice, Poland.
| | - Anna Rzepecka-Stojko
- Department of Drug and Cosmetics Technology, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia in Katowice, Poland.
| | - Ivarne L S Tersariol
- Departament of Molecular Biology, Federal University of São Paulo (UNIFESP), Sao Paulo, SP, Brazil
| | - Tiago Rodrigues
- Center for Natural and Human Sciences (CCNH), Federal University of ABC (UFABC), Santo Andre, SP, Brazil.
| |
Collapse
|
13
|
Martin JH, Galettis P, Flynn A, Schneider J. Phenotype versus genotype to optimize cancer dosing in the clinical setting-focus on 5-fluorouracil and tyrosine kinase inhibitors. Pharmacol Res Perspect 2024; 12:e1182. [PMID: 38429945 PMCID: PMC10907881 DOI: 10.1002/prp2.1182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 12/19/2023] [Accepted: 12/19/2023] [Indexed: 03/03/2024] Open
Abstract
Cancer medicines often have narrow therapeutic windows; toxicity can be severe and sometimes fatal, but inadequate dose intensity reduces efficacy and survival. Determining the optimal dose for each patient is difficult, with body-surface area used most commonly for chemotherapy and flat dosing for tyrosine kinase inhibitors, despite accumulating evidence of a wide range of exposures in individual patients with many receiving a suboptimal dose with these strategies. Therapeutic drug monitoring (measuring the drug concentration in a biological fluid, usually plasma) (TDM) is an accepted and well validated method to guide dose adjustments for individual patients to improve this. However, implementing TDM in routine care has been difficult outside a research context. The development of genotyping of various proteins involved in drug elimination and activity has gained prominence, with several but not all Guideline groups recommending dose reductions for particular variant genotypes. However, there is increasing concern that dosing recommendations are based on limited data sets and may lead to unnecessary underdosing and increased cancer mortality. This Review discusses the evidence surrounding genotyping and TDM to guide decisions around best practice.
Collapse
Affiliation(s)
- Jennifer H. Martin
- Drug Repurposing and Medicines Research ProgramHunter Medical Research InstituteNew Lambton HeightsNew South WalesAustralia
| | - Peter Galettis
- Drug Repurposing and Medicines Research ProgramHunter Medical Research InstituteNew Lambton HeightsNew South WalesAustralia
| | - Alex Flynn
- Drug Repurposing and Medicines Research ProgramHunter Medical Research InstituteNew Lambton HeightsNew South WalesAustralia
| | - Jennifer Schneider
- Drug Repurposing and Medicines Research ProgramHunter Medical Research InstituteNew Lambton HeightsNew South WalesAustralia
| |
Collapse
|
14
|
Muraro E, Montico B, Lum B, Colizzi F, Giurato G, Salvati A, Guerrieri R, Rizzo A, Comaro E, Canzonieri V, Anichini A, Del Vecchio M, Mortarini R, Milione M, Weisz A, Pizzichetta MA, Simpson F, Dolcetti R, Fratta E, Sigalotti L. Antibody dependent cellular cytotoxicity-inducing anti-EGFR antibodies as effective therapeutic option for cutaneous melanoma resistant to BRAF inhibitors. Front Immunol 2024; 15:1336566. [PMID: 38510242 PMCID: PMC10950948 DOI: 10.3389/fimmu.2024.1336566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 02/13/2024] [Indexed: 03/22/2024] Open
Abstract
Introduction About 50% of cutaneous melanoma (CM) patients present activating BRAF mutations that can be effectively targeted by BRAF inhibitors (BRAFi). However, 20% of CM patients exhibit intrinsic drug resistance to BRAFi, while most of the others develop adaptive resistance over time. The mechanisms involved in BRAFi resistance are disparate and globally seem to rewire the cellular signaling profile by up-regulating different receptor tyrosine kinases (RTKs), such as the epidermal growth factor receptor (EGFR). RTKs inhibitors have not clearly demonstrated anti-tumor activity in BRAFi resistant models. To overcome this issue, we wondered whether the shared up-regulated RTK phenotype associated with BRAFi resistance could be exploited by using immune weapons as the antibody-dependent cell cytotoxicity (ADCC)-mediated effect of anti-RTKs antibodies, and kill tumor cells independently from the mechanistic roots. Methods and results By using an in vitro model of BRAFi resistance, we detected increased membrane expression of EGFR, both at mRNA and protein level in 4 out of 9 BRAFi-resistant (VR) CM cultures as compared to their parental sensitive cells. Increased EGFR phosphorylation and AKT activation were observed in the VR CM cultures. EGFR signaling appeared dispensable for maintaining resistance, since small molecule-, antibody- and CRISPR-targeting of EGFR did not restore sensitivity of VR cells to BRAFi. Importantly, immune-targeting of EGFR by the anti-EGFR antibody cetuximab efficiently and specifically killed EGFR-expressing VR CM cells, both in vitro and in humanized mouse models in vivo, triggering ADCC by healthy donors' and patients' peripheral blood cells. Conclusion Our data demonstrate the efficacy of immune targeting of RTKs expressed by CM relapsing on BRAFi, providing the proof-of-concept supporting the assessment of anti-RTK antibodies in combination therapies in this setting. This strategy might be expected to concomitantly trigger the crosstalk of adaptive immune response leading to a complementing T cell immune rejection of tumors.
Collapse
Affiliation(s)
- Elena Muraro
- Immunopathology and Cancer Biomarkers, Department of Translational Research, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy
| | - Barbara Montico
- Immunopathology and Cancer Biomarkers, Department of Translational Research, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy
| | - Benedict Lum
- Frazer Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Francesca Colizzi
- Immunopathology and Cancer Biomarkers, Department of Translational Research, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy
| | - Giorgio Giurato
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Baronissi, Italy
- Genome Research Center for Health - CRGS, Baronissi, Italy
| | - Annamaria Salvati
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Baronissi, Italy
- Genome Research Center for Health - CRGS, Baronissi, Italy
- Molecular Pathology and Medical Genomics Program, AOU 'S. Giovanni di Dio e Ruggi d'Aragona' University of Salerno and Rete Oncologica Campana, Salerno, Italy
| | - Roberto Guerrieri
- Immunopathology and Cancer Biomarkers, Department of Translational Research, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy
| | - Aurora Rizzo
- Immunopathology and Cancer Biomarkers, Department of Translational Research, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy
| | - Elisa Comaro
- Immunopathology and Cancer Biomarkers, Department of Translational Research, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy
| | - Vincenzo Canzonieri
- Division of Pathology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy
- Department of Medical, Surgical and Health Sciences, University of Trieste, Trieste, Italy
| | - Andrea Anichini
- Human Tumors Immunobiology Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Michele Del Vecchio
- Melanoma Unit, Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Roberta Mortarini
- Human Tumors Immunobiology Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Massimo Milione
- Pathology Unit 1, Department of Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Alessandro Weisz
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Baronissi, Italy
- Genome Research Center for Health - CRGS, Baronissi, Italy
- Molecular Pathology and Medical Genomics Program, AOU 'S. Giovanni di Dio e Ruggi d'Aragona' University of Salerno and Rete Oncologica Campana, Salerno, Italy
| | - Maria Antonietta Pizzichetta
- Division of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy
- Department of Dermatology, University of Trieste, Trieste, Italy
| | - Fiona Simpson
- Frazer Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Riccardo Dolcetti
- Translational and Clinical Immunotherapy, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC, Australia
- Department of Microbiology and Immunology, The University of Melbourne, Melbourne, VIC, Australia
- Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| | - Elisabetta Fratta
- Immunopathology and Cancer Biomarkers, Department of Translational Research, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy
| | - Luca Sigalotti
- Oncogenetics and Functional Oncogenomics Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy
| |
Collapse
|
15
|
Stefanello ST, Mizdal CR, Shahin V. Pitstop-2 Upsets The Integrity of Nuclear Pore Complexes (NPCs) by Interaction with β-Propeller Folds of Npc Scaffold Proteins. Adv Biol (Weinh) 2024; 8:e2300360. [PMID: 38129324 DOI: 10.1002/adbi.202300360] [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: 07/21/2023] [Revised: 11/22/2023] [Indexed: 12/23/2023]
Abstract
The small compound Pitstop-2 is a recent potent inhibitor of clathrin-mediated endocytosis (CME), widely used in biomedical research areas. In recent years, however, it is observed that it exhibits CME-independent inhibitory effects on nuclear pore complexes (NPCs), the nucleocytoplasmic gatekeepers. NPCs are elaborate proteinaceous transport nano-machineries of crucial physiological importance rendering them novel targets for various medical applications. They mediate all nucleocytoplasmic transport forming a physiologically essential selective nucleocytoplasmic barrier. The direct Pitstop-2 disruptive effects on NPCs manifested themselves at both the structural and functional integrity levels. Moreover, they are massive, acute, and detectable at concentrations equal to CME-inhibitory concentrations. Pitstop-2 inhibits CME by binding to the terminal β-propeller domain of the heavy chain of clathrin. Several NPC scaffold proteins, critical for the structural and functional integrity of the NPC, possess β-propeller folds. Herein, utilizing computational docking analysis, it is demonstrated that Pitstop-2 exhibits particularly high binding affinities to β-propeller folds of NPC scaffold proteins, similar to its binding affinity to the terminal β-propeller domain of clathrin. The authors, therefore, conclude that Pitstop-2 is a potent disruptor of NPCs, an activity which, separately or in synergy with CME inhibition, may be exploited for a myriad of pharmacological applications.
Collapse
Affiliation(s)
- Sílvio Terra Stefanello
- Institute of Physiology II, University of Münster, Robert-Koch-Str. 27b, 48149, Münster, Germany
| | - Caren Rigon Mizdal
- Institute of Physiology II, University of Münster, Robert-Koch-Str. 27b, 48149, Münster, Germany
| | - Victor Shahin
- Institute of Physiology II, University of Münster, Robert-Koch-Str. 27b, 48149, Münster, Germany
| |
Collapse
|
16
|
Guo H, Zhou C, Zheng M, Zhang J, Wu H, He Q, Ding L, Yang B. Insights into the role of derailed endocytic trafficking pathway in cancer: From the perspective of cancer hallmarks. Pharmacol Res 2024; 201:107084. [PMID: 38295915 DOI: 10.1016/j.phrs.2024.107084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 01/12/2024] [Accepted: 01/25/2024] [Indexed: 02/06/2024]
Abstract
The endocytic trafficking pathway is a highly organized cellular program responsible for the regulation of membrane components and uptake of extracellular substances. Molecules internalized into the cell through endocytosis will be sorted for degradation or recycled back to membrane, which is determined by a series of sorting events. Many receptors, enzymes, and transporters on the membrane are strictly regulated by endocytic trafficking process, and thus the endocytic pathway has a profound effect on cellular homeostasis. However, the endocytic trafficking process is typically dysregulated in cancers, which leads to the aberrant retention of receptor tyrosine kinases and immunosuppressive molecules on cell membrane, the loss of adhesion protein, as well as excessive uptake of nutrients. Therefore, hijacking endocytic trafficking pathway is an important approach for tumor cells to obtain advantages of proliferation and invasion, and to evade immune attack. Here, we summarize how dysregulated endocytic trafficking process triggers tumorigenesis and progression from the perspective of several typical cancer hallmarks. The impact of endocytic trafficking pathway to cancer therapy efficacy is also discussed.
Collapse
Affiliation(s)
- Hongjie Guo
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Chen Zhou
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Mingming Zheng
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jie Zhang
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Honghai Wu
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Qiaojun He
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; The Innovation Institute for Artificial Intelligence in Medicine, Zhejiang University, Hangzhou 310018, China; Cancer Center of Zhejiang University, Hangzhou 310058, China
| | - Ling Ding
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; Nanhu Brain-computer Interface Institute, Hangzhou 311100, China.
| | - Bo Yang
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; School of Medicine, Hangzhou City University, Hangzhou 310015, China; The Innovation Institute for Artificial Intelligence in Medicine, Zhejiang University, Hangzhou 310018, China.
| |
Collapse
|
17
|
Karlsen EA, Walpole E, Simpson F. Steroid Premedication and Monoclonal Antibody Therapy: Should We Reconsider? Curr Treat Options Oncol 2024; 25:275-283. [PMID: 38270799 PMCID: PMC10894762 DOI: 10.1007/s11864-023-01170-4] [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] [Accepted: 11/30/2023] [Indexed: 01/26/2024]
Abstract
OPINION STATEMENT Monoclonal antibody (mAb) therapy is now considered a main component of cancer therapy in Australia. Although traditionally thought of as pure signalling inhibitors, a large proponent of these medications function through antibody-dependent cell-mediated cytotoxicity (ADCC). Currently, most protocols and institutional guidelines for ADCC-mediated mAbs promote the use of corticosteroids as premedication: this is implemented to reduce infusion-related reactions (IRRs) and antiemesis prophylaxis and combat concurrently administered chemotherapy-related syndromes. Concerningly, the inhibitory effects of ADCC by corticosteroids are well documented; henceforth, it is possible the current standard of care is misaligned to the literature surrounding ADCC. Subsequently, clinicians' decisions to act in contrast to this literature may be reducing the efficacy of mAbs. The literature suggests that the redundant use of corticosteroids should be cautioned against when used in conjunction with ADCC-mediated mAbs-this is due to the consequent reduction in anti-tumour activity. Owing to the fact IRRs typically occur upon initial infusion, the authors advocate for individual clinicians and institutional protocols to considering augmenting their practice to corticosteroid premedication at the first dose only, unless clinically indicated. Additionally, product information (PI) and consumer medicine information (CMI) documents distributed by Australian and international regulatory agencies should consider disclosing the risk of concurrent steroids with these medications. Moreover, the authors suggest considering alternative medications for the management of side effects.
Collapse
Affiliation(s)
- Emma-Anne Karlsen
- Frazer Institute, The University of Queensland, Brisbane, Australia.
- Department of General Surgery, Mater Hospital Brisbane, Brisbane, Australia.
- School of Medicine, The University of Queensland, Brisbane, Australia.
- Simpson Laboratory - Frazer Institute, The University of Queensland, 37 Kent Street, Woolloongabba, QLD, 4102, Australia.
| | - Euan Walpole
- School of Medicine, The University of Queensland, Brisbane, Australia
- Division of Cancer Services, Princess Alexandra Hospital, Brisbane, Australia
| | - Fiona Simpson
- Frazer Institute, The University of Queensland, Brisbane, Australia
- Simpson Laboratory - Frazer Institute, The University of Queensland, 37 Kent Street, Woolloongabba, QLD, 4102, Australia
| |
Collapse
|
18
|
Subasic CN, Simpson F, Minchin RF, Kaminskas LM. A PEGylated liposomal formulation of prochlorperazine that limits brain exposure but retains dynamin II activity: A potential adjuvant therapy for cancer patients receiving chemotherapeutic mAbs. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2024; 56:102733. [PMID: 38199450 DOI: 10.1016/j.nano.2024.102733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 12/21/2023] [Accepted: 12/29/2023] [Indexed: 01/12/2024]
Abstract
Anti-cancer monoclonal antibodies often fail to provide therapeutic benefit in receptor-positive patients due to rapid endocytosis of antibody-bound cell surface receptors. High dose co-administration of prochlorperazine (PCZ) inhibits endocytosis and sensitises tumours to mAbs by inhibiting dynamin II but can also introduce neurological side effects. We examined the potential to use PEGylated liposomal formulations of PCZ (LPCZ) to retain the anti-cancer effects of PCZ, but limit brain uptake. Uncharged liposomes showed complete drug encapsulation and pH-dependent drug release, but cationic liposomes showed limited drug encapsulation and lacked pH-dependent drug release. Uncharged LPCZ showed comparable inhibition of EGFR internalisation to free PCZ in KJD cells. After IV administration to rats, LPCZ reduced the plasma clearance and brain uptake of PCZ compared to IV PCZ. The results suggest that LPCZ may offer some benefit over PCZ as an adjunct therapy in cancer patients receiving mAb treatment.
Collapse
Affiliation(s)
- Christopher N Subasic
- School of Biomedical Sciences, University of Queensland, St Lucia, QLD 4072, Australia
| | - Fiona Simpson
- Frazer Institute, University of Queensland, St Lucia, QLD 4072, Australia
| | - Rodney F Minchin
- School of Biomedical Sciences, University of Queensland, St Lucia, QLD 4072, Australia
| | - Lisa M Kaminskas
- School of Biomedical Sciences, University of Queensland, St Lucia, QLD 4072, Australia.
| |
Collapse
|
19
|
Wang P, Zhou R, Zhou R, Feng S, Zhao L, Li W, Lin J, Rajapakse A, Lee CH, Furnari FB, Burgess AW, Gunter JH, Liu G, Ostrikov KK, Richard DJ, Simpson F, Dai X, Thompson EW. Epidermal growth factor potentiates EGFR(Y992/1173)-mediated therapeutic response of triple negative breast cancer cells to cold atmospheric plasma-activated medium. Redox Biol 2024; 69:102976. [PMID: 38052106 PMCID: PMC10746566 DOI: 10.1016/j.redox.2023.102976] [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: 10/26/2023] [Accepted: 11/24/2023] [Indexed: 12/07/2023] Open
Abstract
Cold atmospheric plasma (CAP) holds promise as a cancer-specific treatment that selectively kills various types of malignant cells. We used CAP-activated media (PAM) to utilize a range of the generated short- and long-lived reactive species. Specific antibodies, small molecule inhibitors and CRISPR/Cas9 gene-editing approaches showed an essential role for receptor tyrosine kinases, especially epidermal growth factor (EGF) receptor, in mediating triple negative breast cancer (TNBC) cell responses to PAM. EGF also dramatically enhanced the sensitivity and specificity of PAM against TNBC cells. Site-specific phospho-EGFR analysis, signal transduction inhibitors and reconstitution of EGFR-depleted cells with EGFR-mutants confirmed the role of phospho-tyrosines 992/1173 and phospholipase C gamma signaling in up-regulating levels of reactive oxygen species above the apoptotic threshold. EGF-triggered EGFR activation enhanced the sensitivity and selectivity of PAM effects on TNBC cells. The proposed approach based on the synergy of CAP and EGFR-targeted therapy may provide new opportunities to improve the clinical management of TNBC.
Collapse
Affiliation(s)
- Peiyu Wang
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine, Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, PR China; Centre for Genomics and Personalised Health, School of Biomedical Science, Faculty of Health, Queensland University of Technology, Brisbane, Queensland 4059, Australia; Translational Research Institute, Woolloongabba, Queensland 4102, Australia; State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, PR China
| | - Renwu Zhou
- State Key Laboratory of Electrical Insulation and Power Equipment, Centre for Plasma Biomedicine, School of Electrical Engineering, Xi'an Jiaotong University, Xi'an 710049, PR China
| | - Rusen Zhou
- School of Chemistry and Physics, Queensland University of Technology, Brisbane, Queensland 4000, Australia
| | - Shuo Feng
- Department of Dermatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, PR China
| | - Liqian Zhao
- Department of Neurosurgery, Institute of Brain Disease, Nanfang Hospital of Southern Medical University, Guangzhou 510515, PR China
| | - Wenshao Li
- School of Chemistry and Physics, Queensland University of Technology, Brisbane, Queensland 4000, Australia
| | - Jinyong Lin
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, PR China
| | - Aleksandra Rajapakse
- Centre for Genomics and Personalised Health, School of Biomedical Science, Faculty of Health, Queensland University of Technology, Brisbane, Queensland 4059, Australia; Translational Research Institute, Woolloongabba, Queensland 4102, Australia
| | - Chia-Hwa Lee
- Centre for Genomics and Personalised Health, School of Biomedical Science, Faculty of Health, Queensland University of Technology, Brisbane, Queensland 4059, Australia; Translational Research Institute, Woolloongabba, Queensland 4102, Australia
| | - Frank B Furnari
- Department of Medicine, University of California San Diego, California 92093, USA
| | - Antony W Burgess
- Walter and Elisa Hall Institute, Melbourne, Victoria 3052, Australia
| | - Jennifer H Gunter
- Centre for Genomics and Personalised Health, School of Biomedical Science, Faculty of Health, Queensland University of Technology, Brisbane, Queensland 4059, Australia; Translational Research Institute, Woolloongabba, Queensland 4102, Australia
| | - Gang Liu
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, PR China
| | - Kostya Ken Ostrikov
- School of Chemistry and Physics, Queensland University of Technology, Brisbane, Queensland 4000, Australia
| | - Derek J Richard
- Centre for Genomics and Personalised Health, School of Biomedical Science, Faculty of Health, Queensland University of Technology, Brisbane, Queensland 4059, Australia; Translational Research Institute, Woolloongabba, Queensland 4102, Australia; Cancer and Ageing Research Program, Woolloongabba, Queensland 4102, Australia
| | - Fiona Simpson
- Frazer Institute, The University of Queensland, Brisbane, Queensland 4102, Australia
| | - Xiaofeng Dai
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine, Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, PR China; Department of Dermatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, PR China.
| | - Erik W Thompson
- Centre for Genomics and Personalised Health, School of Biomedical Science, Faculty of Health, Queensland University of Technology, Brisbane, Queensland 4059, Australia; Translational Research Institute, Woolloongabba, Queensland 4102, Australia
| |
Collapse
|
20
|
Zhai Y, Du Y, Li G, Yu M, Hu H, Pan C, Wang D, Shi Z, Yan X, Li X, Jiang T, Zhang W. Trogocytosis of CAR molecule regulates CAR-T cell dysfunction and tumor antigen escape. Signal Transduct Target Ther 2023; 8:457. [PMID: 38143263 PMCID: PMC10749292 DOI: 10.1038/s41392-023-01708-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 10/19/2023] [Accepted: 11/15/2023] [Indexed: 12/26/2023] Open
Abstract
Chimeric antigen receptor (CAR) T-cell therapy has demonstrated clinical response in treating both hematologic malignancies and solid tumors. Although instances of rapid tumor remissions have been observed in animal models and clinical trials, tumor relapses occur with multiple therapeutic resistance mechanisms. Furthermore, while the mechanisms underlying the long-term therapeutic resistance are well-known, short-term adaptation remains less understood. However, more views shed light on short-term adaptation and hold that it provides an opportunity window for long-term resistance. In this study, we explore a previously unreported mechanism in which tumor cells employ trogocytosis to acquire CAR molecules from CAR-T cells, a reversal of previously documented processes. This mechanism results in the depletion of CAR molecules and subsequent CAR-T cell dysfunction, also leading to short-term antigen loss and antigen masking. Such type of intercellular communication is independent of CAR downstream signaling, CAR-T cell condition, target antigen, and tumor cell type. However, it is mainly dependent on antigen density and CAR sensitivity, and is associated with tumor cell cholesterol metabolism. Partial mitigation of this trogocytosis-induced CAR molecule transfer can be achieved by adaptively administering CAR-T cells with antigen density-individualized CAR sensitivities. Together, our study reveals a dynamic process of CAR molecule transfer and refining the framework of clinical CAR-T therapy for solid tumors.
Collapse
Affiliation(s)
- You Zhai
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, PR China
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, PR China
| | - Yicong Du
- Department of Urology, Peking University First Hospital, Institute of Urology, Peking University, National Urological Cancer Center, Beijing, PR China
| | - Guanzhang Li
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, PR China
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, PR China
| | - Mingchen Yu
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, PR China
| | - Huimin Hu
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, PR China
| | - Changqing Pan
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, PR China
| | - Di Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, PR China
| | - Zhongfang Shi
- Department of Pathophysiology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, PR China
| | - Xu Yan
- Department of Pathophysiology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, PR China
| | - Xuesong Li
- Department of Urology, Peking University First Hospital, Institute of Urology, Peking University, National Urological Cancer Center, Beijing, PR China
| | - Tao Jiang
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, PR China.
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, PR China.
- China National Clinical Research Center for Neurological Diseases, Beijing, PR China.
- Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, PR China.
- Research Unit of Accurate Diagnosis, Treatment, and Translational Medicine of Brain Tumors, Chinese Academy of Medical Sciences, Beijing, PR China.
- Chinese Glioma Genome Atlas Network (CGGA) and Asian Glioma Genome Atlas Network (AGGA), Beijing, PR China.
| | - Wei Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, PR China.
- China National Clinical Research Center for Neurological Diseases, Beijing, PR China.
- Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, PR China.
- Chinese Glioma Genome Atlas Network (CGGA) and Asian Glioma Genome Atlas Network (AGGA), Beijing, PR China.
| |
Collapse
|
21
|
Watabe T, Kabayama K, Naka S, Yamamoto R, Kaneda K, Serada S, Ooe K, Toyoshima A, Wang Y, Haba H, Kurimoto K, Kobayashi T, Shimosegawa E, Tomiyama N, Fukase K, Naka T. Immuno-PET and Targeted α-Therapy Using Anti-Glypican-1 Antibody Labeled with 89Zr or 211At: A Theranostic Approach for Pancreatic Ductal Adenocarcinoma. J Nucl Med 2023; 64:1949-1955. [PMID: 37827841 PMCID: PMC10690121 DOI: 10.2967/jnumed.123.266313] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 09/06/2023] [Indexed: 10/14/2023] Open
Abstract
Glypican-1 (GPC1) is overexpressed in several solid cancers and is associated with tumor progression, whereas its expression is low in normal tissues. This study aimed to evaluate the potential of an anti-GPC1 monoclonal antibody (GPC1 mAb) labeled with 89Zr or 211At as a theranostic target in pancreatic ductal adenocarcinoma. Methods: GPC1 mAb clone 01a033 was labeled with 89Zr or 211At with a deferoxamine or decaborane linker, respectively. The internalization ability of GPC1 mAb was evaluated by fluorescence conjugation using a confocal microscope. PANC-1 xenograft mice (n = 6) were intravenously administered [89Zr]GPC1 mAb (0.91 ± 0.10 MBq), and PET/CT scanning was performed for 7 d. Uptake specificity was confirmed through a comparative study using GPC1-positive (BxPC-3) and GPC1-negative (BxPC-3 GPC1-knockout) xenografts (each n = 3) and a blocking study. DNA double-strand breaks were evaluated using the γH2AX antibody. The antitumor effect was evaluated by administering [211At]GPC1 mAb (∼100 kBq) to PANC-1 xenograft mice (n = 10). Results: GPC1 mAb clone 01a033 showed increased internalization ratios over time. One day after administration, a high accumulation of [89Zr]GPC1 mAb was observed in the PANC-1 xenograft (SUVmax, 3.85 ± 0.10), which gradually decreased until day 7 (SUVmax, 2.16 ± 0.30). The uptake in the BxPC-3 xenograft was significantly higher than in the BxPC-3 GPC1-knockout xenograft (SUVmax, 4.66 ± 0.40 and 2.36 ± 0.36, respectively; P = 0.05). The uptake was significantly inhibited in the blocking group compared with the nonblocking group (percentage injected dose per gram, 7.3 ± 1.3 and 12.4 ± 3.0, respectively; P = 0.05). DNA double-strand breaks were observed by adding 150 kBq of [211At]GPC1 and were significantly suppressed by the internalization inhibitor (dynasore), suggesting a substantial contribution of the internalization ability to the antitumor effect. Tumor growth suppression was observed in PANC-1 mice after the administration of [211At]GPC1 mAb. Internalization inhibitors (prochlorperazine) significantly inhibited the therapeutic effect of [211At]GPC1 mAb, suggesting an essential role in targeted α-therapy. Conclusion: [89Zr]GPC1 mAb PET showed high tumoral uptake in the early phase after administration, and targeted α-therapy using [211At]GPC1 mAb showed tumor growth suppression. GPC1 is a promising target for future applications for the precise diagnosis of pancreatic ductal adenocarcinoma and GPC1-targeted theranostics.
Collapse
Affiliation(s)
- Tadashi Watabe
- Department of Nuclear Medicine and Tracer Kinetics, Graduate School of Medicine, Osaka University, Suita, Japan;
- Institute for Radiation Sciences, Osaka University, Suita, Japan
| | - Kazuya Kabayama
- Institute for Radiation Sciences, Osaka University, Suita, Japan
- Department of Chemistry, Graduate School of Science, Osaka University, Toyonaka, Japan
- Forefront Research Center, Graduate School of Science, Osaka University, Toyonaka, Japan
| | - Sadahiro Naka
- Department of Pharmacy, Osaka University Hospital, Suita, Japan
| | - Ryuku Yamamoto
- Department of Chemistry, Graduate School of Science, Osaka University, Toyonaka, Japan
| | - Kazuko Kaneda
- Institute for Radiation Sciences, Osaka University, Suita, Japan
- Forefront Research Center, Graduate School of Science, Osaka University, Toyonaka, Japan
| | - Satoshi Serada
- Institute for Biomedical Sciences Molecular Pathophysiology, Iwate Medical University, Yahaba, Japan
| | - Kazuhiro Ooe
- Institute for Radiation Sciences, Osaka University, Suita, Japan
| | | | - Yang Wang
- Nishina Center for Accelerator-Based Science, RIKEN, Saitama, Japan
| | - Hiromitsu Haba
- Nishina Center for Accelerator-Based Science, RIKEN, Saitama, Japan
| | - Kenta Kurimoto
- Department of Pharmacy, Osaka University Hospital, Suita, Japan
| | - Takanori Kobayashi
- Department of Nuclear Medicine and Tracer Kinetics, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Eku Shimosegawa
- Department of Molecular Imaging in Medicine, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Noriyuki Tomiyama
- Institute for Radiation Sciences, Osaka University, Suita, Japan
- Department of Radiology, Graduate School of Medicine, Osaka University, Suita, Japan; and
| | - Koichi Fukase
- Institute for Radiation Sciences, Osaka University, Suita, Japan
- Department of Chemistry, Graduate School of Science, Osaka University, Toyonaka, Japan
- Forefront Research Center, Graduate School of Science, Osaka University, Toyonaka, Japan
| | - Tetsuji Naka
- Institute for Biomedical Sciences Molecular Pathophysiology, Iwate Medical University, Yahaba, Japan
- Division of Allergy and Rheumatology, Department of Internal Medicine, School of Medicine, Iwate Medical University, Yahaba, Japan
| |
Collapse
|
22
|
Zhang Y, Liang S, Zhang Y, Liu M, Zhang K. Identification of a novel endocytosis‑associated gene signature for prognostic prediction in lung adenocarcinoma. Oncol Lett 2023; 26:511. [PMID: 37920434 PMCID: PMC10618919 DOI: 10.3892/ol.2023.14098] [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: 05/17/2023] [Accepted: 09/21/2023] [Indexed: 11/04/2023] Open
Abstract
Lung cancer is one of the most common malignant solid tumors and the leading cause of cancer-associated mortality worldwide. Endocytosis is an essential physiological activity for cells to maintain membrane homeostasis, and has been reported to serve an important role in tumorigenesis and progression. In the present study, the aim was to construct a prognostic prediction model of endocytosis-associated genes for patients with lung adenocarcinoma (LUAD). The endocytosis-associated gene signature was established using Lasso Cox regression analysis using the training set of the LUAD cohort from The Cancer Genome Atlas (TCGA) database, and verified using two datasets from the Gene Expression Omnibus (GEO) database. Kaplan-Meier survival curves were used to evaluate the effectiveness of the prognostic evaluation of patients with LUAD. Differentially expressed genes were screened in the tumor tissue of patients compared with paired paracancerous tissues. A series of candidate genes associated to the prognosis of patients with LUAD was obtained using univariate Cox's regression analysis. Using the Lasso Cox regression analysis, an appropriate risk model with 18 endocytosis-associated genes was established. A high-risk score was positively correlated with a higher tumor stage and pathologic grade. Patients with LUAD and high-risk scores had shorter survival times, increased intratumor heterogeneities and immune cell infiltration into tumor tissues, compared with those patients with LUAD and low-risk scores. The endocytosis inhibitor chloroquine could repress proliferation and increase the apoptosis of lung cancer cells. In summary, a novel endocytosis-associated gene signature was constructed using TCGA and GEO datasets. Patients with LUAD and high-risk scores, as calculated by the signature, had a poor prognosis and short survival time.
Collapse
Affiliation(s)
- Yixin Zhang
- Department of Blood Transfusion, Tianjin Hospital, Tianjin 300211, P.R. China
| | - Siwen Liang
- School of Optometry & Ophthalmology, Tianjin Medical University, Tianjin 300070, P.R. China
| | - Yan Zhang
- Department of Blood Transfusion, Tianjin Hospital, Tianjin 300211, P.R. China
| | - Minghui Liu
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Kai Zhang
- Department of Blood Transfusion, Tianjin Hospital, Tianjin 300211, P.R. China
| |
Collapse
|
23
|
Zhang Y. Targeting Epidermal Growth Factor Receptor for Cancer Treatment: Abolishing Both Kinase-Dependent and Kinase-Independent Functions of the Receptor. Pharmacol Rev 2023; 75:1218-1232. [PMID: 37339882 PMCID: PMC10595022 DOI: 10.1124/pharmrev.123.000906] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 06/11/2023] [Accepted: 06/13/2023] [Indexed: 06/22/2023] Open
Abstract
Epidermal growth factor receptor (EGFR), a receptor tyrosine kinase, is activated by ligand binding, overexpression, or mutation. It is well known for its tyrosine kinase-dependent oncogenic activities in a variety of human cancers. A large number of EGFR inhibitors have been developed for cancer treatment, including monoclonal antibodies, tyrosine kinase inhibitors, and a vaccine. The EGFR inhibitors are aimed at inhibiting the activation or the activity of EGFR tyrosine kinase. However, these agents have shown efficacy in only a few types of cancers. Drug resistance, both intrinsic and acquired, is common even in cancers where the inhibitors have shown efficacy. The drug resistance mechanism is complex and not fully known. The key vulnerability of cancer cells that are resistant to EGFR inhibitors has not been identified. Nevertheless, it has been increasingly recognized in recent years that EGFR also possesses kinase-independent oncogenic functions and that these noncanonical functions may play a crucial role in cancer resistance to EGFR inhibitors. In this review, both kinase-dependent and -independent activities of EGFR are discussed. Also discussed are the mechanisms of actions and therapeutic activities of clinically used EGFR inhibitors and sustained EGFR overexpression and EGFR interaction with other receptor tyrosine kinases to counter the EGFR inhibitors. Moreover, this review discusses emerging experimental therapeutics that have shown potential for overcoming the limitation of the current EGFR inhibitors in preclinical studies. The findings underscore the importance and feasibility of targeting both kinase-dependent and -independent functions of EGFR to enhance therapeutic efficacy and minimize drug resistance. SIGNIFICANCE STATEMENT: EGFR is a major oncogenic driver and therapeutic target, but cancer resistance to current EGFR inhibitors remains a significant unmet clinical problem. This article reviews the cancer biology of EGFR as well as the mechanisms of actions and the therapeutic efficacies of current and emerging EGFR inhibitors. The findings could potentially lead to development of more effective treatments for EGFR-positive cancers.
Collapse
Affiliation(s)
- Yuesheng Zhang
- Department of Pharmacology and Toxicology, School of Medicine, and Massey Comprehensive Cancer Center, Virginia Commonwealth University, Richmond, Virginia
| |
Collapse
|
24
|
Tim B, Kouznetsova VL, Kesari S, Tsigelny IF. Targeting of insulin receptor endocytosis as a treatment to insulin resistance. J Diabetes Complications 2023; 37:108615. [PMID: 37788593 DOI: 10.1016/j.jdiacomp.2023.108615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 09/02/2023] [Accepted: 09/25/2023] [Indexed: 10/05/2023]
Abstract
BACKGROUND Insulin resistance is the decreased effectiveness of insulin receptor function during signaling of glucose uptake. Insulin receptors are regulated by endocytosis, a process that removes receptors from the cell surface to be marked for degradation or for re-use. OBJECTIVES Our goal was to discover insulin-resistance-related genes that play key roles in endocytosis which could serve as potential biological targets to enhance insulin sensitivity. METHODS The gene mutations related to insulin resistance were elucidated from ClinVar. These were used as the seed set. Using the GeneFriends program, the genes associated with this set were elucidated and used as an enriched set for the next step. The enriched gene set network was visualized by Cytoscape. After that, using the VisANT program, the most significant cluster of genes was identified. With the help of the DAVID program, the most important KEGG pathway corresponding to the gene cluster and insulin resistance was found. Eleven genes part of the KEGG endocytosis pathway were identified. Finally, using the ChEA3 program, seven transcription factors managing these genes were defined. RESULTS Thirty-two genes of pathogenic significance in insulin resistance were elucidated, and then co-expression data for these genes were utilized. These genes were organized into clusters, one of which was singled out for its high node count of 58 genes and low p-value (p = 4.117 × 10-7). DAVID Pathways, a functional annotation tool, helped identify a set of 11 genes from a single cluster associated with the endocytosis pathway related to insulin resistance. These genes (AMPH, BIN1, CBL, DNM1, DNM2, DNM3, ITCH, SH3GL1, SH3GL2, SH3GL3, and SH3KBP1) are all involved in either clathrin-mediated endocytosis of the insulin receptor (IR) or clathrin-independent endocytosis of insulin-resistance-related G protein-coupled receptors (GPCR). They represent prime therapeutic targets to improve insulin sensitivity through modulation of transmembrane cell signaling. Using the ChEA3 database, we also found seven transcription factors (REST, MYPOP, CAMTA2, MYT1L, ZBTB18, NKX6-2, and CXXC5) that control the expression of these 11 genes. Inhibiting these key transcription factors would be another strategy to downregulate endocytosis. CONCLUSION We believe that delaying removal of insulin receptors from the cell surface would prolong signaling of glucose uptake and counteract the symptoms of insulin resistance.
Collapse
Affiliation(s)
- Bryce Tim
- IUL Science Program, San Diego, CA, USA
| | - Valentina L Kouznetsova
- San Diego Supercomputer Center, University of California, San Diego, CA, USA; CureScience Institute, San Diego, CA, USA; BiAna, La Jolla, CA, USA
| | | | - Igor F Tsigelny
- San Diego Supercomputer Center, University of California, San Diego, CA, USA; Department of Neurosciences, University of California, San Diego, CA, USA; CureScience Institute, San Diego, CA, USA; BiAna, La Jolla, CA, USA.
| |
Collapse
|
25
|
Sandker GGW, Middelburg J, Wilbrink E, Molkenboer-Kuenen J, Aarntzen E, van Hall T, Heskamp S. Longitudinal evaluation of the biodistribution and cellular internalization of the bispecific CD3xTRP1 antibody in syngeneic mouse tumor models. J Immunother Cancer 2023; 11:e007596. [PMID: 37899133 PMCID: PMC10619024 DOI: 10.1136/jitc-2023-007596] [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] [Accepted: 10/04/2023] [Indexed: 10/31/2023] Open
Abstract
BACKGROUND CD3 bispecific antibodies (CD3-bsAbs) require binding of both a tumor-associated surface antigen and CD3 for their immunotherapeutic effect. Their efficacy is, therefore, influenced by the tumor uptake and the extracellular dose. To optimize their currently limited efficacy in solid tumors, increased understanding of their pharmacokinetics and in vivo internalization is needed. METHODS Here, were studied the pharmacokinetics and in vivo internalization of CD3xTRP1, a fully murine Fc-inert bsAb, in endogenous TRP1-expressing immunocompetent male C57BL/6J mice bearing TRP1-positive and negative tumors over time. Matching bsAbs lacking TRP1-binding or CD3-binding capacity served as controls. BsAbs were radiolabeled with 111In to investigate their pharmacokinetics, target binding, and biodistribution through SPECT/CT imaging and ex vivo biodistribution analyses. Co-injection of 111In- and 125I-labeled bsAb was performed to investigate the in vivo internalization by comparing tissue concentrations of cellular residing 111In versus effluxing 125I. Antitumor therapy effects were evaluated by monitoring tumor growth and immunohistochemistry. RESULTS SPECT/CT and biodistribution analyses showed that CD3xTRP1 specifically targeted TRP1-positive tumors and CD3-rich lymphoid organ and uptake peaked 24 hours pi (KPC3-TRP1: 37.7%ID/g±5.3%ID/g, spleen: 29.0%ID/g±3.9%ID/g). Studies with control bsAbs demonstrated that uptake of CD3xTRP1 in TRP1-positive tumors and CD3-rich tissues was primarily receptor-mediated. Together with CD3xTRP1 in the circulation being mainly unattached, this indicates that CD3+ T cells are generally not traffickers of CD3-bsAbs to the tumor. Additionally, target-mediated clearance by TRP1-expressing melanocytes was not observed. We further demonstrated rapid internalization of CD3xTRP1 in KPC3-TRP1 tumors (24 hours pi: 54.9%±2.3% internalized) and CD3-rich tissues (spleen, 24 hours pi: 79.7%±0.9% internalized). Therapeutic effects by CD3xTRP1 were observed for TRP1-positive tumors and consisted of high tumor influx of CD8+ T cells and neutrophils, which corresponded with increased necrosis and growth delay. CONCLUSIONS We show that CD3xTRP1 efficiently targets TRP1-positive tumors and CD3-rich tissues primarily through receptor-mediated targeting. We further demonstrate rapid receptor-mediated internalization of CD3xTRP1 in TRP1-positive tumors and CD3-rich tissues. Even though this significantly decreases the therapeutical available dose, CD3xTRP1 still induced effective antitumor T-cell responses and inhibited tumor growth. Together, our data on the pharmacokinetics and mechanism of action of CD3xTRP1 pave the way for further optimization of CD3-bsAb therapies.
Collapse
Affiliation(s)
| | - Jim Middelburg
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands
| | - Evienne Wilbrink
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen, The Netherlands
| | | | - Erik Aarntzen
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Thorbald van Hall
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands
| | - Sandra Heskamp
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
26
|
Szewczyk-Roszczenko OK, Roszczenko P, Shmakova A, Finiuk N, Holota S, Lesyk R, Bielawska A, Vassetzky Y, Bielawski K. The Chemical Inhibitors of Endocytosis: From Mechanisms to Potential Clinical Applications. Cells 2023; 12:2312. [PMID: 37759535 PMCID: PMC10527932 DOI: 10.3390/cells12182312] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 09/07/2023] [Indexed: 09/29/2023] Open
Abstract
Endocytosis is one of the major ways cells communicate with their environment. This process is frequently hijacked by pathogens. Endocytosis also participates in the oncogenic transformation. Here, we review the approaches to inhibit endocytosis, discuss chemical inhibitors of this process, and discuss potential clinical applications of the endocytosis inhibitors.
Collapse
Affiliation(s)
| | - Piotr Roszczenko
- Department of Biotechnology, Medical University of Bialystok, Kilinskiego 1, 15-089 Bialystok, Poland; (P.R.); (A.B.)
| | - Anna Shmakova
- CNRS, UMR 9018, Institut Gustave Roussy, Université Paris-Saclay, 94800 Villejuif, France;
| | - Nataliya Finiuk
- Department of Regulation of Cell Proliferation and Apoptosis, Institute of Cell Biology of National Academy of Sciences of Ukraine, Drahomanov 14/16, 79005 Lviv, Ukraine;
| | - Serhii Holota
- Department of Pharmaceutical, Organic and Bioorganic Chemistry, Danylo Halytsky Lviv National Medical University, Pekarska 69, 79010 Lviv, Ukraine; (S.H.); (R.L.)
| | - Roman Lesyk
- Department of Pharmaceutical, Organic and Bioorganic Chemistry, Danylo Halytsky Lviv National Medical University, Pekarska 69, 79010 Lviv, Ukraine; (S.H.); (R.L.)
| | - Anna Bielawska
- Department of Biotechnology, Medical University of Bialystok, Kilinskiego 1, 15-089 Bialystok, Poland; (P.R.); (A.B.)
| | - Yegor Vassetzky
- CNRS, UMR 9018, Institut Gustave Roussy, Université Paris-Saclay, 94800 Villejuif, France;
| | - Krzysztof Bielawski
- Department of Synthesis and Technology of Drugs, Medical University of Bialystok, Kilinskiego 1, 15-089 Bialystok, Poland;
| |
Collapse
|
27
|
Causer A, Tan X, Lu X, Moseley P, Teoh SM, Molotkov N, McGrath M, Kim T, Simpson PT, Perry C, Frazer IH, Panizza B, Ladwa R, Nguyen Q, Gonzalez-Cruz JL. Deep spatial-omics analysis of Head & Neck carcinomas provides alternative therapeutic targets and rationale for treatment failure. NPJ Precis Oncol 2023; 7:89. [PMID: 37704757 PMCID: PMC10499928 DOI: 10.1038/s41698-023-00444-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 08/30/2023] [Indexed: 09/15/2023] Open
Abstract
Immune checkpoint inhibitor (ICI) therapy has had limited success (<30%) in treating metastatic recurrent Head and Neck Oropharyngeal Squamous Cell Carcinomas (OPSCCs). We postulate that spatial determinants in the tumor play a critical role in cancer therapy outcomes. Here, we describe the case of a male patient diagnosed with p16+ OPSCC and extensive lung metastatic disease who failed Nivolumab and Pembrolizumab/Lenvatinib therapies. Using advanced integrative spatial proteogenomic analysis on the patient's recurrent OPSCC tumors we demonstrate that: (i) unbiased tissue clustering based on spatial transcriptomics (ST) successfully detected tumor cells and enabled the investigation of phenotypic traits such as proliferation or drug-resistance genes in the tumor's leading-edge and core; (ii) spatial proteomic imagining used in conjunction with ST (SpiCi, Spatial Proteomics inferred Cell identification) can resolve the profiling of tumor infiltrating immune cells, (iii) ST data allows for the discovery and ranking of clinically relevant alternative medicines based on their interaction with their matching ligand-receptor. Importantly, when the spatial profiles of ICI pre- and post-failure OPSCC tumors were compared, they exhibited highly similar PD-1/PD-L1low and VEGFAhigh expression, suggesting that these new tumors were not the product of ICI resistance but rather of Lenvatinib dose reduction due to complications. Our work establishes a path for incorporating spatial-omics in clinical settings to facilitate treatment personalization.
Collapse
Affiliation(s)
- Andrew Causer
- Institute of Molecular Biology, The University of Queensland, Brisbane, QLD, Australia
| | - Xiao Tan
- Frazer Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Xuehan Lu
- Frazer Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Philip Moseley
- Frazer Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Siok M Teoh
- Institute of Molecular Biology, The University of Queensland, Brisbane, QLD, Australia
| | - Natalie Molotkov
- Frazer Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Margaret McGrath
- Department of Medical Oncology, Princess Alexandra Hospital, Brisbane, QLD, Australia
| | - Taehyun Kim
- Pathology Queensland, Royal Brisbane & Women's Hospital, Brisbane, QLD, Australia
| | - Peter T Simpson
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| | - Christopher Perry
- Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
- Department of Otolaryngology-Head & Neck surgery, Princess Alexandra Hospital, Brisbane, QLD, Australia
| | - Ian H Frazer
- Frazer Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Benedict Panizza
- Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
- Department of Otolaryngology-Head & Neck surgery, Princess Alexandra Hospital, Brisbane, QLD, Australia
| | - Rahul Ladwa
- Department of Medical Oncology, Princess Alexandra Hospital, Brisbane, QLD, Australia
- Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| | - Quan Nguyen
- Institute of Molecular Biology, The University of Queensland, Brisbane, QLD, Australia.
| | | |
Collapse
|
28
|
Subasic CN, Butcher NJ, Simpson F, Minchin RF, Kaminskas LM. Dose-Dependent Effect of Phenothiazines as Dynamin II Inhibitors on the Uptake of PEGylated Liposomes by Endocytic Cells and In Vivo Pharmacokinetics of PEGylated Liposomal Doxorubicin in Rats. Mol Pharm 2023; 20:4468-4477. [PMID: 37548597 DOI: 10.1021/acs.molpharmaceut.3c00102] [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] [Indexed: 08/08/2023]
Abstract
Dynamin II (dynII) plays a significant role in the internalization pathways of endocytic cells, by allowing membrane invaginations to "bud off". An important class of dynII inhibitors that are used clinically are phenothiazines, such as prochlorperazine (PCZ). PCZ is an antipsychotic drug but is also currently in clinical trials at higher concentrations as an adjuvant in cancer patients that increases the efficacy of monoclonal antibodies at high intravenous doses. It is unknown, however, whether high-dose dynII inhibitors have the potential to alter the pharmacokinetics of co-administered chemotherapeutic nanomedicines that are largely cleared via the mononuclear phagocyte system. This work therefore sought to investigate the impact of clinically relevant concentrations of phenothiazines, PCZ and thioridazine, on in vitro liposome endocytosis and in vivo liposome pharmacokinetics after PCZ infusion in rats. The uptake of fluorescently labeled PEGylated liposomes into differentiated and undifferentiated THP-1 and RAW246.7 cells, and primary human peripheral white blood cells, was investigated via flow cytometry after co-incubation with dynII inhibitors. The IV pharmacokinetics of PEGylated liposomes were also investigated in rats after a 20 min infusion with PCZ. Phenothiazines and dyngo4a reduced the uptake of PEGylated liposomes by THP-1 and RAW264.7 cells in a concentration-dependent manner in vitro. However, dynII inhibitors did not alter the mean uptake of liposomes by human peripheral white blood cells, but endocytic white cells from some donors exhibited sensitivity to phenothiazine exposure. When a clinically relevant dose of PCZ was co-administered with PEGylated liposomal doxorubicin (Caelyx/Doxil) in rats, the pharmacokinetics and biodistribution of liposomes were unaltered. These data suggest that while clinically relevant doses of dynII inhibitors can inhibit the uptake of liposomes by endocytic cells in vitro, they are unlikely to significantly affect the pharmacokinetics of long-circulating, co-administered liposomes.
Collapse
Affiliation(s)
- Christopher N Subasic
- School of Biomedical Sciences, The University of Queensland, St Lucia QLD 4072, Australia
| | - Neville J Butcher
- School of Biomedical Sciences, The University of Queensland, St Lucia QLD 4072, Australia
| | - Fiona Simpson
- Frazer Institute, The University of Queensland, St Lucia QLD 4072, Australia
| | - Rodney F Minchin
- School of Biomedical Sciences, The University of Queensland, St Lucia QLD 4072, Australia
| | - Lisa M Kaminskas
- School of Biomedical Sciences, The University of Queensland, St Lucia QLD 4072, Australia
| |
Collapse
|
29
|
Zeng B, Moi D, Tolley L, Molotkov N, Frazer IH, Perry C, Dolcetti R, Mazzieri R, Cruz JLG. Skin-Grafting and Dendritic Cell "Boosted" Humanized Mouse Models Allow the Pre-Clinical Evaluation of Therapeutic Cancer Vaccines. Cells 2023; 12:2094. [PMID: 37626903 PMCID: PMC10453599 DOI: 10.3390/cells12162094] [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: 06/21/2023] [Revised: 07/25/2023] [Accepted: 08/09/2023] [Indexed: 08/27/2023] Open
Abstract
Vaccines have been hailed as one of the most remarkable medical advancements in human history, and their potential for treating cancer by generating or expanding anti-tumor T cells has garnered significant interest in recent years. However, the limited efficacy of therapeutic cancer vaccines in clinical trials can be partially attributed to the inadequacy of current preclinical mouse models in recapitulating the complexities of the human immune system. In this study, we developed two innovative humanized mouse models to assess the immunogenicity and therapeutic effectiveness of vaccines targeting human papillomavirus (HPV16) antigens and delivering tumor antigens to human CD141+ dendritic cells (DCs). Both models were based on the transference of human peripheral blood mononuclear cells (PBMCs) into immunocompromised HLA-A*02-NSG mice (NSG-A2), where the use of fresh PBMCs boosted the engraftment of human cells up to 80%. The dynamics of immune cells in the PBMC-hu-NSG-A2 mice demonstrated that T cells constituted the vast majority of engrafted cells, which progressively expanded over time and retained their responsiveness to ex vivo stimulation. Using the PBMC-hu-NSG-A2 system, we generated a hyperplastic skin graft model expressing the HPV16-E7 oncogene. Remarkably, human cells populated the skin grafts, and upon vaccination with a DNA vaccine encoding an HPV16-E6/E7 protein, rapid rejection targeted to the E7-expressing skin was detected, underscoring the capacity of the model to mount a vaccine-specific response. To overcome the decline in DC numbers observed over time in PBMC-hu-NSG-A2 animals, we augmented the abundance of CD141+ DCs, the specific targets of our tailored nanoemulsions (TNEs), by transferring additional autologous PBMCs pre-treated in vitro with the growth factor Flt3-L. The Flt3-L treatment bolstered CD141+ DC numbers, leading to potent antigen-specific CD4+ and CD8+ T cell responses in vivo, which caused the regression of pre-established triple-negative breast cancer and melanoma tumors following CD141+ DC-targeting TNE vaccination. Notably, using HLA-A*02-matching PBMCs for humanizing NSG-A2 mice resulted in a delayed onset of graft-versus-host disease and enhanced the efficacy of the TNE vaccination compared with the parental NSG strain. In conclusion, we successfully established two humanized mouse models that exhibited strong antigen-specific responses and demonstrated tumor regression following vaccination. These models serve as valuable platforms for assessing the efficacy of therapeutic cancer vaccines targeting HPV16-dysplastic skin and diverse tumor antigens specifically delivered to CD141+ DCs.
Collapse
Affiliation(s)
- Bijun Zeng
- Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia
- Frazer Institute, The University of Queensland, Brisbane, QLD 4102, Australia
| | - Davide Moi
- Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia
- Frazer Institute, The University of Queensland, Brisbane, QLD 4102, Australia
| | - Lynn Tolley
- Frazer Institute, The University of Queensland, Brisbane, QLD 4102, Australia
| | - Natalie Molotkov
- Frazer Institute, The University of Queensland, Brisbane, QLD 4102, Australia
| | - Ian Hector Frazer
- Frazer Institute, The University of Queensland, Brisbane, QLD 4102, Australia
| | - Christopher Perry
- Faculty of Medicine, The University of Queensland, Brisbane, QLD 4072, Australia
- Department of Otolaryngology, Princess Alexandra Hospital, Brisbane, QLD 4102, Australia
| | - Riccardo Dolcetti
- Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia
- Frazer Institute, The University of Queensland, Brisbane, QLD 4102, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC 3010, Australia
- Department of Microbiology and Immunology, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - Roberta Mazzieri
- Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia
- Frazer Institute, The University of Queensland, Brisbane, QLD 4102, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - Jazmina L. G. Cruz
- Frazer Institute, The University of Queensland, Brisbane, QLD 4102, Australia
| |
Collapse
|
30
|
Odell LR, Jones NC, Chau N, Robertson MJ, Ambrus JI, Deane FM, Young KA, Whiting A, Xue J, Prichard K, Daniel JA, Gorgani NN, O'Brien TJ, Robinson PJ, McCluskey A. The sulfonadyns: a class of aryl sulfonamides inhibiting dynamin I GTPase and clathrin mediated endocytosis are anti-seizure in animal models. RSC Med Chem 2023; 14:1492-1511. [PMID: 37593570 PMCID: PMC10429932 DOI: 10.1039/d2md00371f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 04/15/2023] [Indexed: 08/19/2023] Open
Abstract
We show that dansylcadaverine (1) a known in-cell inhibitor of clathrin mediated endocytosis (CME), moderately inhibits dynamin I (dynI) GTPase activity (IC50 45 μM) and transferrin (Tfn) endocytosis in U2OS cells (IC50 205 μM). Synthesis gave a new class of GTP-competitive dynamin inhibitors, the Sulfonadyns™. The introduction of a terminal cinnamyl moiety greatly enhanced dynI inhibition. Rigid diamine or amide links between the dansyl and cinnamyl moieties were detrimental to dynI inhibition. Compounds with in vitro inhibition of dynI activity <10 μM were tested in-cell for inhibition of CME. These data unveiled a number of compounds, e.g. analogues 33 ((E)-N-(6-{[(3-(4-bromophenyl)-2-propen-1-yl]amino}hexyl)-5-isoquinolinesulfonamide)) and 47 ((E)-N-(3-{[3-(4-bromophenyl)-2-propen-1-yl]amino}propyl)-1-naphthalenesulfonamide)isomers that showed dyn IC50 <4 μM, IC50(CME) <30 μM and IC50(SVE) from 12-265 μM. Both analogues (33 and 47) are at least 10 times more potent that the initial lead, dansylcadaverine (1). Enzyme kinetics revealed these sulfonamide analogues as being GTP competitive inhibitors of dynI. Sulfonadyn-47, the most potent SVE inhibitor observed (IC50(SVE) = 12.3 μM), significantly increased seizure threshold in a 6 Hz mouse psychomotor seizure test at 30 (p = 0.003) and 100 mg kg-1 ip (p < 0.0001), with similar anti-seizure efficacy to the established anti-seizure medication, sodium valproate (400 mg kg-1). The Sulfonadyn™ class of drugs target dynamin and show promise as novel leads for future anti-seizure medications.
Collapse
Affiliation(s)
- Luke R Odell
- Chemistry, Centre for Chemical Biology, School of Environmental & Life Science, The University of Newcastle University Drive Callaghan NSW 2308 Australia +612 4921 5472 +612 4921 6486
| | - Nigel C Jones
- Department of Neuroscience, Central Clinical School, Monash University Melbourne Victoria 3004 Australia
- Department of Neurology, The Alfred Hospital Commercial Road Melbourne Victoria 3004 Australia
- Department of Medicine (Royal Melbourne Hospital), University of Melbourne Parkville Victoria 3052 Australia
| | - Ngoc Chau
- Cell Signaling Unit, Children's Medical Research Institute, The University of Sydney 214 Hawkesbury Road Westmead NSW 2145 Australia +612 8865 2915
| | - Mark J Robertson
- Chemistry, Centre for Chemical Biology, School of Environmental & Life Science, The University of Newcastle University Drive Callaghan NSW 2308 Australia +612 4921 5472 +612 4921 6486
| | - Joseph I Ambrus
- Chemistry, Centre for Chemical Biology, School of Environmental & Life Science, The University of Newcastle University Drive Callaghan NSW 2308 Australia +612 4921 5472 +612 4921 6486
| | - Fiona M Deane
- Chemistry, Centre for Chemical Biology, School of Environmental & Life Science, The University of Newcastle University Drive Callaghan NSW 2308 Australia +612 4921 5472 +612 4921 6486
| | - Kelly A Young
- Chemistry, Centre for Chemical Biology, School of Environmental & Life Science, The University of Newcastle University Drive Callaghan NSW 2308 Australia +612 4921 5472 +612 4921 6486
| | - Ainslie Whiting
- Cell Signaling Unit, Children's Medical Research Institute, The University of Sydney 214 Hawkesbury Road Westmead NSW 2145 Australia +612 8865 2915
| | - Jing Xue
- Cell Signaling Unit, Children's Medical Research Institute, The University of Sydney 214 Hawkesbury Road Westmead NSW 2145 Australia +612 8865 2915
| | - Kate Prichard
- Chemistry, Centre for Chemical Biology, School of Environmental & Life Science, The University of Newcastle University Drive Callaghan NSW 2308 Australia +612 4921 5472 +612 4921 6486
| | - James A Daniel
- Cell Signaling Unit, Children's Medical Research Institute, The University of Sydney 214 Hawkesbury Road Westmead NSW 2145 Australia +612 8865 2915
| | - Nick N Gorgani
- Cell Signaling Unit, Children's Medical Research Institute, The University of Sydney 214 Hawkesbury Road Westmead NSW 2145 Australia +612 8865 2915
| | - Terence J O'Brien
- Department of Neurology, The Alfred Hospital Commercial Road Melbourne Victoria 3004 Australia
- Department of Medicine (Royal Melbourne Hospital), University of Melbourne Parkville Victoria 3052 Australia
| | - Phillip J Robinson
- Cell Signaling Unit, Children's Medical Research Institute, The University of Sydney 214 Hawkesbury Road Westmead NSW 2145 Australia +612 8865 2915
| | - Adam McCluskey
- Chemistry, Centre for Chemical Biology, School of Environmental & Life Science, The University of Newcastle University Drive Callaghan NSW 2308 Australia +612 4921 5472 +612 4921 6486
| |
Collapse
|
31
|
Dai K, Gong C, Xu Y, Ding F, Qi X, Tu X, Yu L, Liu X, Li J, Fan C, Yan H, Yao G. Single-Stranded RNA Origami-Based Epigenetic Immunomodulation. NANO LETTERS 2023; 23:7188-7196. [PMID: 37499095 DOI: 10.1021/acs.nanolett.3c02185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
The integration of functional modules at the molecular level into RNA nanostructures holds great potential for expanding their applications. However, the quantitative integration of nucleoside analogue molecules into RNA nanostructures and their impact on the structure and function of RNA nanostructures remain largely unexplored. Here, we report a transcription-based approach to controllably integrate multiple nucleoside analogues into a 2000 nucleotide (nt) single-stranded RNA (ssRNA) origami nanostructure. The resulting integrated ssRNA origami preserves the morphology and biostability of the original ssRNA origami. Moreover, the integration of nucleoside analogues introduced new biomedical functions to ssRNA origamis, including innate immune recognition and regulation after the precise integration of epigenetic nucleoside analogues and synergistic effects on tumor cell killing after integration of therapeutic nucleoside analogues. This study provides a promising approach for the quantitative integration of functional nucleoside analogues into RNA nanostructures at the molecular level, thereby offering valuable insights for the development of multifunctional ssRNA origamis.
Collapse
Affiliation(s)
- Kun Dai
- School of Chemistry and Chemical Engineering, New Cornerstone Science Laboratory, Frontiers Science Center for Transformative Molecules, Zhangjiang Institute for Advanced Study and National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Chen Gong
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yang Xu
- School of Molecular Sciences and Biodesign Center for Molecular Design and Biomimetics, Biodesign Institute, Arizona State University, Tempe, Arizona 85287, United States
| | - Fei Ding
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acids Chemistry and Nanomedicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Xiaodong Qi
- School of Molecular Sciences and Biodesign Center for Molecular Design and Biomimetics, Biodesign Institute, Arizona State University, Tempe, Arizona 85287, United States
| | - Xinyi Tu
- School of Molecular Sciences and Biodesign Center for Molecular Design and Biomimetics, Biodesign Institute, Arizona State University, Tempe, Arizona 85287, United States
| | - Lu Yu
- School of Molecular Sciences and Biodesign Center for Molecular Design and Biomimetics, Biodesign Institute, Arizona State University, Tempe, Arizona 85287, United States
| | - Xiaoguo Liu
- School of Chemistry and Chemical Engineering, New Cornerstone Science Laboratory, Frontiers Science Center for Transformative Molecules, Zhangjiang Institute for Advanced Study and National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Jiang Li
- Institute of Materiobiology, Department of Chemistry, College of Science, Shanghai University, Shanghai 200444, China
| | - Chunhai Fan
- School of Chemistry and Chemical Engineering, New Cornerstone Science Laboratory, Frontiers Science Center for Transformative Molecules, Zhangjiang Institute for Advanced Study and National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Hao Yan
- School of Molecular Sciences and Biodesign Center for Molecular Design and Biomimetics, Biodesign Institute, Arizona State University, Tempe, Arizona 85287, United States
| | - Guangbao Yao
- School of Chemistry and Chemical Engineering, New Cornerstone Science Laboratory, Frontiers Science Center for Transformative Molecules, Zhangjiang Institute for Advanced Study and National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| |
Collapse
|
32
|
Panikar SS, Keltee N, Berry NK, Shmuel S, Fisher ZT, Brown E, Zidel A, Mabry A, Pereira PMR. Metformin-Induced Receptor Turnover Alters Antibody Accumulation in HER-Expressing Tumors. J Nucl Med 2023; 64:1195-1202. [PMID: 37268425 PMCID: PMC10394312 DOI: 10.2967/jnumed.122.265248] [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: 11/23/2022] [Revised: 03/30/2023] [Indexed: 06/04/2023] Open
Abstract
Metformin has effects beyond its antihyperglycemic properties, including altering the localization of membrane receptors in cancer cells. Metformin decreases human epidermal growth factor receptor (HER) membrane density. Depletion of cell-surface HER decreases antibody-tumor binding for imaging and therapeutic approaches. Here, we used HER-targeted PET to annotate antibody-tumor binding in mice treated with metformin. Methods: Small-animal PET annotated antibody binding in HER-expressing xenografts on administration of an acute versus a daily dose schedule of metformin. Analyses at the protein level in the total, membrane, and internalized cell extracts were performed to determine receptor endocytosis, HER surface and internalized protein levels, and HER phosphorylation. Results: At 24 h after injection of radiolabeled anti-HER antibodies, control tumors had higher antibody accumulation than tumors treated with an acute dose of metformin. These differences were temporal, and by 72 h, tumor uptake in acute cohorts was similar to uptake in control. Additional PET imaging revealed a sustained decrease in tumor uptake on daily metformin treatment compared with control and acute metformin cohorts. The effects of metformin on membrane HER were reversible, and after its removal, antibody-tumor binding was restored. The time- and dose-dependent effects of metformin-induced HER depletion observed preclinically were validated with immunofluorescence, fractionation, and protein analysis cell assays. Conclusion: The findings that metformin decreases cell-surface HER receptors and reduces antibody-tumor binding may have significant implications for the use of antibodies targeting these receptors in cancer treatment and molecular imaging.
Collapse
Affiliation(s)
- Sandeep Surendra Panikar
- Department of Radiology, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri
| | - Nai Keltee
- Department of Radiology, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri
| | - Na-Keysha Berry
- Department of Radiology, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri
| | - Shayla Shmuel
- Department of Radiology, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri
| | - Zachary T Fisher
- Department of Radiology, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri
| | - Emma Brown
- Department of Radiology, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri
| | - Abbey Zidel
- Department of Radiology, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri
- Department of Biology, Washington University School of Medicine, St. Louis, Missouri; and
| | - Alex Mabry
- Department of Radiology, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri
- Department of Biological and Biomedical Sciences, Washington University School of Medicine, St. Louis, Missouri
| | - Patrícia M R Pereira
- Department of Radiology, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri;
| |
Collapse
|
33
|
Zhao S, Zhuang W, Han B, Song Z, Guo W, Luo F, Wu L, Hu Y, Wang H, Dong X, Jiang D, Wang M, Miao L, Wang Q, Zhang J, Fu Z, Huang Y, Xu C, Hu L, Li L, Hu R, Yang Y, Li M, Yang X, Zhang L, Huang Y, Fang W. Phase 1b trial of anti-EGFR antibody JMT101 and Osimertinib in EGFR exon 20 insertion-positive non-small-cell lung cancer. Nat Commun 2023; 14:3468. [PMID: 37308490 DOI: 10.1038/s41467-023-39139-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 05/31/2023] [Indexed: 06/14/2023] Open
Abstract
EGFR exon 20 insertion (20ins)-positive non-small-cell lung cancer (NSCLC) is an uncommon disease with limited therapeutic options and dismal prognosis. Here we report the activity, tolerability, potential mechanisms of response and resistance for dual targeting EGFR 20ins with JMT101 (anti-EGFR monoclonal antibody) plus osimertinib from preclinical models and an open label, multi-center phase 1b trial (NCT04448379). Primary endpoint of the trial is tolerability. Secondary endpoints include objective response rate, duration of response, disease control rate, progression free survival, overall survival, the pharmacokinetic profile of JMT101, occurrence of anti-drug antibodies and correlation between biomarkers and clinical outcomes. A total of 121 patients are enrolled to receive JMT101 plus osimertinib 160 mg. The most common adverse events are rash (76.9%) and diarrhea (63.6%). The confirmed objective response rate is 36.4%. Median progression-free survival is 8.2 months. Median duration of response is unreached. Subgroup analyses were performed by clinicopathological features and prior treatments. In patients with platinum-refractory diseases (n = 53), confirmed objective response rate is 34.0%, median progression-free survival is 9.2 months and median duration of response is 13.3 months. Responses are observed in distinct 20ins variants and intracranial lesions. Intracranial disease control rate is 87.5%. Confirmed intracranial objective response rate is 25%.
Collapse
Affiliation(s)
- Shen Zhao
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Wu Zhuang
- Department of Thoracic Oncology, Fujian Cancer Hospital, Fuzhou, China
| | - Baohui Han
- Department of Pulmonary Medicine, Shanghai Chest Hospital, Shanghai, China
| | - Zhengbo Song
- Department of Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, China
| | - Wei Guo
- Department of Respiratory Medicine, Shanxi Provincial Cancer Hospital, Taiyuan, China
| | - Feng Luo
- Lung Cancer Center, West China School of Medicine and West China Hospital, Sichuan University, Chengdu, China
| | - Lin Wu
- Department of Thoracic Medicine, Hunan Cancer Hospital, Changsha, China
| | - Yi Hu
- Department of Medical Oncology, Chinese PLA General Hospital, Beijing, China
| | - Huijuan Wang
- Department of Medical Oncology, Henan Cancer Hospital, Zhengzhou, China
| | - Xiaorong Dong
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Da Jiang
- Department of Medical Oncology, The Fourth Hospital of Hebei Medical University and Hebei Tumor Hospital, Shijiazhuang, China
| | - Mingxia Wang
- Department of Clinical Pharmacology, The Fourth Hospital of Hebei Medical University and Hebei Tumor Hospital, Shijiazhuang, China
| | - Liyun Miao
- Department of Respiratory and Critical Care Medicine, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Qian Wang
- Department of Respiratory Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| | - Junping Zhang
- Department of Medical Oncology, Shanxi Bethune Hospital, Taiyuan, China
| | - Zhenming Fu
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yihua Huang
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Chunwei Xu
- Department of Respiratory Medicine, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Longyu Hu
- HaploX Biotechnology Co,. Ltd., Shenzhen, China
| | - Lei Li
- Clinical Science Division, CSPC Pharmaceutical Group Co., Ltd, Shijiazhuang, China
| | - Rong Hu
- Clinical Science Division, CSPC Pharmaceutical Group Co., Ltd, Shijiazhuang, China
| | - Yang Yang
- Clinical Science Division, CSPC Pharmaceutical Group Co., Ltd, Shijiazhuang, China
| | - Mengke Li
- Clinical Science Division, CSPC Pharmaceutical Group Co., Ltd, Shijiazhuang, China
| | - Xiugao Yang
- Clinical Science Division, CSPC Pharmaceutical Group Co., Ltd, Shijiazhuang, China.
| | - Li Zhang
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China.
| | - Yan Huang
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China.
| | - Wenfeng Fang
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China.
| |
Collapse
|
34
|
Banushi B, Joseph SR, Lum B, Lee JJ, Simpson F. Endocytosis in cancer and cancer therapy. Nat Rev Cancer 2023:10.1038/s41568-023-00574-6. [PMID: 37217781 DOI: 10.1038/s41568-023-00574-6] [Citation(s) in RCA: 41] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/11/2023] [Indexed: 05/24/2023]
Abstract
Endocytosis is a complex process whereby cell surface proteins, lipids and fluid from the extracellular environment are packaged, sorted and internalized into cells. Endocytosis is also a mechanism of drug internalization into cells. There are multiple routes of endocytosis that determine the fate of molecules, from degradation in the lysosomes to recycling back to the plasma membrane. The overall rates of endocytosis and temporal regulation of molecules transiting through endocytic pathways are also intricately linked with signalling outcomes. This process relies on an array of factors, such as intrinsic amino acid motifs and post-translational modifications. Endocytosis is frequently disrupted in cancer. These disruptions lead to inappropriate retention of receptor tyrosine kinases on the tumour cell membrane, changes in the recycling of oncogenic molecules, defective signalling feedback loops and loss of cell polarity. In the past decade, endocytosis has emerged as a pivotal regulator of nutrient scavenging, response to and regulation of immune surveillance and tumour immune evasion, tumour metastasis and therapeutic drug delivery. This Review summarizes and integrates these advances into the understanding of endocytosis in cancer. The potential to regulate these pathways in the clinic to improve cancer therapy is also discussed.
Collapse
Affiliation(s)
- Blerida Banushi
- Frazer Institute, University of Queensland, Woolloongabba, Queensland, Australia
| | - Shannon R Joseph
- Frazer Institute, University of Queensland, Woolloongabba, Queensland, Australia
| | - Benedict Lum
- Frazer Institute, University of Queensland, Woolloongabba, Queensland, Australia
| | - Jason J Lee
- Frazer Institute, University of Queensland, Woolloongabba, Queensland, Australia
| | - Fiona Simpson
- Frazer Institute, University of Queensland, Woolloongabba, Queensland, Australia.
| |
Collapse
|
35
|
Placidi G, Mattu C, Ciardelli G, Campa CC. Small molecules targeting endocytic uptake and recycling pathways. Front Cell Dev Biol 2023; 11:1125801. [PMID: 36968200 PMCID: PMC10036367 DOI: 10.3389/fcell.2023.1125801] [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: 12/16/2022] [Accepted: 02/23/2023] [Indexed: 03/12/2023] Open
Abstract
Over the past years a growing number of studies highlighted the pivotal role of intracellular trafficking in cell physiology. Among the distinct transport itineraries connecting the endocytic system, both internalization (endocytosis) and recycling (endocytic recycling) pathways were found fundamental to ensure cellular sensing, cell-to-cell communication, cellular division, and collective cell migration in tissue specific-contexts. Consistently, the dysregulation of endocytic trafficking pathways is correlated with several human diseases including both cancers and neurodegeneration. Aimed at suppress specific intracellular trafficking routes involved in disease onset and progression, huge efforts have been made to identify small molecule inhibitors with suitable pharmacological properties for in vivo administration. Here, we review most used drugs and recently discovered small molecules able to block endocytosis and endocytic recycling pathways. We characterize such pharmacological inhibitors by emphasizing their target specificity, molecular affinity, biological activity and efficacy in both in vitro and in vivo experimental models.
Collapse
Affiliation(s)
- Giampaolo Placidi
- Italian Institute for Genomic Medicine, Candiolo, Italy
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy
| | - Clara Mattu
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy
| | - Gianluca Ciardelli
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy
- Chemical-Physical Processes, National Research Council (CNR-IPCF), Pisa, Italy
| | - Carlo C. Campa
- Italian Institute for Genomic Medicine, Candiolo, Italy
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
| |
Collapse
|
36
|
The endocytosis inhibitor dynasore induces a DNA damage response pathway that can be manipulated for enhanced apoptosis. Biochem Biophys Res Commun 2023; 645:1-9. [PMID: 36657293 DOI: 10.1016/j.bbrc.2023.01.035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 01/04/2023] [Accepted: 01/12/2023] [Indexed: 01/15/2023]
Abstract
Endocytosis has been shown to play an important role in cancer proliferation and metastasis. Recent studies have accumulated evidence that endocytosis inhibitors suppress in vitro and in vivo proliferation and migration. In addition, endocytosis inhibition has been shown to induce apoptosis, but its mechanism remains largely unclear. In this study, we found that the endocytosis inhibitor dynasore causes a cell viability reduction in multiple cancer cell lines, especially in hematopoietic cancers. Dynasore induced massive apoptosis and an S-phase progression delay. In addition, dynasore activated the ATR-Chk1 DNA damage response, which suggests a single-stranded DNA exposure induced by DNA replication stress. Furthermore, an ATR inhibitor sensitized the dynasore-induced apoptosis. These findings suggest that endocytosis inhibitors may have an ability to suppress DNA replication, a common mechanism of genotoxic chemotherapies targeting cancer, and that the anti-cancer effects of endocytosis inhibitors may be sensitized by DNA damage response inhibitors.
Collapse
|
37
|
Head and neck cancer patient-derived tumouroid cultures: opportunities and challenges. Br J Cancer 2023; 128:1807-1818. [PMID: 36765173 PMCID: PMC10147637 DOI: 10.1038/s41416-023-02167-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 01/11/2023] [Accepted: 01/16/2023] [Indexed: 02/12/2023] Open
Abstract
Head and neck cancers (HNC) are the seventh most prevalent cancer type globally. Despite their common categorisation, HNCs are a heterogeneous group of malignancies arising in various anatomical sites within the head and neck region. These cancers exhibit different clinical and biological manifestations, and this heterogeneity also contributes to the high rates of treatment failure and mortality. To evaluate patients who will respond to a particular treatment, there is a need to develop in vitro model systems that replicate in vivo tumour status. Among the methods developed, patient-derived cancer organoids, also known as tumouroids, recapitulate in vivo tumour characteristics including tumour architecture. Tumouroids have been used for general disease modelling and genetic instability studies in pan-cancer research. However, a limited number of studies have thus far been conducted using tumouroid-based drug screening. Studies have concluded that tumouroids can play an essential role in bringing precision medicine for highly heterogenous cancer types such as HNC.
Collapse
|
38
|
Wong JKM, Dolcetti R, Rhee H, Simpson F, Souza-Fonseca-Guimaraes F. Weaponizing natural killer cells for solid cancer immunotherapy. Trends Cancer 2023; 9:111-121. [PMID: 36379852 DOI: 10.1016/j.trecan.2022.10.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 10/16/2022] [Accepted: 10/19/2022] [Indexed: 11/13/2022]
Abstract
Enhancing natural killer (NK) cell-based innate immunity has become a promising strategy for immunotherapy against hard-to-cure solid cancers. Monoclonal antibody (mAb) therapy has been used to activate NK-cell-mediated antibody-dependent cellular cytotoxicity (ADCC) towards solid cancers. Cancer cells, however, can subvert immunosurveillance using multiple immunosuppressive mechanisms, which may hamper NK cell ADCC. Mechanisms to safely enhance ADCC by NK cells, such as utilizing temporary inhibition of receptor endocytosis to increase antibody presentation from target to effector cells can now be used to enhance NK-cell-mediated ADCC against solid tumors. This review summarizes and discusses the recent advances in the field and highlights current and potential future use of immunotherapies to maximize the therapeutic efficacy of innate anticancer immunity.
Collapse
Affiliation(s)
- Joshua K M Wong
- The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, QLD 4102, Australia
| | - Riccardo Dolcetti
- The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, QLD 4102, Australia; Peter MacCallum Cancer Centre, Melbourne, Victoria 3000, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Victoria 3010, Australia; Department of Microbiology and Immunology, The University of Melbourne, Victoria 3010, Australia
| | - Handoo Rhee
- Princess Alexandra Hospital and Queen Elizabeth Jubilee II Hospital, Woolloongabba, QLD 4102, Australia; The School of Medicine, The University of Queensland, Woolloongabba, QLD 4102, Australia
| | - Fiona Simpson
- The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, QLD 4102, Australia
| | | |
Collapse
|
39
|
Cai X, Shi W, Lian J, Zhang G, Cai Y, Zhu L. Characterization of immune landscape and development of a novel N7-methylguanine-related gene signature to aid therapy in recurrent aphthous stomatitis. Inflamm Res 2023; 72:133-148. [PMID: 36352034 DOI: 10.1007/s00011-022-01665-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 10/24/2022] [Accepted: 10/26/2022] [Indexed: 11/11/2022] Open
Abstract
OBJECTIVES Recurrent aphthous stomatitis (RAS) is the most common inflammatory disease of the oral mucosa resulting in an impaired life quality and even leading to tumors in susceptible populations. N7-Methylguanine (m7G) plays a vital role in various cellular activities but has not yet been investigated in RAS. We aimed at picturing the immune landscape and constructing an m7G-related gene signature, and investigating candidate drugs and gene-disease association to aid therapy for RAS. METHODS For our study, m7G-related differentially expressed genes (DEGs) were screened. We outlined the immune microenvironment and studied the correlations between the m7G-related DEGs and immune cells/pathways. We performed functional enrichment analyses and constructed the protein-protein interaction (PPI) and multifactor regulatory network in RAS. The m7G-related hub genes were extracted to formulate the corresponding m7G predictive signature. RESULTS We obtained 11 m7G-related DEGs and studied a comprehensive immune infiltration landscape, which indicated several immune markers as possible immunotherapeutic targets. The PPI and multifactor regulatory network was constructed and 4 hub genes (DDX58, IFI27, IFIT5, and PML) were identified, followed by validation of the corresponding m7G predictive signature for RAS. GO and KEGG analyses revealed the participation of JAK-STAT and several immune-related pathways. Finally, we suggested candidate drugs and gene-disease associations for potential RAS medical interventions. CONCLUSIONS The present study pictured a comprehensive immune infiltration landscape and suggested that m7G played a vital role in RAS through immune-related pathways. This study provided new insight for the future investigation of the mechanisms and therapeutic strategies for RAS.
Collapse
Affiliation(s)
- Xueyao Cai
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, 639 Zhi-Zao-Ju Road, Huangpu District, Shanghai, 200011, China
| | - Wenjun Shi
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, 639 Zhi-Zao-Ju Road, Huangpu District, Shanghai, 200011, China
| | - Jie Lian
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, 639 Zhi-Zao-Ju Road, Huangpu District, Shanghai, 200011, China
| | - Guoyou Zhang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, 639 Zhi-Zao-Ju Road, Huangpu District, Shanghai, 200011, China
| | - Yuchen Cai
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, 639 Zhi-Zao-Ju Road, Huangpu District, Shanghai, 200011, China.
| | - Lian Zhu
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, 639 Zhi-Zao-Ju Road, Huangpu District, Shanghai, 200011, China.
| |
Collapse
|
40
|
Hosseini R, Asef-Kabiri L, Sarvnaz H, Ghanavatinejad A, Rezayat F, Eskandari N, Akbari ME. Blockade of exosome release alters HER2 trafficking to the plasma membrane and gives a boost to Trastuzumab. CLINICAL & TRANSLATIONAL ONCOLOGY : OFFICIAL PUBLICATION OF THE FEDERATION OF SPANISH ONCOLOGY SOCIETIES AND OF THE NATIONAL CANCER INSTITUTE OF MEXICO 2023; 25:185-198. [PMID: 36018441 DOI: 10.1007/s12094-022-02925-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 08/05/2022] [Indexed: 01/07/2023]
Abstract
OBJECTIVE(S) Exosomal HER2 has been evidenced to interfere with antibody-induced anti-tumor effects. However, whether the blockade of HER2+ exosomes release would affect antibody-mediated tumor inhibition has yet to be investigated. METHODS Exosomes derived from BT-474, SK-BR3 and SK-OV3 (HER2-overexpressing tumor cells) and MDA-MB-231 cells (HER2 negative) were purified and characterized by bicinchoninic acid (BCA) assay, western blotting and Transmission electron microscopy (TEM). Inhibition of exosome release was achieved by neutral sphingomyelinase-2 (nSMase-2) inhibitor, GW4869. The effects of exosome blockade on the anti-proliferative effects, apoptosis induction, and antibody-mediated cellular cytotoxicity (ADCC) activity of Trastuzumab were examined using MTT, flow cytometry, and LDH release assays. Also, the effects of exosome inhibition on the surface expression and endocytosis/internalization of HER2 were studied by flow cytometry. RESULTS Purified exosomes derived from HER2 overexpressing cancer cells were positive for HER2 protein. Blockade of exosome release was able to significantly improve apoptosis induction, anti-proliferative and ADCC responses of Trastuzumab dose dependently. The pretreatment of Trastuzumab/purified NK cells, but not PBMCs, with HER2+ exosomes could also decrease the ADCC effects of Trastuzumab. Exosome inhibition also remarkably downregulated surface HER2 levels in a time-dependent manner, but does not affect its endocytosis/internalization. CONCLUSION Based on our findings, HER2+ exosomes may benefit tumor progression by dually suppressing Trastuzumab-induced tumor growth inhibition and cytotoxicity of NK cells. It seems that concomitant blocking of exosome release might be an effective approach for improving the therapeutic effects of Trastuzumab, and potentially other HER2-directed mAbs. In addition, the exosome secretion pathway possibly contributes to the HER2 trafficking to plasma membrane, since the blockade of exosome secretion decreased surface HER2 levels.
Collapse
Affiliation(s)
- Reza Hosseini
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Leila Asef-Kabiri
- Cancer Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hamzeh Sarvnaz
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Alireza Ghanavatinejad
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Rezayat
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Nahid Eskandari
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.
| | | |
Collapse
|
41
|
Odell LR, Robertson MJ, Young KA, McGeachie AB, Quan A, Robinson PJ, McCluskey A. Prodrugs of the Archetypal Dynamin Inhibitor Bis-T-22. ChemMedChem 2022; 17:e202200400. [PMID: 36351775 PMCID: PMC10947042 DOI: 10.1002/cmdc.202200400] [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: 07/21/2022] [Revised: 10/06/2022] [Indexed: 11/11/2022]
Abstract
The Bis-T series of compounds comprise some of the most potent inhibitors of dynamin GTPase activity yet reported, e. g., (2E,2'E)-N,N'-(propane-1,3-diyl)bis(2-cyano-3-(3,4-dihydroxyphenyl)acrylamide) (2), Bis-T-22. The catechol moieties are believed to limit cell permeability, rendering these compounds largely inactive in cells. To solve this problem, a prodrug strategy was envisaged and eight ester analogues were synthesised. The shortest and bulkiest esters (acetate and butyl/tert-butyl) were found to be insoluble under physiological conditions, whilst the remaining five were soluble and stable under these conditions. These five were analysed for plasma stability and half-lives ranged from ∼2.3 min (propionic ester 4), increasing with size and bulk, to greater than 24 hr (dimethyl carbamate 10). Similar profiles where observed with the rate of formation of Bis-T-22 with half-lives ranging from ∼25 mins (propionic ester 4). Propionic ester 4 was chosen to undergo further testing and was found to inhibit endocytosis in a dose-dependent manner with IC50 ∼8 μM, suggesting this compound is able to effectively cross the cell membrane where it is rapidly hydrolysed to the desired Bis-T-22 parent compound.
Collapse
Affiliation(s)
- Luke R. Odell
- The University of NewcastleUniversity DriveCallaghanNSW 2308Australia
- Present address: Department of Medicinal ChemistryUppsala UniversityBox 57475123UppsalaSweden
| | - Mark J Robertson
- The University of NewcastleUniversity DriveCallaghanNSW 2308Australia
- Present address: Chemistry, College of Science & EngineeringJames Cook UniversityTownsvilleQLD 4814Australia
| | - Kelly A Young
- The University of NewcastleUniversity DriveCallaghanNSW 2308Australia
| | - Andrew B. McGeachie
- Cell Signalling UnitChildren's Medical Research InstituteThe University of Sydney214 Hawkesbury RoadWestmeadNSW 2145Australia
| | - Annie Quan
- Cell Signalling UnitChildren's Medical Research InstituteThe University of Sydney214 Hawkesbury RoadWestmeadNSW 2145Australia
| | - Phillip J. Robinson
- Cell Signalling UnitChildren's Medical Research InstituteThe University of Sydney214 Hawkesbury RoadWestmeadNSW 2145Australia
| | - Adam McCluskey
- The University of NewcastleUniversity DriveCallaghanNSW 2308Australia
| |
Collapse
|
42
|
Furuya G, Katoh H, Atsumi S, Hashimoto I, Komura D, Hatanaka R, Senga S, Hayashi S, Akita S, Matsumura H, Miura A, Mita H, Nakakido M, Nagatoishi S, Sugiyama A, Suzuki R, Konishi H, Yamamoto A, Abe H, Hiraoka N, Aoki K, Kato Y, Seto Y, Yoshimura C, Miyadera K, Tsumoto K, Ushiku T, Ishikawa S. Nucleic acid-triggered tumoral immunity propagates pH-selective therapeutic antibodies through tumor-driven epitope spreading. Cancer Sci 2022; 114:321-338. [PMID: 36136061 PMCID: PMC9807517 DOI: 10.1111/cas.15596] [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: 08/11/2022] [Revised: 09/09/2022] [Accepted: 09/12/2022] [Indexed: 01/07/2023] Open
Abstract
Important roles of humoral tumor immunity are often pointed out; however, precise profiles of dominant antigens and developmental mechanisms remain elusive. We systematically investigated the humoral antigens of dominant intratumor immunoglobulin clones found in human cancers. We found that approximately half of the corresponding antigens were restricted to strongly and densely negatively charged polymers, resulting in simultaneous reactivities of the antibodies to both densely sulfated glycosaminoglycans (dsGAGs) and nucleic acids (NAs). These anti-dsGAG/NA antibodies matured and expanded via intratumoral immunological driving force of innate immunity via NAs. These human cancer-derived antibodies exhibited acidic pH-selective affinity across both antigens and showed specific reactivity to diverse spectrums of human tumor cells. The antibody-drug conjugate exerted therapeutic effects against multiple cancers in vivo by targeting cell surface dsGAG antigens. This study reveals that intratumoral immunological reactions propagate tumor-oriented immunoglobulin clones and demonstrates a new therapeutic modality for the universal treatment of human malignancies.
Collapse
Affiliation(s)
- Genta Furuya
- Department of Preventive medicine, Graduate School of MedicineThe University of TokyoTokyoJapan
| | - Hiroto Katoh
- Department of Preventive medicine, Graduate School of MedicineThe University of TokyoTokyoJapan
| | - Shinichiro Atsumi
- Department of Preventive medicine, Graduate School of MedicineThe University of TokyoTokyoJapan
| | - Itaru Hashimoto
- Department of Preventive medicine, Graduate School of MedicineThe University of TokyoTokyoJapan
| | - Daisuke Komura
- Department of Preventive medicine, Graduate School of MedicineThe University of TokyoTokyoJapan
| | - Ryo Hatanaka
- Discovery and Preclinical Research DivisionTaiho Pharmaceutical Co., Ltd.IbarakiJapan
| | - Shogo Senga
- Discovery and Preclinical Research DivisionTaiho Pharmaceutical Co., Ltd.IbarakiJapan
| | - Shuto Hayashi
- Department of Preventive medicine, Graduate School of MedicineThe University of TokyoTokyoJapan
| | - Shoji Akita
- Discovery and Preclinical Research DivisionTaiho Pharmaceutical Co., Ltd.IbarakiJapan
| | - Hirofumi Matsumura
- Discovery and Preclinical Research DivisionTaiho Pharmaceutical Co., Ltd.IbarakiJapan
| | - Akihiro Miura
- Discovery and Preclinical Research DivisionTaiho Pharmaceutical Co., Ltd.IbarakiJapan
| | - Hideaki Mita
- Department of Preventive medicine, Graduate School of MedicineThe University of TokyoTokyoJapan
| | - Makoto Nakakido
- Laboratory of Medical Proteomics, Institute of Medical ScienceThe University of TokyoTokyoJapan
| | - Satoru Nagatoishi
- Laboratory of Medical Proteomics, Institute of Medical ScienceThe University of TokyoTokyoJapan
| | - Akira Sugiyama
- Laboratory of Systems Biology and MedicineResearch Center for Advanced Science and Technology, The University of TokyoTokyoJapan
| | - Ryohei Suzuki
- Department of Preventive medicine, Graduate School of MedicineThe University of TokyoTokyoJapan
| | - Hiroki Konishi
- Department of Preventive medicine, Graduate School of MedicineThe University of TokyoTokyoJapan
| | - Asami Yamamoto
- Department of Preventive medicine, Graduate School of MedicineThe University of TokyoTokyoJapan
| | - Hiroyuki Abe
- Department of Pathology, Graduate School of MedicineThe University of TokyoTokyoJapan
| | - Nobuyoshi Hiraoka
- Department of Analytical PathologyNational Cancer Center Research InstituteTokyoJapan
| | - Kazunori Aoki
- Division of Molecular and Cellular MedicineNational Cancer Center Research InstituteTokyoJapan
| | - Yasumasa Kato
- Department of Oral Function and Molecular BiologyOhu University School of DentistryFukushimaJapan
| | - Yasuyuki Seto
- Department of Gastrointestinal SurgeryGraduate School of Medicine, The University of TokyoTokyoJapan
| | - Chihoko Yoshimura
- Discovery and Preclinical Research DivisionTaiho Pharmaceutical Co., Ltd.IbarakiJapan
| | - Kazutaka Miyadera
- Discovery and Preclinical Research DivisionTaiho Pharmaceutical Co., Ltd.IbarakiJapan
| | - Kouhei Tsumoto
- Laboratory of Medical Proteomics, Institute of Medical ScienceThe University of TokyoTokyoJapan
| | - Tetsuo Ushiku
- Department of Pathology, Graduate School of MedicineThe University of TokyoTokyoJapan
| | - Shumpei Ishikawa
- Department of Preventive medicine, Graduate School of MedicineThe University of TokyoTokyoJapan
| |
Collapse
|
43
|
O’Donnell JS, Isaacs A, Jakob V, Lebas C, Barnes JB, Reading PC, Young PR, Watterson D, Dubois PM, Collin N, Chappell KJ. Characterization and comparison of novel adjuvants for a prefusion clamped MERS vaccine. Front Immunol 2022; 13:976968. [PMID: 36119058 PMCID: PMC9478912 DOI: 10.3389/fimmu.2022.976968] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 07/26/2022] [Indexed: 11/13/2022] Open
Abstract
Various chemical adjuvants are available to augment immune responses to non-replicative, subunit vaccines. Optimized adjuvant selection can ensure that vaccine-induced immune responses protect against the diversity of pathogen-associated infection routes, mechanisms of infectious spread, and pathways of immune evasion. In this study, we compare the immune response of mice to a subunit vaccine of Middle Eastern respiratory syndrome coronavirus (MERS-CoV) spike protein, stabilized in its prefusion conformation by a proprietary molecular clamp (MERS SClamp) alone or formulated with one of six adjuvants: either (i) aluminium hydroxide, (ii) SWE, a squalene-in-water emulsion, (iii) SQ, a squalene-in-water emulsion containing QS21 saponin, (iv) SMQ, a squalene-in-water emulsion containing QS21 and a synthetic toll-like receptor 4 (TLR4) agonist 3D-6-acyl Phosphorylated HexaAcyl Disaccharide (3D6AP); (v) LQ, neutral liposomes containing cholesterol, 1.2-dioleoyl-sn-glycero-3-phosphocholine (DOPC) and QS21, (vi) or LMQ, neutral liposomes containing cholesterol, DOPC, QS21, and 3D6AP. All adjuvanted formulations induced elevated antibody titers which where greatest for QS21-containing formulations. These had elevated neutralization capacity and induced higher frequencies of IFNƔ and IL-2-producing CD4+ and CD8+ T cells. Additionally, LMQ-containing formulations skewed the antibody response towards IgG2b/c isotypes, allowing for antibody-dependent cellular cytotoxicity. This study highlights the utility of side-by-side adjuvant comparisons in vaccine development.
Collapse
Affiliation(s)
- Jake S. O’Donnell
- The Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, Australia
- The School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia
| | - Ariel Isaacs
- The Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, Australia
- The School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia
| | | | - Celia Lebas
- Vaccine Formulation Institute, Geneva, Switzerland
| | - James B. Barnes
- The WHO Collaborating Centre for Reference and Research on Influenza, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Patrick C. Reading
- The WHO Collaborating Centre for Reference and Research on Influenza, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
- Department of Microbiology and Immunology, The University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Paul R. Young
- The Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, Australia
- The School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia
- Australian Infectious Disease Research Centre, The University of Queensland, Brisbane, QLD, Australia
| | - Daniel Watterson
- The Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, Australia
- The School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia
- Australian Infectious Disease Research Centre, The University of Queensland, Brisbane, QLD, Australia
| | | | - Nicolas Collin
- Vaccine Formulation Institute, Geneva, Switzerland
- *Correspondence: Keith J. Chappell, ; Nicolas Collin,
| | - Keith J. Chappell
- The Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, Australia
- The School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia
- Australian Infectious Disease Research Centre, The University of Queensland, Brisbane, QLD, Australia
- *Correspondence: Keith J. Chappell, ; Nicolas Collin,
| |
Collapse
|
44
|
Zhai Y, Dong S, Li H, Zhang Y, Shami P, Chen M. Antibody-mediated depletion of programmed death 1-positive (PD-1 +) cells. J Control Release 2022; 349:425-433. [PMID: 35820540 PMCID: PMC10699550 DOI: 10.1016/j.jconrel.2022.07.010] [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: 03/08/2022] [Revised: 06/17/2022] [Accepted: 07/06/2022] [Indexed: 10/17/2022]
Abstract
PD-1 immune checkpoint has been intensively investigated in pathogenesis and treatments for cancer and autoimmune diseases. Cells that express PD-1 (PD-1+ cells) draw ever-increasing attention in cancer and autoimmune disease research although the role of PD-1+ cells in the progression and treatments of these diseases remains largely ambiguous. One definite approach to elucidate their roles is to deplete these cells in disease settings and examine how the depletion impacts disease progression and treatments. To execute the depletion, we designed and generated the first depleting antibody (D-αPD-1) that specifically ablates PD-1+ cells. D-αPD-1 has the same variable domains as an anti-mouse PD-1 blocking antibody (RMP1-14). The constant domains of D-αPD-1 were derived from mouse IgG2a heavy and κ-light chain, respectively. D-αPD-1 was verified to bind with mouse PD-1 as well as mouse FcγRIV, an immuno-activating Fc receptor. The cell depletion effect of D-αPD-1 was confirmed in vivo using a PD-1+ cell transferring model. Since transferred PD-1+ cells, EL4 cells, are tumorigenic and EL4 tumors are lethal to host mice, the depleting effect of D-αPD-1 was also manifested by an absolute survival among the antibody-treated mice while groups receiving control treatments had median survival time of merely approximately 30 days. Furthermore, we found that D-αPD-1 leads to elimination of PD-1+ cells through antibody-dependent cell-mediate phagocytosis (ADCP) and complement-dependent cytotoxicity (CDC) mechanisms. These results, altogether, confirmed the specificity and effectiveness of D-αPD-1. The results also highlighted that D-αPD-1 is a robust tool to study PD-1+ cells in cancer and autoimmune diseases and a potential therapeutic for these diseases.
Collapse
Affiliation(s)
- Yujia Zhai
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT 84112, USA
| | - Shuyun Dong
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT 84112, USA
| | - Haojia Li
- Division of Epidemiology, Department of Internal Medicine, University of Utah, Salt Lake City, UT 84132, USA
| | - Yue Zhang
- Division of Epidemiology, Department of Internal Medicine, University of Utah, Salt Lake City, UT 84132, USA
| | - Paul Shami
- Division of Hematology and Hematologic Malignancies, Department of Internal Medicine, and Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Mingnan Chen
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT 84112, USA.
| |
Collapse
|
45
|
Cong VT, Houng JL, Kavallaris M, Chen X, Tilley RD, Gooding JJ. How can we use the endocytosis pathways to design nanoparticle drug-delivery vehicles to target cancer cells over healthy cells? Chem Soc Rev 2022; 51:7531-7559. [PMID: 35938511 DOI: 10.1039/d1cs00707f] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Targeted drug delivery in cancer typically focuses on maximising the endocytosis of drugs into the diseased cells. However, there has been less focus on exploiting the differences in the endocytosis pathways of cancer cells versus non-cancer cells. An understanding of the endocytosis pathways in both cancer and non-cancer cells allows for the design of nanoparticles to deliver drugs to cancer cells whilst restricting healthy cells from taking up anticancer drugs, thus efficiently killing the cancer cells. Herein we compare the differences in the endocytosis pathways of cancer and healthy cells. Second, we highlight the importance of the physicochemical properties of nanoparticles (size, shape, stiffness, and surface chemistry) on cellular uptake and how they can be adjusted to selectively target the dominated endocytosis pathway of cancer cells over healthy cells and to deliver anticancer drug to the target cells. The review generates new thought in the design of cancer-selective nanoparticles based on the endocytosis pathways.
Collapse
Affiliation(s)
- Vu Thanh Cong
- School of Chemistry, University of New South Wales, Sydney, NSW 2052, Australia. .,Australian Centre for NanoMedicine, University of New South Wales, Sydney, NSW 2052, Australia
| | - Jacinta L Houng
- School of Chemistry, University of New South Wales, Sydney, NSW 2052, Australia. .,Australian Centre for NanoMedicine, University of New South Wales, Sydney, NSW 2052, Australia
| | - Maria Kavallaris
- Australian Centre for NanoMedicine, University of New South Wales, Sydney, NSW 2052, Australia.,Children's Cancer Institute, Lowy Cancer Research Centre, University of New South Wales, Sydney, NSW 2052, Australia.,School of Clinical Medicine, UNSW Medicine & Health, University of New South Wales, Sydney, NSW 2052, Australia
| | - Xin Chen
- School of Chemical Engineering and Technology, Shaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical Engineering, Xi'an Jiao Tong University, Xi'an, China
| | - Richard D Tilley
- School of Chemistry, University of New South Wales, Sydney, NSW 2052, Australia.
| | - J Justin Gooding
- School of Chemistry, University of New South Wales, Sydney, NSW 2052, Australia. .,Australian Centre for NanoMedicine, University of New South Wales, Sydney, NSW 2052, Australia
| |
Collapse
|
46
|
Harnessing natural killer cells for cancer immunotherapy: dispatching the first responders. Nat Rev Drug Discov 2022; 21:559-577. [PMID: 35314852 PMCID: PMC10019065 DOI: 10.1038/s41573-022-00413-7] [Citation(s) in RCA: 81] [Impact Index Per Article: 40.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/24/2022] [Indexed: 02/07/2023]
Abstract
Natural killer (NK) cells have crucial roles in the innate immunosurveillance of cancer and viral infections. They are 'first responders' that can spontaneously recognize abnormal cells in the body, rapidly eliminate them through focused cytotoxicity mechanisms and potently produce pro-inflammatory cytokines and chemokines that recruit and activate other immune cells to initiate an adaptive response. From the initial discovery of the diverse cell surface receptors on NK cells to the characterization of regulatory events that control their function, our understanding of the basic biology of NK cells has improved dramatically in the past three decades. This advanced knowledge has revealed increased mechanistic complexity, which has opened the doors to the development of a plethora of exciting new therapeutics that can effectively manipulate and target NK cell functional responses, particularly in cancer patients. Here, we summarize the basic mechanisms that regulate NK cell biology, review a wide variety of drugs, cytokines and antibodies currently being developed and used to stimulate NK cell responses, and outline evolving NK cell adoptive transfer approaches to treat cancer.
Collapse
|
47
|
Liu H, Wu W, Sun G, Chia T, Cao L, Liu X, Guan J, Fu F, Yao Y, Wu Z, Zhou S, Wang J, Lu J, Kuang Z, Wu M, He L, Shao Z, Wu D, Chen B, Xu W, Wang Z, He K. Optimal target saturation of ligand-blocking anti-GITR antibody IBI37G5 dictates FcγR-independent GITR agonism and antitumor activity. Cell Rep Med 2022; 3:100660. [PMID: 35732156 PMCID: PMC9245059 DOI: 10.1016/j.xcrm.2022.100660] [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: 12/08/2021] [Revised: 03/26/2022] [Accepted: 05/20/2022] [Indexed: 11/29/2022]
Abstract
Glucocorticoid-induced tumor necrosis factor receptor (GITR) is a co-stimulatory receptor and an important target for cancer immunotherapy. We herein present a potent FcγR-independent GITR agonist IBI37G5 that can effectively activate effector T cells and synergize with anti-programmed death 1 (PD1) antibody to eradicate established tumors. IBI37G5 depends on both antibody bivalency and GITR homo-dimerization for efficient receptor cross-linking. Functional analyses reveal bell-shaped dose responses due to the unique 2:2 antibody-receptor stoichiometry required for GITR activation. Antibody self-competition is observed after concentration exceeded that of 100% receptor occupancy (RO), which leads to antibody monovalent binding and loss of activity. Retrospective pharmacokinetics/pharmacodynamics analysis demonstrates that the maximal efficacy is achieved at medium doses with drug exposure near saturating GITR occupancy during the dosing cycle. Finally, we propose an alternative dose-finding strategy that does not rely on the traditional maximal tolerated dose (MTD)-based paradigm but instead on utilizing the RO-function relations as biomarker to guide the clinical translation of GITR and similar co-stimulatory agonists.
Collapse
Affiliation(s)
- Huisi Liu
- Department of Immunology, Innovent Guoqing Academy, Innovent Biologics (Suzhou) Co., Ltd., Suzhou, China
| | - Weiwei Wu
- Department of Pharmacology and Preclinical Studies, Innovent Guoqing Academy, Innovent Biologics (Suzhou) Co., Ltd., Suzhou, China
| | - Gangyu Sun
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Tiongsun Chia
- Department of Immunology, Innovent Guoqing Academy, Innovent Biologics (Suzhou) Co., Ltd., Suzhou, China
| | - Lei Cao
- Department of Pharmacology and Preclinical Studies, Innovent Guoqing Academy, Innovent Biologics (Suzhou) Co., Ltd., Suzhou, China
| | - Xiaodan Liu
- Department of Immunology, Innovent Guoqing Academy, Innovent Biologics (Suzhou) Co., Ltd., Suzhou, China
| | - Jian Guan
- Department of Immunology, Innovent Guoqing Academy, Innovent Biologics (Suzhou) Co., Ltd., Suzhou, China
| | - Fenggen Fu
- Department of Antibody Discovery and Protein Engineering, Guoqing Academy, Innovent Biologics (Suzhou) Co., Ltd., Suzhou, China
| | - Ying Yao
- Department of Pharmacology and Preclinical Studies, Innovent Guoqing Academy, Innovent Biologics (Suzhou) Co., Ltd., Suzhou, China
| | - Zhihai Wu
- Department of Antibody Discovery and Protein Engineering, Guoqing Academy, Innovent Biologics (Suzhou) Co., Ltd., Suzhou, China
| | - Shuaixiang Zhou
- Department of Antibody Discovery and Protein Engineering, Guoqing Academy, Innovent Biologics (Suzhou) Co., Ltd., Suzhou, China
| | - Jie Wang
- Department of Pharmacology and Preclinical Studies, Innovent Guoqing Academy, Innovent Biologics (Suzhou) Co., Ltd., Suzhou, China
| | - Jia Lu
- Department of Pharmacology and Preclinical Studies, Innovent Guoqing Academy, Innovent Biologics (Suzhou) Co., Ltd., Suzhou, China
| | - Zhihui Kuang
- Department of Pharmacology and Preclinical Studies, Innovent Guoqing Academy, Innovent Biologics (Suzhou) Co., Ltd., Suzhou, China
| | - Min Wu
- Department of Pharmacology and Preclinical Studies, Innovent Guoqing Academy, Innovent Biologics (Suzhou) Co., Ltd., Suzhou, China
| | - Luan He
- Department of Immunology, Innovent Guoqing Academy, Innovent Biologics (Suzhou) Co., Ltd., Suzhou, China
| | - Zhiyuan Shao
- Department of Antibody Discovery and Protein Engineering, Guoqing Academy, Innovent Biologics (Suzhou) Co., Ltd., Suzhou, China
| | - Dongdong Wu
- Department of Pharmacology and Preclinical Studies, Innovent Guoqing Academy, Innovent Biologics (Suzhou) Co., Ltd., Suzhou, China
| | - Bingliang Chen
- Department of Pharmacology and Preclinical Studies, Innovent Guoqing Academy, Innovent Biologics (Suzhou) Co., Ltd., Suzhou, China
| | - Wenqing Xu
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Zhizhi Wang
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China.
| | - Kaijie He
- Department of Immunology, Innovent Guoqing Academy, Innovent Biologics (Suzhou) Co., Ltd., Suzhou, China.
| |
Collapse
|
48
|
Kuo YC, Kuo CF, Jenkins K, Hung AFH, Chang WC, Park M, Aguilar B, Starr R, Hibbard J, Brown C, Williams JC. Antibody-based redirection of universal Fabrack-CAR T cells selectively kill antigen bearing tumor cells. J Immunother Cancer 2022; 10:jitc-2021-003752. [PMID: 35728874 PMCID: PMC9214433 DOI: 10.1136/jitc-2021-003752] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/23/2022] [Indexed: 11/07/2022] Open
Abstract
Background Chimeric antigen receptor (CAR) T cells engineered to recognize and target tumor associated antigens have made a profound impact on the quality of life for many patients with cancer. However, tumor heterogeneity and intratumoral immune suppression reduce the efficacy of this approach, allowing for tumor cells devoid of the target antigen to seed disease recurrence. Here, we address the complexity of tumor heterogeneity by developing a universal CAR. Method We constructed a universal Fabrack-CAR with an extracellular domain composed of the non-tumor targeted, cyclic, twelve residue meditope peptide that binds specifically to an engineered binding pocket within the Fab arm of monoclonal antibodies (mAbs). As this site is readily grafted onto therapeutic mAbs, the antigen specificity of these universal Fabrack-CAR T cells is simply conferred by administering mAbs with specificity to the heterogeneous tumor. Results Using in vitro and in vivo studies with multiple meditope-engineered mAbs, we show the feasibility, specificity, and robustness of this approach. These studies demonstrate antigen- and antibody-specific T cell activation, proliferation, and IFNγ production, selective killing of target cells in a mixed population, and tumor regression in animal models. Conclusion Collectively, these findings support the feasibility of this universal Fabrack-CAR T cell approach and provide the rationale for future clinical use in cancer immunotherapy.
Collapse
Affiliation(s)
- Yi-Chiu Kuo
- Department of Molecular Medicine, City of Hope National Medical Center, Duarte, California, USA
| | - Cheng-Fu Kuo
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, California, USA.,Irell and Manella Graduate School of Biological Sciences, City of Hope National Medical Center, Duarte, California, USA
| | - Kurt Jenkins
- Department of Molecular Medicine, City of Hope National Medical Center, Duarte, California, USA.,Irell and Manella Graduate School of Biological Sciences, City of Hope National Medical Center, Duarte, California, USA
| | - Alfur Fu-Hsin Hung
- Department of Molecular Medicine, City of Hope National Medical Center, Duarte, California, USA
| | - Wen-Chung Chang
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, California, USA
| | - Miso Park
- Department of Molecular Medicine, City of Hope National Medical Center, Duarte, California, USA
| | - Brenda Aguilar
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, California, USA
| | - Renate Starr
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, California, USA
| | - Jonathan Hibbard
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, California, USA
| | - Christine Brown
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, California, USA
| | - John C Williams
- Department of Molecular Medicine, City of Hope National Medical Center, Duarte, California, USA
| |
Collapse
|
49
|
Zou Y, Zheng S, Xie X, Ye F, Hu X, Tian Z, Yan SM, Yang L, Kong Y, Tang Y, Tian W, Xie J, Deng X, Zeng Y, Chen ZS, Tang H, Xie X. N6-methyladenosine regulated FGFR4 attenuates ferroptotic cell death in recalcitrant HER2-positive breast cancer. Nat Commun 2022; 13:2672. [PMID: 35562334 PMCID: PMC9106694 DOI: 10.1038/s41467-022-30217-7] [Citation(s) in RCA: 98] [Impact Index Per Article: 49.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 04/21/2022] [Indexed: 12/26/2022] Open
Abstract
Intrinsic and acquired anti-HER2 resistance remains a major hurdle for treating HER2-positive breast cancer. Using genome-wide CRISPR/Cas9 screening in vitro and in vivo, we identify FGFR4 as an essential gene following anti-HER2 treatment. FGFR4 inhibition enhances susceptibility to anti-HER2 therapy in resistant breast cancer. Mechanistically, m6A-hypomethylation regulated FGFR4 phosphorylates GSK-3β and activates β-catenin/TCF4 signaling to drive anti-HER2 resistance. Notably, suppression of FGFR4 dramatically diminishes glutathione synthesis and Fe2+ efflux efficiency via the β-catenin/TCF4-SLC7A11/FPN1 axis, resulting in excessive ROS production and labile iron pool accumulation. Ferroptosis, a unique iron-dependent form of oxidative cell death, is triggered after FGFR4 inhibition. Experiments involving patient-derived xenografts and organoids reveals a synergistic effect of anti-FGFR4 with anti-HER2 therapy in breast cancer with either intrinsic or acquired resistance. Together, these results pinpoint a mechanism of anti-HER2 resistance and provide a strategy for overcoming resistance via FGFR4 inhibition in recalcitrant HER2-positive breast cancer. Anti-HER2 resistance causes treatment failure in HER2-positive breast cancers. Here the authors identify FGFR4 as one of the vulnerabilities of anti-HER2 resistant breast cancer and show that FGRR4 inhibition enhances sensitivity to anti-HER2 treatment in these resistant cells by triggering ferroptosis.
Collapse
Affiliation(s)
- Yutian Zou
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Shaoquan Zheng
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Xinhua Xie
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Feng Ye
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Xiaoqian Hu
- School of Biomedical Sciences, Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Zhi Tian
- College of Pharmacy, University of South Florida, Tampa, FL, USA
| | - Shu-Mei Yan
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Lu Yang
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Yanan Kong
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Yuhui Tang
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Wenwen Tian
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Jindong Xie
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Xinpei Deng
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Yan Zeng
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Zhe-Sheng Chen
- College of Pharmacy and Health Sciences, St. John's University, Queens, NY, USA.
| | - Hailin Tang
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.
| | - Xiaoming Xie
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.
| |
Collapse
|
50
|
Caveolin-1 temporal modulation enhances antibody drug efficacy in heterogeneous gastric cancer. Nat Commun 2022; 13:2526. [PMID: 35534471 PMCID: PMC9085816 DOI: 10.1038/s41467-022-30142-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 04/19/2022] [Indexed: 11/11/2022] Open
Abstract
Resistance mechanisms and heterogeneity in HER2-positive gastric cancers (GC) limit Trastuzumab benefit in 32% of patients, and other targeted therapies have failed in clinical trials. Using patient samples, patient-derived xenografts (PDXs), partially humanized biological models, and HER2-targeted imaging technologies we demonstrate the role of caveolin-1 (CAV1) as a complementary biomarker in GC selection for Trastuzumab therapy. In retrospective analyses of samples from patients enrolled on Trastuzumab trials, the CAV1-high profile associates with low membrane HER2 density and low patient survival. We show a negative correlation between CAV1 tumoral protein levels – a major protein of cholesterol-rich membrane domains – and Trastuzumab-drug conjugate TDM1 tumor uptake. Finally, CAV1 depletion using knockdown or pharmacologic approaches (statins) increases antibody drug efficacy in tumors with incomplete HER2 membranous reactivity. In support of these findings, background statin use in patients associates with enhanced antibody efficacy. Together, this work provides preclinical justification and clinical evidence that require prospective investigation of antibody drugs combined with statins to delay drug resistance in tumors. Clinical evidences have demonstrated limited efficacy of HER2-targeted therapies in patients with gastric cancer (GC). Here the authors show that survival benefit to anti-HER2 antibody Trastuzumab is reduced in GC patients with high levels of the caveolin-1 and that, in preclinical cancer models, antibody drug efficacy can be improved by modulating caveolin-1 levels with cholesterol-depleting drugs, statins.
Collapse
|