1
|
Xie W, Kong Y, Ren C, Wen Y, Ying M, Xing H. Chemistries on the inner leaflet of the cell membrane. Chem Commun (Camb) 2025. [PMID: 39810742 DOI: 10.1039/d4cc05186f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
The cell membrane, characterized by its inherent asymmetry, functions as a dynamic barrier that regulates numerous cellular activities. This Highlight aims to provide the chemistry community with a comprehensive overview of the intriguing and underexplored inner leaflet, encompassing both fundamental biology and emerging synthetic modification strategies. We begin by describing the asymmetric nature of the plasma membrane, with a focus on the distinct roles of lipids, proteins, and glycan chains, highlighting the composition and biofunctions of the inner leaflet and the biological mechanisms that sustain membrane asymmetry. Next, we explore chemical biological strategies for engineering the inner leaflet, including genetic engineering, transmembrane peptides, and liposome fusion-based transport. In the perspective section, we discuss the challenges in developing chemistries for the inner leaflet of the cell membrane, aiming to inspire researchers and collaborators to explore this field and address its unanswered biological questions.
Collapse
Affiliation(s)
- Wenxue Xie
- Institute of Chemical Biology and Nanomedicine, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, China.
| | - Yuhan Kong
- Institute of Chemical Biology and Nanomedicine, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, China.
| | - Cong Ren
- Institute of Chemical Biology and Nanomedicine, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, China.
| | - Yujian Wen
- Institute of Chemical Biology and Nanomedicine, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, China.
| | | | - Hang Xing
- Institute of Chemical Biology and Nanomedicine, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, China.
- Research Institute of Hunan University in Chongqing, Chongqing, 401100, China
| |
Collapse
|
2
|
He H, Deng X, Wang Z, Chen J. Recent progress in the development of peptide-drug conjugates (PDCs) for cancer therapy. Eur J Med Chem 2024; 284:117204. [PMID: 39731788 DOI: 10.1016/j.ejmech.2024.117204] [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: 11/04/2024] [Revised: 12/10/2024] [Accepted: 12/22/2024] [Indexed: 12/30/2024]
Abstract
Peptide-drug conjugates (PDCs) are emerging therapeutic agents composed of peptides, linkers, and payloads, which possess favorable targeting capability and can deliver enough payloads to the tumor sites with minimized impact on healthy tissues. However, only a few PDCs have been approved for clinical use so far. To advance the research on PDCs, this review summarizes the approved PDCs, and PDCs in clinical and preclinical stages based on the payload types. Additionally, the biological activity and pharmacokinetic properties of preclinical PDCs are detailedly described. Lastly, the challenges and future development directions of PDCs are discussed. This review aims to inspire insights into the development of PDCs for cancer treatment.
Collapse
Affiliation(s)
- Haiqi He
- Guangdong Provincial Key Laboratory of New Drug Screening, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Xin Deng
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinic Al Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Zhijie Wang
- Guangdong Provincial Key Laboratory of New Drug Screening, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China; Shenzhen Key Laboratory of Viral Oncology, Ministry of Science and Innovation, Shenzhen Hospital, Southern Medical University, Shenzhen, 518100, China
| | - Jianjun Chen
- Guangdong Provincial Key Laboratory of New Drug Screening, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
3
|
Ankrom E, Dalesandro B, Pires MM, Thévenin D. Selective Recruitment of Antibodies to Cancer Cells and Immune Cell-mediated Killing via In Situ Click Chemistry. ChemMedChem 2024; 19:e202400356. [PMID: 39087480 PMCID: PMC11617666 DOI: 10.1002/cmdc.202400356] [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: 07/15/2023] [Revised: 07/14/2024] [Accepted: 01/17/2024] [Indexed: 08/02/2024]
Abstract
Many current cancer immunotherapies function by redirecting immune system components to recognize cancer biomarkers and initiate a cytotoxic attack. The lack of a universal tumor biomarker limits the therapeutic potential of these approaches. However, one feature characteristic of nearly all solid tumors is extracellular acidity. This inherent acidity provides the basis for targeted drug delivery via the pH-low insertion peptide (pHLIP), which selectively accumulates in tumors in vivo due to a pH-dependent membrane insertion propensity. Previously, we established that we could selectively decorate cancer cells with antigen-pHLIP conjugates to facilitate antibody recruitment and subsequent killing by engineered effector cells via antibody-dependent cellular cytotoxicity (ADCC). Here, we present a novel strategy for opsonizing antibodies on target cell surfaces using click chemistry. We utilize pHLIP to facilitate selective tetrazine - trans-cyclooctene ligation of human IgGs to the cancer cell surface and induce ADCC. We demonstrate that our approach activates the primary ADCC signaling pathway via CD16a (FcγRIIIa) receptors on effector cells and induces the killing of cancer cell targets by engineered NK cells.
Collapse
Affiliation(s)
- Emily Ankrom
- Department of ChemistryLehigh UniversityBethlehem, Pennsylvania18015USA
| | - Brianna Dalesandro
- Department of ChemistryUniversity of VirginiaCharlottesville, Virginia22904USA
| | - Marcos M. Pires
- Department of ChemistryUniversity of VirginiaCharlottesville, Virginia22904USA
| | - Damien Thévenin
- Department of ChemistryLehigh UniversityBethlehem, Pennsylvania18015USA
| |
Collapse
|
4
|
Knepper LE, Ankrom ET, Thévenin D. Enhancing Anti-Cancer Immune Response by Acidosis-Sensitive Nanobody Display. J Membr Biol 2024; 257:391-401. [PMID: 39254684 PMCID: PMC11584308 DOI: 10.1007/s00232-024-00322-3] [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: 07/16/2024] [Accepted: 08/16/2024] [Indexed: 09/11/2024]
Abstract
One of the main challenges with many cancer immunotherapies is that biomarkers are needed for targeting. These biomarkers are often associated with tumors but are not specific to a particular tumor and can lead to damage in healthy tissues, resistance to treatment, or the need for customization for different types of cancer due to variations in targets. A promising alternative approach is to target the acidic microenvironment found in most solid tumor types. This can be achieved using the pH (Low) Insertion Peptide (pHLIP), which inserts selectively into cell membranes under acidic conditions, sparing healthy tissues. pHLIP has shown potential for imaging, drug delivery, and surface display. For instance, we previously used pHLIP to display epitopes on the surfaces of cancer cells, enabling antibody-mediated immune cell recruitment and selective killing of cancer cells. In this study, we further explored this concept by directly fusing an anti-CD16 nanobody, which activates natural killer (NK) cells, to pHLIP, eliminating the need for antibody recruitment. Our results demonstrated the insertion of pH-sensitive agents into cancer cells, activation of the CD16 receptor on effector cells, and successful targeting and destruction of cancer cells by high-affinity CD16+ NK cells in two cancer cell lines.
Collapse
Affiliation(s)
- Leah E Knepper
- Department of Chemistry, Lehigh University, Bethlehem, PA, 18015, USA
| | - Emily T Ankrom
- Department of Chemistry, Lehigh University, Bethlehem, PA, 18015, USA
| | - Damien Thévenin
- Department of Chemistry, Lehigh University, Bethlehem, PA, 18015, USA.
| |
Collapse
|
5
|
Sun X, Setrerrahmane S, Li C, Hu J, Xu H. Nucleic acid drugs: recent progress and future perspectives. Signal Transduct Target Ther 2024; 9:316. [PMID: 39609384 PMCID: PMC11604671 DOI: 10.1038/s41392-024-02035-4] [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] [Received: 10/31/2023] [Revised: 09/20/2024] [Accepted: 10/25/2024] [Indexed: 11/30/2024] Open
Abstract
High efficacy, selectivity and cellular targeting of therapeutic agents has been an active area of investigation for decades. Currently, most clinically approved therapeutics are small molecules or protein/antibody biologics. Targeted action of small molecule drugs remains a challenge in medicine. In addition, many diseases are considered 'undruggable' using standard biomacromolecules. Many of these challenges however, can be addressed using nucleic therapeutics. Nucleic acid drugs (NADs) are a new generation of gene-editing modalities characterized by their high efficiency and rapid development, which have become an active research topic in new drug development field. However, many factors, including their low stability, short half-life, high immunogenicity, tissue targeting, cellular uptake, and endosomal escape, hamper the delivery and clinical application of NADs. Scientists have used chemical modification techniques to improve the physicochemical properties of NADs. In contrast, modified NADs typically require carriers to enter target cells and reach specific intracellular locations. Multiple delivery approaches have been developed to effectively improve intracellular delivery and the in vivo bioavailability of NADs. Several NADs have entered the clinical trial recently, and some have been approved for therapeutic use in different fields. This review summarizes NADs development and evolution and introduces NADs classifications and general delivery strategies, highlighting their success in clinical applications. Additionally, this review discusses the limitations and potential future applications of NADs as gene therapy candidates.
Collapse
Affiliation(s)
- Xiaoyi Sun
- Jiangsu Province Engineering Research Center of Synthetic Peptide Drug Discovery and Evaluation, China Pharmaceutical University, Nanjing, 210009, China
| | | | - Chencheng Li
- Jiangsu Province Engineering Research Center of Synthetic Peptide Drug Discovery and Evaluation, China Pharmaceutical University, Nanjing, 210009, China
| | - Jialiang Hu
- Jiangsu Province Engineering Research Center of Synthetic Peptide Drug Discovery and Evaluation, China Pharmaceutical University, Nanjing, 210009, China
| | - Hanmei Xu
- Jiangsu Province Engineering Research Center of Synthetic Peptide Drug Discovery and Evaluation, China Pharmaceutical University, Nanjing, 210009, China.
| |
Collapse
|
6
|
Wang C, Wang B, Zhang Q, Zhang S. Tumor microenvironment-responsive cell-penetrating peptides: Design principle and precision delivery. Colloids Surf B Biointerfaces 2024; 242:114100. [PMID: 39024717 DOI: 10.1016/j.colsurfb.2024.114100] [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: 05/08/2024] [Revised: 06/29/2024] [Accepted: 07/15/2024] [Indexed: 07/20/2024]
Abstract
Cell-penetrating peptides (CPPs) are promising vehicles for intracellular delivery of different cargoes. Although various CPPs are designed for targeted delivery of nanomedicines and anticancer drugs, their clinical approval is hampered by a lack of selectivity. In recent years, new approaches have been explored to address this drawback, and distinct strategies for tumor microenvironment (TME)-responsive activation have been developed. In this review, we first introduce the cellular uptake mechanisms of CPPs. We next extensively discuss the design principles and precision delivery of TME-responsive CPPs. Nine kinds of single stimulus-responsive CPPs, five kinds of multiple stimuli-responsive CPPs, three kinds of TME-responsive targeting CPPs, and two kinds of reversibly activatable CPPs (RACPPs) are systemically summarized. Then, TME-responsive CPPs for nanomedical applications are further discussed. Finally, we describe the translational applications of TME-responsive CPPs for anticancer drug delivery. These commentaries provide an insight into the design of next-generation activatable CPPs (ACPPs) for selective delivery of nanomedicines and anticancer drugs.
Collapse
Affiliation(s)
- Chenhui Wang
- Department of Cell Biology, School of Medicine, Nankai University, Tianjin 300071, China
| | - Bo Wang
- Department of Cell Biology, School of Medicine, Nankai University, Tianjin 300071, China
| | - Qing Zhang
- Department of Laboratory Medicine, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China
| | - Sihe Zhang
- Department of Cell Biology, School of Medicine, Nankai University, Tianjin 300071, China.
| |
Collapse
|
7
|
Liu X, Ullah I, Yuan Y. Tumor Acidity-Triggered Bioorthogonal Reactions for Biomedical Applications. Chembiochem 2024; 25:e202400452. [PMID: 38940000 DOI: 10.1002/cbic.202400452] [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: 05/28/2024] [Revised: 06/27/2024] [Accepted: 06/27/2024] [Indexed: 06/29/2024]
Abstract
Cancer is one of the most serious threats to human health. Over the past few years, researchers have incrementally uncovered the pivotal role of tumor acidity in tumor formation, development, and treatment. In addition, bioorthogonal reactions have been widely used in tumor diagnosis and therapy, owing to their advantageous characteristics, including small ligand size, biocompatibility, fast reaction kinetics, and high chemospecificity. Consequently, bioorthogonal reactions triggered by tumor acidity have become an emerging strategy in biomedical applications. On this basis, we first elucidate the concept and major strategies of tumor acidity-triggered bioorthogonal reactions. Additionally, we review the progress in biomedical applications, with a particular focus on their importance in disease diagnosis and treatment. Finally, clinical challenges and future trends are also outlooked.
Collapse
Affiliation(s)
- Xiajian Liu
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 511442, China
| | - Ihsan Ullah
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 511442, China
| | - Youyong Yuan
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 511442, China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, China
| |
Collapse
|
8
|
Shi T, Sun M, Tuerhong S, Li M, Wang J, Wang Y, Zheng Q, Zou L, Lu C, Sun Z, Zou Z, Shao J, Du J, Li R, Liu B, Meng F. Acidity-targeting transition-aided universal chimeric antigen receptor T-cell (ATT-CAR-T) therapy for the treatment of solid tumors. Biomaterials 2024; 309:122607. [PMID: 38759487 DOI: 10.1016/j.biomaterials.2024.122607] [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: 11/22/2023] [Revised: 04/29/2024] [Accepted: 05/07/2024] [Indexed: 05/19/2024]
Abstract
The use of CAR-T cells in treating solid tumors frequently faces significant challenges, mainly due to the heterogeneity of tumor antigens. This study assessed the efficacy of an acidity-targeting transition-aided universal chimeric antigen receptor T (ATT-CAR-T) cell strategy, which is facilitated by an acidity-targeted transition. Specifically, the EGFRvIII peptide was attached to the N-terminus of a pH-low insertion peptide. Triggered by the acidic conditions of the tumor microenvironment, this peptide alters its structure and selectively integrates into the membrane of solid tumor cells. The acidity-targeted transition component effectively relocated the EGFRvIII peptide across various tumor cell membranes; thus, allowing the direct destruction of these cells by EGFRvIII-specific CAR-T cells. This method was efficient even when endogenous antigens were absent. In vivo tests showed marked antigen modification within the acidic tumor microenvironment using this component. Integrating this component with CAR-T cell therapy showed high effectiveness in combating solid tumors. These results highlight the capability of ATT-CAR-T cell therapy to address the challenges presented by tumor heterogeneity and expand the utility of CAR-T cell therapy in the treatment of solid tumors.
Collapse
Affiliation(s)
- Tianyu Shi
- Department of Oncology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing 210008, China
| | - Mengna Sun
- Department of Oncology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing 210008, China
| | - Subiyinuer Tuerhong
- Department of Oncology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing 210008, China
| | - Mengru Li
- Department of Oncology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing 210008, China
| | - Jiayu Wang
- Department of Oncology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing 210008, China
| | - Yingxin Wang
- Department of Oncology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing 210008, China
| | - Qinghua Zheng
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital, The Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China
| | - Lu Zou
- Department of Oncology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing 210008, China
| | - Changchang Lu
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Zhichen Sun
- Department of Oncology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China
| | - Zhengyun Zou
- Department of Oncology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing 210008, China
| | - Jie Shao
- Department of Oncology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China
| | - Juan Du
- Department of Oncology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China
| | - Rutian Li
- Department of Oncology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China.
| | - Baorui Liu
- Department of Oncology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China.
| | - Fanyan Meng
- Department of Oncology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing 210008, China; Department of Laboratory Medicine, Nanjing Drum Tower Hospital, The Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China.
| |
Collapse
|
9
|
Knepper LE, Ankrom ET, Thévenin D. Enhancing Anti-Cancer Immune Response by Acidosis-sensitive Nanobody Display. RESEARCH SQUARE 2024:rs.3.rs-4750804. [PMID: 39184093 PMCID: PMC11343302 DOI: 10.21203/rs.3.rs-4750804/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
One of the main challenges with many cancer immuno-therapies is that they depend on biomarkers for targeting. These biomarkers are often associated with tumors but are not specific to a particular tumor, which can lead to damage in healthy tissues, resistance to treatment, and the need for customization for different types of cancer due to the variations in targets. A promising alternative approach is to target the acidic microenvironment found in most solid tumor types. This can be achieved using the pH (Low) Insertion Peptide (pHLIP), which inserts selectively into cell membranes in acidic conditions, sparing healthy tissues. pHLIP has shown potential for imaging, drug delivery, and surface display. For instance, we previously used pHLIP to display epitopes on the surfaces of cancer cells, enabling antibody-mediated immune cell recruitment and selective killing of cancer cells. In this study, we further this concept by directly fusing an anti-CD16 nanobody, which activates Natural Killer (NK) cells, to pHLIP, eliminating the need for antibody recruitment. Our results demonstrate pH-sensitive insertion into cancer cells, activation of the CD16 receptor on effector cells, and successful targeting and destruction of cancer cells by high-affinity CD16 + NK cells in two cancer cell lines.
Collapse
|
10
|
Feng J, Zheng H, Zhang Y, Wu H, Wang M, Sun Y, Zhang M, Xiao H, Qiao C, Wang J, Luo L, Li X, Feng J, Shi Y, Zheng Y, Wang Y, Sheng D, Chen G. pHLIP-fused PD-L1 engages avelumab to elicit NK cytotoxicity under acidic conditions. Heliyon 2024; 10:e30551. [PMID: 38756565 PMCID: PMC11096742 DOI: 10.1016/j.heliyon.2024.e30551] [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: 11/28/2023] [Revised: 04/28/2024] [Accepted: 04/29/2024] [Indexed: 05/18/2024] Open
Abstract
Natural killer (NK) cells represent key player in immune surveillance to eliminate transformed or malignant cells. One of mechanisms of action of NK cells is antibody-dependent cell-mediated cytotoxicity (ADCC) by recognizing tumor antigens on the surface of cancer cells. However, the heterogeneity of tumor antigens and the scarcity of membrane surface targets significantly restrict this strategy. Recently, we constructed a new cargo by tethering a low pH insertion peptide (pHLIP) to the C terminus of the ectodomain of programed death ligand-1 (PD-L1) and demonstrated its ability to modulate immune responses. Herein, the potential application of PD-L1-pHLIP in cancer therapy was determined. pHLIP tethering had no effect on the binding capacity of PD-L1 protein to an anti-PD-L1 antibody (i.e. avelumab). Association of pHLIP rendered PD-L1 segment display on the surface of cellular membrane in the acidic buffer instead of the neutral solution. Importantly, plate-coated or beads-coupled PD-L1-pHLIP enable robust activation and expression of cytotoxic mediators of NK cells via engaging avelumab. Overall, this work provides proof of concept that recombinant PD-L1 protein decorated on the cellular membrane driven by pHLIP in combination with appropriate monoclonal antibody has potentials to elicit NK cytotoxicity, which may represent a novel and promising therapeutic avenue in cancer.
Collapse
Affiliation(s)
- Junjuan Feng
- Inner Mongolia Key Lab of Molecular Biology, School of Basic Medical Sciences, Inner Mongolia Medical University, Hohhot, Inner Mongolia, 010058, PR China
- State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and Toxicology, Beijing, 100850, PR China
| | - Hang Zheng
- Inner Mongolia Key Lab of Molecular Biology, School of Basic Medical Sciences, Inner Mongolia Medical University, Hohhot, Inner Mongolia, 010058, PR China
- State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and Toxicology, Beijing, 100850, PR China
| | - Yuting Zhang
- Inner Mongolia Key Lab of Molecular Biology, School of Basic Medical Sciences, Inner Mongolia Medical University, Hohhot, Inner Mongolia, 010058, PR China
- State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and Toxicology, Beijing, 100850, PR China
| | - Haiyan Wu
- State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and Toxicology, Beijing, 100850, PR China
| | - Mianjing Wang
- Inner Mongolia Key Lab of Molecular Biology, School of Basic Medical Sciences, Inner Mongolia Medical University, Hohhot, Inner Mongolia, 010058, PR China
- State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and Toxicology, Beijing, 100850, PR China
| | - Ying Sun
- Inner Mongolia Key Lab of Molecular Biology, School of Basic Medical Sciences, Inner Mongolia Medical University, Hohhot, Inner Mongolia, 010058, PR China
| | - Min Zhang
- State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and Toxicology, Beijing, 100850, PR China
| | - He Xiao
- State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and Toxicology, Beijing, 100850, PR China
| | - Chunxia Qiao
- State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and Toxicology, Beijing, 100850, PR China
| | - Jing Wang
- State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and Toxicology, Beijing, 100850, PR China
| | - Longlong Luo
- State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and Toxicology, Beijing, 100850, PR China
| | - Xinying Li
- State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and Toxicology, Beijing, 100850, PR China
| | - Jiannan Feng
- State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and Toxicology, Beijing, 100850, PR China
| | - Yanchun Shi
- Inner Mongolia Key Lab of Molecular Biology, School of Basic Medical Sciences, Inner Mongolia Medical University, Hohhot, Inner Mongolia, 010058, PR China
| | - Yuanqiang Zheng
- Inner Mongolia Key Lab of Molecular Biology, School of Basic Medical Sciences, Inner Mongolia Medical University, Hohhot, Inner Mongolia, 010058, PR China
| | - Yi Wang
- Department of Hematology, The Fifth Medical Center, Chinese PLA General Hospital, Beijing, 100071, PR China
| | - Dongsheng Sheng
- Department of Thoracic surgery, The Fifth Medical Center, Chinese PLA General Hospital, Beijing, 100071, PR China
| | - Guojiang Chen
- State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and Toxicology, Beijing, 100850, PR China
| |
Collapse
|
11
|
Chirizzi C, Pellegatta S, Gori A, Falco J, Rubiu E, Acerbi F, Bombelli FB. Next-generation agents for fluorescence-guided glioblastoma surgery. Bioeng Transl Med 2024; 9:e10608. [PMID: 38818124 PMCID: PMC11135154 DOI: 10.1002/btm2.10608] [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: 06/29/2023] [Revised: 08/09/2023] [Accepted: 09/05/2023] [Indexed: 06/01/2024] Open
Abstract
Glioblastoma is a fast-growing and aggressive form of brain cancer. Even with maximal treatment, patients show a low median survival and are often subjected to a high recurrence incidence. The currently available treatments require multimodal management, including maximal safe surgical resection, followed by radiation and chemotherapy. Because of the infiltrative glioblastoma nature, intraoperative differentiation of cancer tissue from normal brain parenchyma is very challenging, and this accounts for the low rate of complete tumor resection. For these reasons, clinicians have increasingly used various intraoperative adjuncts to improve surgical results, such as fluorescent agents. However, most of the existing fluorophores show several limitations such as poor selectivity, photostability, photosensitization and high costs. This could limit their application to successfully improve glioblastoma resection. In the present perspective, we highlight the possibility to develop next-generation fluorescent tools able to more selectively label cancer cells during surgical resection.
Collapse
Affiliation(s)
- Cristina Chirizzi
- Department of Chemistry, Materials and Chemical Engineering “Giulio Natta”Politecnico di MilanoMilanoItaly
| | - Serena Pellegatta
- Unit of Immunotherapy of Brain TumorsFondazione IRCCS Istituto Neurologico Carlo BestaMilanItaly
- Unit of NeuroncologyFondazione IRCCS Istituto Neurologico Carlo BestaMilanItaly
| | - Alessandro Gori
- National Research Council of Italy, Istituto di Scienze e Tecnologie Chimiche (SCITEC‐CNR)MilanItaly
| | - Jacopo Falco
- Neurosurgical Unit 2, Department of NeurosurgeryFondazione IRCCS Istituto Neurologico Carlo BestaMilanItaly
| | - Emanuele Rubiu
- Neurosurgical Unit 2, Department of NeurosurgeryFondazione IRCCS Istituto Neurologico Carlo BestaMilanItaly
| | - Francesco Acerbi
- Neurosurgical Unit 2, Department of NeurosurgeryFondazione IRCCS Istituto Neurologico Carlo BestaMilanItaly
- Experimental Microsurgical Laboratory, Department of NeurosurgeryFondazione IRCCS Istituto Neurologico Carlo BestaMilanoItaly
| | | |
Collapse
|
12
|
Lefevre C, Thibaut MM, Loumaye A, Thissen JP, Neyrinck AM, Navez B, Delzenne NM, Feron O, Bindels LB. Tumoral acidosis promotes adipose tissue depletion by fostering adipocyte lipolysis. Mol Metab 2024; 83:101930. [PMID: 38570069 PMCID: PMC11027574 DOI: 10.1016/j.molmet.2024.101930] [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: 01/15/2024] [Revised: 03/20/2024] [Accepted: 03/28/2024] [Indexed: 04/05/2024] Open
Abstract
OBJECTIVE Tumour progression drives profound alterations in host metabolism, such as adipose tissue depletion, an early event of cancer cachexia. As fatty acid consumption by cancer cells increases upon acidosis of the tumour microenvironment, we reasoned that fatty acids derived from distant adipose lipolysis may sustain tumour fatty acid craving, leading to the adipose tissue loss observed in cancer cachexia. METHODS To evaluate the pro-lipolytic capacities of acid-exposed cancer cells, primary mouse adipocytes from subcutaneous and visceral adipose tissue were exposed to pH-matched conditioned medium from human and murine acid-exposed cancer cells (pH 6.5), compared to naive cancer cells (pH 7.4). To further address the role of tumoral acidosis on adipose tissue loss, a pH-low insertion peptide was injected into tumour-bearing mice, and tumoral acidosis was neutralised with a sodium bicarbonate buffer. Prolipolytic mediators were identified by transcriptomic approaches and validated on murine and human adipocytes. RESULTS Here, we reveal that acid-exposed cancer cells promote lipolysis from subcutaneous and visceral adipocytes and that dampening acidosis in vivo inhibits adipose tissue depletion. We further found a set of well-known prolipolytic factors enhanced upon acidosis adaptation and unravelled a role for β-glucuronidase (GUSB) as a promising new actor in adipocyte lipolysis. CONCLUSIONS Tumoral acidosis promotes the mobilization of fatty acids derived from adipocytes via the release of soluble factors by cancer cells. Our work paves the way for therapeutic approaches aimed at tackling cachexia by targeting the tumour acidic compartment.
Collapse
Affiliation(s)
- Camille Lefevre
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium.
| | - Morgane M Thibaut
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Audrey Loumaye
- Department of Endocrinology and Nutrition, Cliniques Universitaires Saint-Luc, Brussels, Belgium; Department of Endocrinology, Diabetology and Nutrition, IREC, UCLouvain, Brussels, Belgium
| | - Jean-Paul Thissen
- Department of Endocrinology and Nutrition, Cliniques Universitaires Saint-Luc, Brussels, Belgium; Department of Endocrinology, Diabetology and Nutrition, IREC, UCLouvain, Brussels, Belgium
| | - Audrey M Neyrinck
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Benoit Navez
- Department of Abdominal Surgery and Transplantation, Cliniques universitaires Saint-Luc, Brussels, Belgium
| | - Nathalie M Delzenne
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Olivier Feron
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, Brussels, Belgium; Welbio Department, WEL Research Institute, Wavre, Belgium
| | - Laure B Bindels
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium; Welbio Department, WEL Research Institute, Wavre, Belgium.
| |
Collapse
|
13
|
Frolova AY, Pakhomov AA, Kakuev DL, Sungurova AS, Dremina AA, Mamontova ED, Deyev SM, Martynov VI. Hybrid protein-peptide system for the selective pH-dependent binding and photodynamic ablation of cancer cells. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY. B, BIOLOGY 2023; 249:112803. [PMID: 37924677 DOI: 10.1016/j.jphotobiol.2023.112803] [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: 06/05/2023] [Revised: 10/02/2023] [Accepted: 10/24/2023] [Indexed: 11/06/2023]
Abstract
Creating new tools for the early diagnosis and treatment of cancer is one of the most important and intensively developing areas of modern medicine. Currently, photodynamic cancer therapy (PDT) is attracting increasing attention as a unique modality of minimally invasive treatment and due to the absence of acquired resistance. However, PDT is associated with undesirable activities, such as non-specific photodynamic effects of sunlight on healthy tissues. Therefore, an important fundamental task is the development of improved PDT agents that selectively act on the affected areas. Here, we report the development of a hybrid protein-peptide system for the selective pH-dependent binding and subsequent photodynamic cancer cells ablation. It is known that a distinctive feature of cancer cells is a decreased pH level in the extracellular space. In this study we exploited a peptide fragment (pHLIP) as a targeting module, which spontaneously binds and embeds into the cell membrane when pH decreases below neutral. A mutant of miniSOG protein fused to pHLIP was used as a photosensitizing constituent. We demonstrate that this protein-peptide photosensitizing system selectively binds to HeLa cells at pH below 6.8 and kills them when exposed to light. These findings demonstrate the feasibility of using genetically encoded MiniSOG fusions with pHLIP for the targeted delivery of PSs to cancer cells and subsequent highly precise photodynamic therapy.
Collapse
Affiliation(s)
- Anastasiya Yu Frolova
- M.M. Shemyakin-Y.A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117997, Russian Federation
| | - Alexey A Pakhomov
- M.M. Shemyakin-Y.A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117997, Russian Federation.
| | - Dmitry L Kakuev
- M.M. Shemyakin-Y.A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117997, Russian Federation
| | - Anna S Sungurova
- M.M. Shemyakin-Y.A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117997, Russian Federation
| | - Anastasiya A Dremina
- M.M. Shemyakin-Y.A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117997, Russian Federation
| | - Elizaveta D Mamontova
- M.M. Shemyakin-Y.A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117997, Russian Federation
| | - Sergey M Deyev
- M.M. Shemyakin-Y.A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117997, Russian Federation
| | - Vladimir I Martynov
- M.M. Shemyakin-Y.A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117997, Russian Federation
| |
Collapse
|
14
|
Yang J, Qu J, Teng X, Zhu W, Xu Y, Yang Y, Qian X. Tumor Microenvironment-Responsive Hydrogel for Direct Extracellular ATP Imaging-Guided Surgical Resection with Clear Boundaries. Adv Healthc Mater 2023; 12:e2301084. [PMID: 37219912 DOI: 10.1002/adhm.202301084] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 05/20/2023] [Indexed: 05/24/2023]
Abstract
Most solid tumors are clinically treated using surgical resection, and the presence of residual tumor tissues at the surgical margins often determines tumor survival and recurrence. Herein, a hydrogel (Apt-HEX/Cp-BHQ1 Gel, termed AHB Gel) is developed for fluorescence-guided surgical resection. AHB Gel is constructed by tethering a polyacrylamide hydrogel and ATP-responsive aptamers together. It exhibits strong fluorescence under high ATP concentrations corresponding to the TME (100-500 µm) but shows little fluorescence at low ATP concentrations (10-100 nm) such as those in normal tissues. AHB Gel can rapidly (within 3 min) emit fluorescence after exposure to ATP, and the fluorescence signal only occurs at sites exposed to high ATP, resulting in a clear boundary between the ATP-high and ATP-low regions. In vivo, AHB Gel exhibits specific tumor-targeting capacity with no fluorescence response in normal tissue, providing clear tumor boundaries. In addition, AHB Gel has good storage stability, which is conducive to its future clinical application. In summary, AHB Gel is a novel tumor microenvironment-targeted DNA-hybrid hydrogel for ATP-based fluorescence imaging. It can enable the precise imaging of tumor tissues, showing promising application in fluorescence-guided surgeries in the future.
Collapse
Affiliation(s)
- Jingyi Yang
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, P. R. China
| | - Jiahao Qu
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, P. R. China
| | - Xuanming Teng
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, P. R. China
| | - Weiping Zhu
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, P. R. China
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, 130 Mei Long Road, Shanghai, 200237, P. R. China
| | - Yufang Xu
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, P. R. China
| | - Yangyang Yang
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, P. R. China
| | - Xuhong Qian
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, P. R. China
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, P. R. China
| |
Collapse
|
15
|
Chu T, Cao B, Wang P, Li B, Ren J, Nie G, Wei J, Li S. Tumor-Targeted Delivery of IL-2 by Fusing with a pH Low Insertion Peptide for Antitumor Immunotherapy. Bioconjug Chem 2023; 34:1894-1901. [PMID: 37787935 DOI: 10.1021/acs.bioconjchem.3c00401] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/04/2023]
Abstract
As a pleiotropic cytokine, interleukin-2 (IL-2) can effectively regulate lymphocyte proliferation, survival, and active antitumor immune responses in tumor microenvironments. Although the ability of IL-2 to boost immune responses was reported in cancer patients, its short circulating half-life and high toxicity hinder its broad and continual clinical application. Herein, we developed a novel tumor target agent by fusing pH low insertion peptides (pHLIP) with IL-2, forming the fusion protein pHLIP-IL2. Based on the low pH insertion property of pHLIP, the pHLIP-IL2 fusion protein could be selectively delivered to the acidic tumor microenvironments and then promote the proliferation of killer immune cells to elicit tumor regression. We found that pHLIP-IL2 fusion proteins can be significantly enriched in tumor tissues and can effectively reduce tumor size in diverse tumor models, including breast cancer and melanoma, without apparent adverse effects. These data suggest that the pHLIP-IL2 fusion protein may be a promising solution for the continual and extensive application of IL-2, and pHLIP-IL2 is a potential and valuable therapeutic drug for cancer patients with antitumor immunotherapy.
Collapse
Affiliation(s)
- Tianjiao Chu
- College of Pharmaceutical Science, Jilin University, Changchun 130021, China
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
| | - Bowei Cao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
| | - Peina Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
- College of Basic Medicine, Hebei Medical University, Shijiazhuang 050017, Hebei Province, China
| | - Bozhao Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
| | - Jinna Ren
- Hospital of Stomatology, Jilin University, Changchun 130021, China
| | - Guangjun Nie
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
- GBA Research Innovation Institute for Nanotechnology, Guangzhou 510530, China
| | - Jingyan Wei
- College of Pharmaceutical Science, Jilin University, Changchun 130021, China
| | - Suping Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
- GBA Research Innovation Institute for Nanotechnology, Guangzhou 510530, China
| |
Collapse
|
16
|
Wachira FW, Githirwa DC, McPartlon T, Nazarenko V, Gonzales JJC, Gazura MM, Leen C, Clary HR, Alston C, Klees LM, Yao L, An M. D-to-E and T19V Variants of the pH-Low Insertion Peptide and Their Doxorubicin Conjugates Interact with Membrane at Higher pH Ranges Than WT. Biochemistry 2023; 62:2997-3011. [PMID: 37793002 DOI: 10.1021/acs.biochem.3c00218] [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] [Indexed: 10/06/2023]
Abstract
To improve targeted cargo delivery to cancer cells, pH-Low Insertion Peptide (pHLIP) variants were developed to interact with the membrane at pH values higher than those of the WT. The Asp-to-Glu variants aim to increase side chain pKa without disturbing the sequence of protonations that underpin membrane insertion. The Thr19 variants represent efforts to perturb the critical Pro20 residue. To study the effect of cargo on pHLIP insertion, doxorubicin (Dox), a fluorescent antineoplastic drug, was conjugated to selected variants near the inserting C-terminus. Variants and conjugates were characterized on a POPC membrane using Trp and Dox fluorescence methods to define the entire pH range of insertion (pHinitial-pHfinal). Compared to WT with a pHi-pHf range of 6.7-5.6, D25E-D31E-D33E, D14E-D25E-D31E-D33E, and T19V-D25E variants demonstrated higher pHi-pHf ranges of 7.3-6.1, 7.3-6.3, and 8.2-5.4, respectively. The addition of Dox expanded the pHi-pHf range, mainly by shifting pHi to higher pH values (e.g., WT pHLIP-Dox has a pHi-pHf range of 7.7-5.2). Despite the low Hill coefficient observed for the conjugates, D14E-D25E-D31E-D33E pHLIP-Dox completed insertion by a pHf of 5.7. However, the Dox cargo remained in the hydrophobic membrane interior after pHLIP insertion, which may impede drug release. Finally, a logistic function can describe pHLIP insertion as a peripheral-to-TM (start-to-finish) two-state transition; wherever possible, we discuss data deviating from such sigmoidal fitting in support of the idea that pH-specific intermediate states distinct from the initial peripheral state and the final TM state exist at intervening pH values.
Collapse
Affiliation(s)
- Faith W Wachira
- Department of Chemistry, State University of New York (SUNY), Binghamton University, Binghamton, New York 13902, United States
| | - Dancan C Githirwa
- Department of Chemistry, State University of New York (SUNY), Binghamton University, Binghamton, New York 13902, United States
| | - Thomas McPartlon
- Department of Chemistry, State University of New York (SUNY), Binghamton University, Binghamton, New York 13902, United States
| | - Vladyslav Nazarenko
- Department of Chemistry, State University of New York (SUNY), Binghamton University, Binghamton, New York 13902, United States
| | - Jerel J C Gonzales
- Department of Chemistry, State University of New York (SUNY), Binghamton University, Binghamton, New York 13902, United States
| | - Makenzie M Gazura
- Department of Chemistry, State University of New York (SUNY), Binghamton University, Binghamton, New York 13902, United States
| | - Caitlin Leen
- Department of Chemistry, State University of New York (SUNY), Binghamton University, Binghamton, New York 13902, United States
| | - Hannah R Clary
- Department of Chemistry, State University of New York (SUNY), Binghamton University, Binghamton, New York 13902, United States
| | - Claire Alston
- Department of Chemistry, State University of New York (SUNY), Binghamton University, Binghamton, New York 13902, United States
| | - Lukas M Klees
- Department of Chemistry, State University of New York (SUNY), Binghamton University, Binghamton, New York 13902, United States
| | - Lan Yao
- Department of Chemistry, State University of New York (SUNY), Binghamton University, Binghamton, New York 13902, United States
- Department of Physics, SUNY, Binghamton University, Binghamton, New York 13902, United States
| | - Ming An
- Department of Chemistry, State University of New York (SUNY), Binghamton University, Binghamton, New York 13902, United States
| |
Collapse
|
17
|
Lu X, Wang X, Gao S, Chen Z, Bai R, Wang Y. Bioparameter-directed nanoformulations as MRI CAs enable the specific visualization of hypoxic tumour. Analyst 2023; 148:4967-4981. [PMID: 37724375 DOI: 10.1039/d3an00972f] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/20/2023]
Abstract
A malignant tumour has hypoxic cells of varying degrees. The more severe the hypoxic degree, the more difficult the prognosis of the tumour and the higher the recurrence rate. Therefore, tumour hypoxia imaging is crucial. Magnetic resonance imaging (MRI) shows its strength in high resolution, depth of penetration and noninvasiveness. However, it needs more excellent contrast agents (CAs) to combat the complex tumour microenvironment (TME) and increased targeting of tumours to enhance clinical safety. Many research studies have focused on developing hypoxia-responsive MRI CAs that take advantage of the unique characteristics of hypoxic tumours. The low oxygen pressure, acidic TME, and up-regulated redox molecule levels found in hypoxic tumours serve as biological stimuli for nanoformulations that can accurately image the hypoxic region. This review highlights the importance of developing bioparameter-directed nanoformulations as MRI CAs for accurate tumour diagnosis. The design strategies and mechanisms of tumour-hypoxia imaging with nanoformulations are exemplified, with a focus on pH-responsiveness, redox-responsiveness, and p(O2)-responsiveness. The promising future of bioparameter-responsive nanoformulations for accurate tumour diagnosis and personalised cancer treatment is discussed.
Collapse
Affiliation(s)
- Xinyi Lu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, 100190, P. R. China.
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Xin Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, 100190, P. R. China.
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Susu Gao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, 100190, P. R. China.
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Ziwei Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, 100190, P. R. China.
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Ru Bai
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, 100190, P. R. China.
| | - Yaling Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, 100190, P. R. China.
| |
Collapse
|
18
|
Mc Larney BE, Kim M, Roberts S, Skubal M, Hsu HT, Ogirala A, Pratt EC, Pillarsetty NVK, Heller DA, Lewis JS, Grimm J. Ambient Light Resistant Shortwave Infrared Fluorescence Imaging for Preclinical Tumor Delineation via the pH Low-Insertion Peptide Conjugated to Indocyanine Green. J Nucl Med 2023; 64:1647-1653. [PMID: 37620049 PMCID: PMC10586478 DOI: 10.2967/jnumed.123.265686] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 06/12/2023] [Indexed: 08/26/2023] Open
Abstract
Shortwave infrared (900-1,700 nm) fluorescence imaging (SWIRFI) has shown significant advantages over visible (400-650 nm) and near-infrared (700-900 nm) fluorescence imaging (reduced autofluorescence, improved contrast, tissue resolution, and depth sensitivity). However, there is a major lag in the clinical translation of preclinical SWIRFI systems and targeted SWIRFI probes. Methods: We preclinically show that the pH low-insertion peptide conjugated to indocyanine green (pHLIP ICG), currently in clinical trials, is an excellent candidate for cancer-targeted SWIRFI. Results: pHLIP ICG SWIRFI achieved picomolar sensitivity (0.4 nM) with binary and unambiguous tumor screening and resection up to 96 h after injection in an orthotopic breast cancer mouse model. SWIRFI tumor screening and resection had ambient light resistance (possible without gating or filtering) with outstanding signal-to-noise ratio (SNR) and contrast-to-noise ratio (CNR) values at exposures from 10 to 0.1 ms. These SNR and CNR values were also found for the extended emission of pHLIP ICG in vivo (>1,100 nm, 300 ms). Conclusion: SWIRFI sensitivity and ambient light resistance enabled continued tracer clearance tracking with unparalleled SNR and CNR values at video rates for tumor delineation (achieving a tumor-to-muscle ratio above 20). In total, we provide a direct precedent for the democratic translation of an ambient light resistant SWIRFI and pHLIP ICG ecosystem, which can instantly improve tumor resection.
Collapse
Affiliation(s)
| | - Mijin Kim
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Sheryl Roberts
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Magdalena Skubal
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Hsiao-Ting Hsu
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Anuja Ogirala
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Edwin C Pratt
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Naga Vara Kishore Pillarsetty
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Radiology, Weill Cornell Medicine, New York, New York; and
| | - Daniel A Heller
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Pharmacology, Weill Cornell Medicine, New York, New York
| | - Jason S Lewis
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Pharmacology, Weill Cornell Medicine, New York, New York
- Department of Radiology, Weill Cornell Medicine, New York, New York; and
| | - Jan Grimm
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York;
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Pharmacology, Weill Cornell Medicine, New York, New York
- Department of Radiology, Weill Cornell Medicine, New York, New York; and
- Molecular Imaging Therapy Service, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
19
|
Hajjar S, Zhou X. pH sensing at the intersection of tissue homeostasis and inflammation. Trends Immunol 2023; 44:807-825. [PMID: 37714775 PMCID: PMC10543622 DOI: 10.1016/j.it.2023.08.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 08/13/2023] [Accepted: 08/13/2023] [Indexed: 09/17/2023]
Abstract
pH is tightly maintained at cellular, tissue, and systemic levels, and altered pH - particularly in the acidic range - is associated with infection, injury, solid tumors, and physiological and pathological inflammation. However, how pH is sensed and regulated and how it influences immune responses remain poorly understood at the tissue level. Applying conceptual frameworks of homeostatic and inflammatory circuitries, we categorize cellular and tissue components engaged in pH regulation, drawing parallels from established cases in physiology. By expressing various intracellular (pHi) and extracellular pH (pHe)-sensing receptors, the immune system may integrate information on tissue and cellular states into the regulation of homeostatic and inflammatory programs. We introduce the novel concept of resistance and adaptation responses to rationalize pH-dependent immunomodulation intertwined with homeostatic equilibrium and inflammatory control. We discuss emerging challenges and opportunities in understanding the immunological roles of pH sensing, which might reveal new strategies to combat inflammation and restore tissue homeostasis.
Collapse
Affiliation(s)
- Stephanie Hajjar
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Boston, MA 02115, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA; Broad Institute of MIT and Harvard, 300 Longwood Ave, Boston, MA 02115, USA
| | - Xu Zhou
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Boston, MA 02115, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA; Broad Institute of MIT and Harvard, 300 Longwood Ave, Boston, MA 02115, USA.
| |
Collapse
|
20
|
Zhou Q, Xiang J, Qiu N, Wang Y, Piao Y, Shao S, Tang J, Zhou Z, Shen Y. Tumor Abnormality-Oriented Nanomedicine Design. Chem Rev 2023; 123:10920-10989. [PMID: 37713432 DOI: 10.1021/acs.chemrev.3c00062] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/17/2023]
Abstract
Anticancer nanomedicines have been proven effective in mitigating the side effects of chemotherapeutic drugs. However, challenges remain in augmenting their therapeutic efficacy. Nanomedicines responsive to the pathological abnormalities in the tumor microenvironment (TME) are expected to overcome the biological limitations of conventional nanomedicines, enhance the therapeutic efficacies, and further reduce the side effects. This Review aims to quantitate the various pathological abnormalities in the TME, which may serve as unique endogenous stimuli for the design of stimuli-responsive nanomedicines, and to provide a broad and objective perspective on the current understanding of stimuli-responsive nanomedicines for cancer treatment. We dissect the typical transport process and barriers of cancer drug delivery, highlight the key design principles of stimuli-responsive nanomedicines designed to tackle the series of barriers in the typical drug delivery process, and discuss the "all-into-one" and "one-for-all" strategies for integrating the needed properties for nanomedicines. Ultimately, we provide insight into the challenges and future perspectives toward the clinical translation of stimuli-responsive nanomedicines.
Collapse
Affiliation(s)
- Quan Zhou
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Department of Cell Biology, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Jiajia Xiang
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Department of Cell Biology, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Nasha Qiu
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| | - Yechun Wang
- Department of Cell Biology, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Ying Piao
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| | - Shiqun Shao
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| | - Jianbin Tang
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| | - Zhuxian Zhou
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| | - Youqing Shen
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- State Key Laboratory of Chemical Engineering, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
21
|
Xing W, Li X, Zhou Y, Li M, Zhu M. Lactate metabolic pathway regulates tumor cell metastasis and its use as a new therapeutic target. EXPLORATION OF MEDICINE 2023:541-559. [DOI: https:/doi.org/10.37349/emed.2023.00160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 04/17/2023] [Indexed: 09/04/2023] Open
Abstract
Abnormal energy metabolism is one of the ten hallmarks of tumors, and tumor cell metabolism provides energy and a suitable microenvironment for tumorigenesis and metastasis. Tumor cells can consume large amounts of glucose and produce large amounts of lactate through glycolysis even in the presence of oxygen, a process called aerobic glycolysis, also known as the Warburg effect. Lactate is the end product of the aerobic glycolysis. Lactate dehydrogenase A (LDHA), which is highly expressed in cancer cells, promotes lactate production and transports lactate to the tumor microenvironment and is taken up by surrounding stromal cells under the action of monocarboxylate transporter 1/4 (MCT1/4), which in turn influences the immune response and enhances the invasion and metastasis of cancer cells. Therapeutic strategies targeting lactate metabolism have been intensively investigated, focusing on its metastasis-promoting properties and various target inhibitors; AZD3965, an MCT1 inhibitor, has entered phase I clinical trials, and the LDHA inhibitor N-hydroxyindole (NHI) has shown cancer therapeutic activity in pre-clinical studies. Interventions targeting lactate metabolism are emerging as a promising option for cancer therapy, with chemotherapy or radiotherapy combined with lactate-metabolism-targeted drugs adding to the effectiveness of cancer treatment. Based on current research, this article outlines the role of lactate metabolism in tumor metastasis and the potential value of inhibitors targeting lactate metabolism in cancer therapy.
Collapse
Affiliation(s)
- Weimei Xing
- Hainan Provincial Key Laboratory of Carcinogenesis and Intervention, Hainan Medical University, Haikou 571199, Hainan, China
| | - Xiaowei Li
- Hainan Provincial Key Laboratory of Carcinogenesis and Intervention, Hainan Medical University, Haikou 571199, Hainan, China
| | - Yuli Zhou
- Hainan Provincial Key Laboratory of Carcinogenesis and Intervention, Hainan Medical University, Haikou 571199, Hainan, China
| | - Mengsen Li
- Hainan Provincial Key Laboratory of Carcinogenesis and Intervention, Hainan Medical University, Haikou 571199, Hainan, China; Department of Medical Oncology, Second Affiliated Hospital, Hainan Medical University, Haikou 570311, Hainan, China; Institution of Tumour, First Affiliated Hospital, Hainan Medical University, Haikou 570102, Hainan, China
| | - Mingyue Zhu
- Hainan Provincial Key Laboratory of Carcinogenesis and Intervention, Hainan Medical University, Haikou 571199, Hainan, China
| |
Collapse
|
22
|
Li Z, Zheng Y, Yan J, Yan Y, Peng C, Wang Z, Liu H, Liu Y, Zhou Y, Ding M. Self-Assembly of Poly(Amino Acid)s Mediated by Secondary Conformations. Chembiochem 2023; 24:e202300132. [PMID: 37340829 DOI: 10.1002/cbic.202300132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 06/16/2023] [Accepted: 06/20/2023] [Indexed: 06/22/2023]
Abstract
Self-assembly of block copolymers has recently drawn great attention due to its remarkable performance and wide variety of applications in biomedicine, biomaterials, microelectronics, photoelectric materials, catalysts, etc. Poly(amino acid)s (PAAs), formed by introducing synthetic amino acids into copolymer backbones, are able to fold into different secondary conformations when compared with traditional amphiphilic copolymers. Apart from changing the chemical composition and degree of polymerization of copolymers, the self-assembly behaviors of PAAs could be controlled by their secondary conformations, which are more flexible and adjustable for fine structure tailoring. In this article, we summarize the latest findings on the variables that influence secondary conformations, in particular the regulation of order-to-order conformational changes and the approaches used to manage the self-assembly behaviors of PAAs. These strategies include controlling pH, redox reactions, coordination, light, temperature, and so on. Hopefully, we can provide valuable perspectives that will be useful for the future development and use of synthetic PAAs.
Collapse
Affiliation(s)
- Zifen Li
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065, China
| | - Yi Zheng
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065, China
| | - Jingyue Yan
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065, China
| | - Yue Yan
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065, China
| | - Chuan Peng
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065, China
| | - Zuojie Wang
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065, China
| | - Hang Liu
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065, China
| | - Yang Liu
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065, China
| | - Yeqiang Zhou
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065, China
| | - Mingming Ding
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065, China
| |
Collapse
|
23
|
Qiu C, Xia F, Zhang J, Shi Q, Meng Y, Wang C, Pang H, Gu L, Xu C, Guo Q, Wang J. Advanced Strategies for Overcoming Endosomal/Lysosomal Barrier in Nanodrug Delivery. RESEARCH (WASHINGTON, D.C.) 2023; 6:0148. [PMID: 37250954 PMCID: PMC10208951 DOI: 10.34133/research.0148] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 04/27/2023] [Indexed: 05/31/2023]
Abstract
Nanocarriers have therapeutic potential to facilitate drug delivery, including biological agents, small-molecule drugs, and nucleic acids. However, their efficiency is limited by several factors; among which, endosomal/lysosomal degradation after endocytosis is the most important. This review summarizes advanced strategies for overcoming endosomal/lysosomal barriers to efficient nanodrug delivery based on the perspective of cellular uptake and intracellular transport mechanisms. These strategies include promoting endosomal/lysosomal escape, using non-endocytic methods of delivery to directly cross the cell membrane to evade endosomes/lysosomes and making a detour pathway to evade endosomes/lysosomes. On the basis of the findings of this review, we proposed several promising strategies for overcoming endosomal/lysosomal barriers through the smarter and more efficient design of nanodrug delivery systems for future clinical applications.
Collapse
Affiliation(s)
- Chong Qiu
- Artemisinin Research Center, and Institute of Chinese Materia Medica,
China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Fei Xia
- Artemisinin Research Center, and Institute of Chinese Materia Medica,
China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Junzhe Zhang
- Artemisinin Research Center, and Institute of Chinese Materia Medica,
China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Qiaoli Shi
- Artemisinin Research Center, and Institute of Chinese Materia Medica,
China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Yuqing Meng
- Artemisinin Research Center, and Institute of Chinese Materia Medica,
China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Chen Wang
- Artemisinin Research Center, and Institute of Chinese Materia Medica,
China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Huanhuan Pang
- Artemisinin Research Center, and Institute of Chinese Materia Medica,
China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Liwei Gu
- Artemisinin Research Center, and Institute of Chinese Materia Medica,
China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Chengchao Xu
- Artemisinin Research Center, and Institute of Chinese Materia Medica,
China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Qiuyan Guo
- Artemisinin Research Center, and Institute of Chinese Materia Medica,
China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Jigang Wang
- Artemisinin Research Center, and Institute of Chinese Materia Medica,
China Academy of Chinese Medical Sciences, Beijing 100700, China
- Department of Nephrology, and Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital, The First Affiliated Hospital,
Southern University of Science and Technology, Shenzhen, Guangdong 518020, China
| |
Collapse
|
24
|
Shtykalova S, Deviatkin D, Freund S, Egorova A, Kiselev A. Non-Viral Carriers for Nucleic Acids Delivery: Fundamentals and Current Applications. Life (Basel) 2023; 13:903. [PMID: 37109432 PMCID: PMC10142071 DOI: 10.3390/life13040903] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 03/21/2023] [Accepted: 03/24/2023] [Indexed: 03/31/2023] Open
Abstract
Over the past decades, non-viral DNA and RNA delivery systems have been intensively studied as an alternative to viral vectors. Despite the most significant advantage over viruses, such as the lack of immunogenicity and cytotoxicity, the widespread use of non-viral carriers in clinical practice is still limited due to the insufficient efficacy associated with the difficulties of overcoming extracellular and intracellular barriers. Overcoming barriers by non-viral carriers is facilitated by their chemical structure, surface charge, as well as developed modifications. Currently, there are many different forms of non-viral carriers for various applications. This review aimed to summarize recent developments based on the essential requirements for non-viral carriers for gene therapy.
Collapse
Affiliation(s)
- Sofia Shtykalova
- Department of Genomic Medicine, D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductology, Mendeleevskaya Line 3, 199034 Saint-Petersburg, Russia
- Faculty of Biology, Saint-Petersburg State University, Universitetskaya Embankment 7-9, 199034 Saint-Petersburg, Russia
| | - Dmitriy Deviatkin
- Department of Genomic Medicine, D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductology, Mendeleevskaya Line 3, 199034 Saint-Petersburg, Russia
- Faculty of Biology, Saint-Petersburg State University, Universitetskaya Embankment 7-9, 199034 Saint-Petersburg, Russia
| | - Svetlana Freund
- Department of Genomic Medicine, D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductology, Mendeleevskaya Line 3, 199034 Saint-Petersburg, Russia
- Faculty of Biology, Saint-Petersburg State University, Universitetskaya Embankment 7-9, 199034 Saint-Petersburg, Russia
| | - Anna Egorova
- Department of Genomic Medicine, D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductology, Mendeleevskaya Line 3, 199034 Saint-Petersburg, Russia
| | - Anton Kiselev
- Department of Genomic Medicine, D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductology, Mendeleevskaya Line 3, 199034 Saint-Petersburg, Russia
| |
Collapse
|
25
|
Mahto AK, Kumari S, Akbar S, Paroha S, Sahoo PK, Kumar A, Dewangan RP. Peptide-Based Therapeutics and Drug Delivery Systems. DRUGS AND A METHODOLOGICAL COMPENDIUM 2023:173-211. [DOI: 10.1007/978-981-19-7952-1_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
26
|
Gayle S, Paradis T, Jones K, Vasquez J, Paralkar VM. Antigen-independent tumor targeting by CBX-12 (alphalex ™-exatecan) induces long-term antitumor immunity. Immunotherapy 2022; 14:1467-1480. [PMID: 36597724 DOI: 10.2217/imt-2022-0121] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Aims: To determine whether antigen-independent targeting of the TOP1 inhibitor exatecan to tumor with a pH-sensitive peptide (CBX-12) produces superior synergy with immunotherapy compared with unconjugated exatecan. Materials & methods: In vitro and ex vivo functional assays were performed via FACS and ELISA assays. In vivo efficacy was evaluated in the syngeneic CT26 model. Results: CBX-12 combined with anti-PD-1 or anti-CTLA4 results in delayed tumor growth and complete response, with cured animals displaying long-term antitumor immunity. CBX-12 stimulates expression of MHC 1 and PD-L1 and is an inducer of immunogenic cell death, producing long-term immune recognition of tumor cells and resultant antitumor immunity. Conclusion: The authors' data provide the rationale for exploring immunotherapy combinations with CBX-12 in clinical trials.
Collapse
Affiliation(s)
| | | | - Kelli Jones
- Cybrexa Therapeutics, New Haven, CT 06511, USA
| | - Juan Vasquez
- Section of Hematology & Oncology, Department of Pediatrics, Yale University School of Medicine, New Haven, CT 06520, USA
| | | |
Collapse
|
27
|
Visca H, DuPont M, Moshnikova A, Crawford T, Engelman DM, Andreev OA, Reshetnyak YK. pHLIP Peptides Target Acidity in Activated Macrophages. Mol Imaging Biol 2022; 24:874-885. [PMID: 35604527 PMCID: PMC9681937 DOI: 10.1007/s11307-022-01737-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 03/19/2022] [Accepted: 04/27/2022] [Indexed: 12/29/2022]
Abstract
PURPOSE Acidity can be a useful alternative biomarker for the targeting of metabolically active cells in certain diseased tissues, as in acute inflammation or aggressive tumors. We investigated the targeting of activated macrophages by pH low insertion peptides (pHLIPs), an established technology for targeting cell-surface acidity. PROCEDURES The uptake of fluorescent pHLIPs by activated macrophages was studied in cell cultures, in a mouse model of lung inflammation, and in a mouse tumor model. Fluorescence microscopy, whole-body and organ imaging, immunohistochemistry, and FACS analysis were employed. RESULTS We find that cultured, activated macrophages readily internalize pHLIPs. The uptake is higher in glycolytic macrophages activated by LPS and INF-γ compared to macrophages activated by IL-4/IL-13. Fluorescent pHLIPs target LPS-induced lung inflammation in mice. In addition to marking cancer cells within the tumor microenvironment, fluorescent pHLIPs target CD45+, CD11b+, F4/80+, and CD206+ tumor-associated macrophages with no significant targeting of other immune cells. Also, fluorescent pHLIPs target CD206-positive cells found in the inguinal lymph nodes of animals inoculated with breast cancer cells in mammary fat pads. CONCLUSIONS pHLIP peptides sense low cell surface pH, which triggers their insertion into the cell membrane. Unlike cancerous cells, activated macrophages do not retain inserted pHLIPs on their surfaces, instead their highly active membrane recycling moves the pHLIPs into endosomes. Targeting activated macrophages in diseased tissues may enable clinical visualization and therapeutic opportunities.
Collapse
Affiliation(s)
- Hannah Visca
- Physics Department, University of Rhode Island, Kingston, RI, USA
| | - Michael DuPont
- Physics Department, University of Rhode Island, Kingston, RI, USA
| | - Anna Moshnikova
- Physics Department, University of Rhode Island, Kingston, RI, USA
| | - Troy Crawford
- Physics Department, University of Rhode Island, Kingston, RI, USA
| | - Donald M Engelman
- Department of Molecular Biophysics and Biochemistry, Yale, New Haven, CT, USA
| | - Oleg A Andreev
- Physics Department, University of Rhode Island, Kingston, RI, USA
| | | |
Collapse
|
28
|
Multiplexed Imaging Reveals the Spatial Relationship of the Extracellular Acidity-Targeting pHLIP with Necrosis, Hypoxia, and the Integrin-Targeting cRGD Peptide. Cells 2022; 11:cells11213499. [DOI: 10.3390/cells11213499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 10/24/2022] [Accepted: 11/02/2022] [Indexed: 11/09/2022] Open
Abstract
pH (low) insertion peptides (pHLIPs) have been developed for cancer imaging and therapy targeting the acidic extracellular microenvironment. However, the characteristics of intratumoral distribution (ITD) of pHLIPs are not yet fully understood. This study aimed to reveal the details of the ITD of pHLIPs and their spatial relationship with other tumor features of concern. The fluorescent dye-labeled pHLIPs were intravenously administered to subcutaneous xenograft mouse models of U87MG and IGR-OV1 expressing αVβ3 integrins (using large necrotic tumors). The αVβ3 integrin-targeting Cy5.5-RAFT-c(-RGDfK-)4 was used as a reference. In vivo and ex vivo fluorescence imaging, whole-tumor section imaging, fluorescence microscopy, and multiplexed fluorescence colocalization analysis were performed. The ITD of fluorescent dye-labeled pHLIPs was heterogeneous, having a high degree of colocalization with necrosis. A direct one-to-one comparison of highly magnified images revealed the cellular localization of pHLIP in pyknotic, karyorrhexis, and karyolytic necrotic cells. pHLIP and hypoxia were spatially contiguous but not overlapping cellularly. The hypoxic region was found between the ITDs of pHLIP and the cRGD peptide and the Ki-67 proliferative activity remained detectable in the pHLIP-accumulated regions. The results provide a better understanding of the characteristics of ITD of pHLIPs, leading to new insights into the theranostic applications of pHLIPs.
Collapse
|
29
|
Zhou W, Jia Y, Liu Y, Chen Y, Zhao P. Tumor Microenvironment-Based Stimuli-Responsive Nanoparticles for Controlled Release of Drugs in Cancer Therapy. Pharmaceutics 2022; 14:2346. [PMID: 36365164 PMCID: PMC9694300 DOI: 10.3390/pharmaceutics14112346] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 10/22/2022] [Accepted: 10/28/2022] [Indexed: 07/22/2023] Open
Abstract
With the development of nanomedicine technology, stimuli-responsive nanocarriers play an increasingly important role in antitumor therapy. Compared with the normal physiological environment, the tumor microenvironment (TME) possesses several unique properties, including acidity, high glutathione (GSH) concentration, hypoxia, over-expressed enzymes and excessive reactive oxygen species (ROS), which are closely related to the occurrence and development of tumors. However, on the other hand, these properties could also be harnessed for smart drug delivery systems to release drugs specifically in tumor tissues. Stimuli-responsive nanoparticles (srNPs) can maintain stability at physiological conditions, while they could be triggered rapidly to release drugs by specific stimuli to prolong blood circulation and enhance cancer cellular uptake, thus achieving excellent therapeutic performance and improved biosafety. This review focuses on the design of srNPs based on several stimuli in the TME for the delivery of antitumor drugs. In addition, the challenges and prospects for the development of srNPs are discussed, which can possibly inspire researchers to develop srNPs for clinical applications in the future.
Collapse
Affiliation(s)
- Weixin Zhou
- Department of Medical Ultrasound, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yujie Jia
- Institute of Biomedical Engineering and Technology, Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200065, China
| | - Yani Liu
- Department of Medical Ultrasound, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yan Chen
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Pengxuan Zhao
- Department of Medical Ultrasound, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
30
|
Liu CX, Wang B, Zhu WP, Xu YF, Yang YY, Qian XH. An Endoplasmic Reticulum (ER)‐Targeting DNA Nanodevice for Autophagy‐Dependent Degradation of Proteins in Membrane‐Bound Organelles. Angew Chem Int Ed Engl 2022; 61:e202205509. [DOI: 10.1002/anie.202205509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Indexed: 11/09/2022]
Affiliation(s)
- Caixia X. Liu
- Shanghai Key Laboratory of Chemical Biology School of Pharmacy East China University of Science and Technology 130 Meilong Road Shanghai 200237 P.R. China
| | - Bin Wang
- Shanghai Key Laboratory of Chemical Biology School of Pharmacy East China University of Science and Technology 130 Meilong Road Shanghai 200237 P.R. China
| | - Weiping P. Zhu
- Shanghai Key Laboratory of Chemical Biology School of Pharmacy East China University of Science and Technology 130 Meilong Road Shanghai 200237 P.R. China
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism School of Pharmacy East China University of Science and Technology 130 Mei Long Road Shanghai 200237 China
| | - Yufang F. Xu
- Shanghai Key Laboratory of Chemical Biology School of Pharmacy East China University of Science and Technology 130 Meilong Road Shanghai 200237 P.R. China
| | - Yangyang Y. Yang
- Shanghai Key Laboratory of Chemical Biology School of Pharmacy East China University of Science and Technology 130 Meilong Road Shanghai 200237 P.R. China
| | - Xuhong H. Qian
- Shanghai Key Laboratory of Chemical Biology School of Pharmacy East China University of Science and Technology 130 Meilong Road Shanghai 200237 P.R. China
- State Key Laboratory of Bioreactor Engineering East China University of Science and Technology 130 Meilong Road Shanghai 200237 P.R. China
| |
Collapse
|
31
|
Moshnikova A, Golijanin B, Amin A, Doyle J, Kott O, Gershman B, DuPont M, Li Y, Lu X, Engelman DM, Andreev OA, Reshetnyak YK, Golijanin D. Targeting Bladder Urothelial Carcinoma with pHLIP-ICG and Inhibition of Urothelial Cancer Cell Proliferation by pHLIP-amanitin. FRONTIERS IN UROLOGY 2022; 2:868919. [PMID: 36439552 PMCID: PMC9691284 DOI: 10.3389/fruro.2022.868919] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Acidity is a useful biomarker for the targeting of metabolically active-cells in tumors. pH Low Insertion Peptides (pHLIPs) sense the pH at the surfaces of tumor cells and can facilitate intracellular delivery of cell-permeable and cell-impermeable cargo molecules. In this study we have shown the targeting of malignant lesions in human bladders by fluorescent pHLIP agents, intracellular delivery of amanitin toxin by pHLIP for the inhibition of urothelial cancer cell proliferation, and enhanced potency of pHLIP-amanitin for cancer cells with 17p loss, a mutation frequently present in urothelial cancers. Twenty-eight ex-vivo bladder specimens, from patients undergoing robotic assisted laparoscopic radical cystectomy for bladder cancer, were treated via intravesical incubation for 15-60 minutes with pHLIP conjugated to indocyanine green (ICG) or IR-800 near infrared fluorescent (NIRF) dyes at concentrations of 4-8 μM. White light cystoscopy identified 47/58 (81%) and NIRF pHLIP cystoscopy identified 57/58 (98.3%) of malignant lesions of different subtypes and stages selected for histopathological processing. pHLIP NIRF imaging improved diagnosis by 17.3% (p < 0.05). All carcinoma-in-situ cases missed by white light cystoscopy were targeted by pHLIP agents and were diagnosed by NIRF imaging. We also investigated the interactions of pHLIP-amanitin with urothelial cancer cells of different grades. pHLIP-amanitin produced concentration- and pH-dependent inhibition of the proliferation of urothelial cancer cells treated for 2 hrs at concentrations up to 4 μM. A 3-4x enhanced cytotoxicity of pHLIP-amanitin was observed for cells with a 17p loss after 2 hrs of treatment at pH6. Potentially, pHLIP technology may improve the management of urothelial cancers, including imaging of malignant lesions using pHLIP-ICG for diagnosis and surgery, and the use of pHLIP-amanitin for treatment of superficial bladder cancers via intravesical instillation.
Collapse
Affiliation(s)
- Anna Moshnikova
- Physics Department, University of Rhode Island, Kingston, RI, USA
| | - Borivoj Golijanin
- Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School of Brown University, The Miriam Hospital, Providence, RI, USA
- Division of Urology, Department of Surgery, Brown University, The Miriam Hospital, Providence, RI, USA
| | - Ali Amin
- Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School of Brown University, The Miriam Hospital, Providence, RI, USA
| | - Joshua Doyle
- Physics Department, University of Rhode Island, Kingston, RI, USA
- Current address: Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, CT, USA
| | - Ohad Kott
- Division of Urology, Department of Surgery, Brown University, The Miriam Hospital, Providence, RI, USA
| | - Boris Gershman
- Division of Urology, Department of Surgery, Brown University, The Miriam Hospital, Providence, RI, USA
- Current address: Division of Urologic Surgery, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Michael DuPont
- Physics Department, University of Rhode Island, Kingston, RI, USA
| | - Yujing Li
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Xiongbin Lu
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
- Melvin & Bren Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Donald M. Engelman
- Department of Molecular Biophysics and Biochemistry, Yale, New Haven, CT, USA
| | - Oleg A. Andreev
- Physics Department, University of Rhode Island, Kingston, RI, USA
| | | | - Dragan Golijanin
- Division of Urology, Department of Surgery, Brown University, The Miriam Hospital, Providence, RI, USA
| |
Collapse
|
32
|
Di Z, Lu X, Zhao J, Jaklenec A, Zhao Y, Langer R, Li L. Mild Acidosis-Directed Signal Amplification in Tumor Microenvironment via Spatioselective Recruitment of DNA Amplifiers. Angew Chem Int Ed Engl 2022; 61:e202205436. [PMID: 35652128 DOI: 10.1002/anie.202205436] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Indexed: 12/29/2022]
Abstract
DNA biotechnology offers intriguing opportunities for amplification-based sensitive detection. However, spatiotemporally-controlled manipulation of signal amplification for in situ imaging of the tumor microenvironment remains an outstanding challenge. Here, we demonstrate a DNA-based strategy that can spatial-selectively amplify the acidic signal in the extracellular milieu of the tumor to achieve specific imaging with improved sensitivity. The strategy, termed mild acidosis-targeted amplification (MAT-amp), leverages the specific acidic microenvironment to engineer tumor cells with artificial DNA receptors through a pH (low) insertion peptide, which permits controlled recruitment of fluorescent amplifiers via a hybridization chain reaction. The acidosis-responsive amplification cascade enables significant fluorescence enhancement in tumors with a reduced background signal in normal tissues, leading to improved signal-to-background ratio. These results highlight the utility of MAT-amp for in situ imaging of the microenvironment characterized by pH disequilibrium.
Collapse
Affiliation(s)
- Zhenghan Di
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China.,College of Materials Science and Optoelectronic Technology, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xueguang Lu
- Key Laboratory of Colloid, Interface and Chemical Thermodynamics, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China.,David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Jian Zhao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China.,College of Materials Science and Optoelectronic Technology, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Ana Jaklenec
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Yuliang Zhao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
| | - Robert Langer
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Lele Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China.,College of Materials Science and Optoelectronic Technology, University of Chinese Academy of Sciences, Beijing, 100049, China
| |
Collapse
|
33
|
Liu C, Wang B, Zhu W, Xu Y, Yang Y, Qian X. An ER‐targeting DNA Nanodevice for Autophagy‐dependent Degradation of Proteins in Membrane‐bound Organelles. Angew Chem Int Ed Engl 2022. [DOI: 10.1002/ange.202205509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- Caixia Liu
- East China University of Science and Technology School of Pharmacy CHINA
| | - Bin Wang
- East China University of Science and Technology School of Pharmacy Shanghai CHINA
| | - Weiping Zhu
- East China University of Science and Technology School of Pharmacy CHINA
| | - Yufang Xu
- East China University of Science and Technology School of Pharmacy School of Pharmacy CHINA
| | - Yangyang Yang
- East China University of Science and Technology School of Pharmacy Meilong Road 130 200237 Shanghai CHINA
| | - Xuhong Qian
- East China University of Science and Technology School of Pharmacy CHINA
| |
Collapse
|
34
|
van der Heide CD, Dalm SU. Radionuclide imaging and therapy directed towards the tumor microenvironment: a multi-cancer approach for personalized medicine. Eur J Nucl Med Mol Imaging 2022; 49:4616-4641. [PMID: 35788730 PMCID: PMC9606105 DOI: 10.1007/s00259-022-05870-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 06/09/2022] [Indexed: 12/19/2022]
Abstract
Targeted radionuclide theranostics is becoming more and more prominent in clinical oncology. Currently, most nuclear medicine compounds researched for cancer theranostics are directed towards targets expressed in only a small subset of cancer types, limiting clinical applicability. The identification of cancer-specific targets that are (more) universally expressed will allow more cancer patients to benefit from these personalized nuclear medicine–based interventions. A tumor is not merely a collection of cancer cells, it also comprises supporting stromal cells embedded in an altered extracellular matrix (ECM), together forming the tumor microenvironment (TME). Since the TME is less genetically unstable than cancer cells, and TME phenotypes can be shared between cancer types, it offers targets that are more universally expressed. The TME is characterized by the presence of altered processes such as hypoxia, acidity, and increased metabolism. Next to the ECM, the TME consists of cancer-associated fibroblasts (CAFs), macrophages, endothelial cells forming the neo-vasculature, immune cells, and cancer-associated adipocytes (CAAs). Radioligands directed at the altered processes, the ECM, and the cellular components of the TME have been developed and evaluated in preclinical and clinical studies for targeted radionuclide imaging and/or therapy. In this review, we provide an overview of the TME targets and their corresponding radioligands. In addition, we discuss what developments are needed to further explore the TME as a target for radionuclide theranostics, with the hopes of stimulating the development of novel TME radioligands with multi-cancer, or in some cases even pan-cancer, application.
Collapse
Affiliation(s)
| | - Simone U Dalm
- Department of Radiology & Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands.
| |
Collapse
|
35
|
Jo U, Murai Y, Agama KK, Sun Y, Saha LK, Yang X, Arakawa Y, Gayle S, Jones K, Paralkar V, Sundaram RK, Doorn JV, Vasquez JC, Bindra RS, Choi WS, Pommier Y. TOP1-DNA Trapping by Exatecan and Combination Therapy with ATR Inhibitor. Mol Cancer Ther 2022; 21:1090-1102. [PMID: 35439320 PMCID: PMC9256811 DOI: 10.1158/1535-7163.mct-21-1000] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 02/18/2022] [Accepted: 04/12/2022] [Indexed: 01/07/2023]
Abstract
Exatecan and deruxtecan are antineoplastic camptothecin derivatives in development as tumor-targeted-delivery warheads in various formulations including peptides, liposomes, polyethylene glycol nanoparticles, and antibody-drug conjugates. Here, we report the molecular pharmacology of exatecan compared with the clinically approved topoisomerase I (TOP1) inhibitors and preclinical models for validating biomarkers and the combination of exatecan with ataxia telangiectasia and Rad3-related kinase (ATR) inhibitors. Modeling exatecan binding at the interface of a TOP1 cleavage complex suggests two novel molecular interactions with the flanking DNA base and the TOP1 residue N352, in addition to the three known interactions of camptothecins with the TOP1 residues R364, D533, and N722. Accordingly, exatecan showed much stronger TOP1 trapping, higher DNA damage, and apoptotic cell death than the classical TOP1 inhibitors used clinically. We demonstrate the value of SLFN11 expression and homologous recombination (HR) deficiency (HRD) as predictive biomarkers of response to exatecan. We also show that exatecan kills cancer cells synergistically with the clinical ATR inhibitor ceralasertib (AZD6738). To establish the translational potential of this combination, we tested CBX-12, a clinically developed pH-sensitive peptide-exatecan conjugate that selectively targets cancer cells and is currently in clinical trials. The combination of CBX-12 with ceralasertib significantly suppressed tumor growth in mouse xenografts. Collectively, our results demonstrate the potency of exatecan as a TOP1 inhibitor and its clinical potential in combination with ATR inhibitors, using SLFN11 and HRD as predictive biomarkers.
Collapse
Affiliation(s)
- Ukhyun Jo
- Developmental Therapeutics Branch and Laboratory of Molecular Pharmacology, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Yasuhisa Murai
- Developmental Therapeutics Branch and Laboratory of Molecular Pharmacology, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
- Department of Gastroenterology and Hematology, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Keli K. Agama
- Developmental Therapeutics Branch and Laboratory of Molecular Pharmacology, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Yilun Sun
- Developmental Therapeutics Branch and Laboratory of Molecular Pharmacology, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Liton Kumar Saha
- Developmental Therapeutics Branch and Laboratory of Molecular Pharmacology, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Xi Yang
- Developmental Therapeutics Branch and Laboratory of Molecular Pharmacology, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Yasuhiro Arakawa
- Developmental Therapeutics Branch and Laboratory of Molecular Pharmacology, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | | | - Kelli Jones
- Cybrexa Therapeutics, New Haven, CT 06511, USA
| | | | - Ranjini K. Sundaram
- Department of Therapeutic Radiology, Yale School of Medicine, New Haven, CT 06511, USA
| | - Jinny Van Doorn
- Department of Therapeutic Radiology, Yale School of Medicine, New Haven, CT 06511, USA
| | - Juan C. Vasquez
- Department of Pediatrics, Yale School of Medicine, New Haven, CT 06511, USA
| | - Ranjit S. Bindra
- Department of Therapeutic Radiology, Yale School of Medicine, New Haven, CT 06511, USA
| | - Woo Suk Choi
- Laboratory of Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD 20892, USA
| | - Yves Pommier
- Developmental Therapeutics Branch and Laboratory of Molecular Pharmacology, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| |
Collapse
|
36
|
Di Z, Lu X, Zhao J, Jaklenec A, Zhao Y, Langer R, Li L. Mild Acidosis‐Directed Signal Amplification in Tumor Microenvironment via Spatioselective Recruitment of DNA Amplifiers. Angew Chem Int Ed Engl 2022. [DOI: 10.1002/ange.202205436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
- Zhenghan Di
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience National Center for Nanoscience and Technology Beijing 100190 China
- College of Materials Science and Optoelectronic Technology University of Chinese Academy of Sciences Beijing 100049 China
| | - Xueguang Lu
- Key Laboratory of Colloid, Interface and Chemical Thermodynamics Institute of Chemistry Chinese Academy of Sciences Beijing 100190 China
- David H. Koch Institute for Integrative Cancer Research Massachusetts Institute of Technology Cambridge MA 02139 USA
| | - Jian Zhao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience National Center for Nanoscience and Technology Beijing 100190 China
- College of Materials Science and Optoelectronic Technology University of Chinese Academy of Sciences Beijing 100049 China
| | - Ana Jaklenec
- David H. Koch Institute for Integrative Cancer Research Massachusetts Institute of Technology Cambridge MA 02139 USA
| | - Yuliang Zhao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience National Center for Nanoscience and Technology Beijing 100190 China
| | - Robert Langer
- David H. Koch Institute for Integrative Cancer Research Massachusetts Institute of Technology Cambridge MA 02139 USA
| | - Lele Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience National Center for Nanoscience and Technology Beijing 100190 China
- College of Materials Science and Optoelectronic Technology University of Chinese Academy of Sciences Beijing 100049 China
| |
Collapse
|
37
|
Ataka K, Drauschke J, Stulberg V, Koksch B, Heberle J. pH-induced insertion of pHLIP into a lipid bilayer: In-situ SEIRAS characterization of a folding intermediate at neutral pH. BIOCHIMICA ET BIOPHYSICA ACTA. BIOMEMBRANES 2022; 1864:183873. [PMID: 35104491 DOI: 10.1016/j.bbamem.2022.183873] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 11/22/2021] [Accepted: 01/20/2022] [Indexed: 06/14/2023]
Abstract
The pH low insertion peptide (pHLIP) is a pH-sensitive cell penetrating peptide that transforms from an unstructured coil on the membrane surface at pH > 7, to a transmembrane (TM) α-helix at pH < 5. By exploiting this unique property, pHLIP attracts interest as a potential tool for drug delivery and visualisation of acidic tissues produced by various maladies such as cancer, inflammation, hypoxia etc. Even though the structures of initial and end states of pHLIP insertion have been widely accepted, the intermediate structures in between these two states are less clear. Here, we have applied in situ Surface-Enhanced Infrared Absorption spectroscopy to examine the pH-induced insertion and folding processes of pHLIP into a solid-supported lipid bilayer. We show that formation of partially helical structure already takes place at pH only slightly below 7.0, but with the helical axis parallel to the membrane surface. The peptide starts to reorientate its helix from horizontal to vertical direction, accompanied by the insertion into the TM region at pH < 6.2. Further insertion into the TM region of the peptide results in an increase of inherent α-helical structure and complete secondary structure formation at pH 5.3. Analysis of the changes of the carboxylate vibrational bands upon pH titration shows two distinctive groups of aspartates and glutamates with pKa values of 4.5 and 6.3, respectively. Comparison to the amide bands of the peptide backbone suggests that the latter Asp/Glu groups are directly involved in the conformational changes of pHLIP in the respective intermediate states.
Collapse
Affiliation(s)
- Kenichi Ataka
- Experimental Molecular Biophysics, Department of Physics, Freie Universität Berlin, Arnimallee 14, 14195 Berlin, Germany.
| | - Janina Drauschke
- Experimental Molecular Biophysics, Department of Physics, Freie Universität Berlin, Arnimallee 14, 14195 Berlin, Germany
| | - Valentina Stulberg
- Institute for Chemistry and Biochemistry, Department of Biology, Chemistry, Pharmacy, Freie Universität Berlin, Arnimallee 20, 14195, Berlin, Germany
| | - Beate Koksch
- Institute for Chemistry and Biochemistry, Department of Biology, Chemistry, Pharmacy, Freie Universität Berlin, Arnimallee 20, 14195, Berlin, Germany
| | - Joachim Heberle
- Experimental Molecular Biophysics, Department of Physics, Freie Universität Berlin, Arnimallee 14, 14195 Berlin, Germany.
| |
Collapse
|
38
|
Bauer D, Visca H, Weerakkody A, Carter LM, Samuels Z, Kaminsky S, Andreev OA, Reshetnyak YK, Lewis JS. PET Imaging of Acidic Tumor Environment With 89Zr-labeled pHLIP Probes. Front Oncol 2022; 12:882541. [PMID: 35664740 PMCID: PMC9160799 DOI: 10.3389/fonc.2022.882541] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 04/19/2022] [Indexed: 11/13/2022] Open
Abstract
Acidosis of the tumor microenvironment is a hallmark of tumor progression and has emerged as an essential biomarker for cancer diagnosis, prognosis, and evaluation of treatment response. A tool for quantitatively visualizing the acidic tumor environment could significantly advance our understanding of the behavior of aggressive tumors, improving patient management and outcomes. 89Zr-labeled pH-low insertion peptides (pHLIP) are a class of radiopharmaceutical imaging probes for the in vivo analysis of acidic tumor microenvironments via positron emission tomography (PET). Their unique structure allows them to sense and target acidic cancer cells. In contrast to traditional molecular imaging agents, pHLIP's mechanism of action is pH-dependent and does not rely on the presence of tumor-specific molecular markers. In this study, one promising acidity-imaging PET probe ([89Zr]Zr-DFO-Cys-Var3) was identified as a candidate for clinical translation.
Collapse
Affiliation(s)
- David Bauer
- Department of Radiology and the Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Hannah Visca
- Department of Physics, University of Rhode Island, Kingston, RI, United States
| | - Anuradha Weerakkody
- Department of Physics, University of Rhode Island, Kingston, RI, United States
| | - Lukas M. Carter
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Zachary Samuels
- Department of Radiology and the Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Spencer Kaminsky
- Department of Radiology and the Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Oleg A. Andreev
- Department of Physics, University of Rhode Island, Kingston, RI, United States
| | - Yana K. Reshetnyak
- Department of Physics, University of Rhode Island, Kingston, RI, United States
| | - Jason S. Lewis
- Department of Radiology and the Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, United States
- Department of Radiology, Weill Cornell Medical College, New York, NY, United States
- Department of Pharmacology Program, Weill Cornell Medical College, New York, NY, United States
| |
Collapse
|
39
|
Sikorski EL, Wehr J, Ferraro NJ, Rizzo SM, Pires MM, Thévenin D. Selective Display of a Chemoattractant Agonist on Cancer Cells Activates the Formyl Peptide Receptor 1 on Immune Cells. Chembiochem 2022; 23:e202100521. [PMID: 35199442 PMCID: PMC9035110 DOI: 10.1002/cbic.202100521] [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: 09/29/2021] [Revised: 01/26/2022] [Indexed: 11/11/2022]
Abstract
Current immunotherapeutics often work by directing components of the immune system to recognize biomarkers on the surface of cancer cells to generate an immune response. However, variable changes in biomarker distribution and expression can result in inconsistent patient response. The development of a more universal tumor-homing strategy has the potential to improve selectivity and extend therapy to cancers with decreased expression or absence of specific biomarkers. Here, we designed a bifunctional agent that exploits the inherent acidic microenvironment of most solid tumors to selectively graft the surface of cancer cells with a formyl peptide receptor ligand (FPRL). Our approach is based on the pH(Low) insertion peptide (pHLIP), a unique peptide that selectively targets tumors in vivo by anchoring to cancer cells in a pH-dependent manner. We establish that selectively remodeling cancer cells with a pHLIP-based FPRL activates formyl peptide receptors on recruited immune cells, potentially initiating an immune response towards tumors.
Collapse
Affiliation(s)
- Eden L. Sikorski
- Department of Chemistry, Lehigh University. Bethlehem, Pennsylvania 18015, United States
| | - Janessa Wehr
- Department of Chemistry, Lehigh University. Bethlehem, Pennsylvania 18015, United States
| | - Noel J. Ferraro
- Department of Chemistry, University of Virginia, Charlottesville, Virginia 22904, United States
| | - Sophia M. Rizzo
- Department of Chemistry, Lehigh University. Bethlehem, Pennsylvania 18015, United States
| | - Marcos M. Pires
- Department of Chemistry, University of Virginia, Charlottesville, Virginia 22904, United States
| | - Damien Thévenin
- Department of Chemistry, Lehigh University. Bethlehem, Pennsylvania 18015, United States
| |
Collapse
|
40
|
Lee PS, MacDonald KG, Massi E, Chew PV, Bee C, Perkins P, Chau B, Thudium K, Lohre J, Nandi P, Deyanova EG, Barman I, Gudmundsson O, Dollinger G, Sproul T, Engelhardt JJ, Strop P, Rajpal A. Improved therapeutic index of an acidic pH-selective antibody. MAbs 2022; 14:2024642. [PMID: 35192429 PMCID: PMC8865267 DOI: 10.1080/19420862.2021.2024642] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Although therapeutically efficacious, ipilimumab can exhibit dose-limiting toxicity that prevents maximal efficacious clinical outcomes and can lead to discontinuation of treatment. We hypothesized that an acidic pH-selective ipilimumab (pH Ipi), which preferentially and reversibly targets the acidic tumor microenvironment over the neutral periphery, may have a more favorable therapeutic index. While ipilimumab has pH-independent CTLA-4 affinity, pH Ipi variants have been engineered to have up to 50-fold enhanced affinity to CTLA-4 at pH 6.0 compared to pH 7.4. In hCTLA-4 knock-in mice, these variants have maintained anti-tumor activity and reduced peripheral activation, a surrogate marker for toxicity. pH-sensitive therapeutic antibodies may be a differentiating paradigm and a novel modality for enhanced tumor targeting and improved safety profiles.
Collapse
Affiliation(s)
- Peter S Lee
- Discovery Biotherapeutics, Bristol Myers Squibb, Redwood City, CA, USA
| | | | - Evan Massi
- Immuno-Oncology Research, Bristol Myers Squibb, Redwood City, CA, USA
| | - Pamela V Chew
- Oncology Biology, Gilead Sciences, Foster City, CA, USA
| | - Christine Bee
- Discovery Biology, Frontier Medicines, South San Francisco, CA, USA
| | - Padma Perkins
- Immuno-Oncology Research, Bristol Myers Squibb, Redwood City, CA, USA
| | - Bryant Chau
- Kyverna, Synthetic Biology, Emeryville, CA, USA
| | - Kent Thudium
- Immuno-Oncology Research, Bristol Myers Squibb, Redwood City, CA, USA
| | - Jack Lohre
- In Vivo Pharmacology, Bristol Myers Squibb, Redwood City, CA, USA
| | - Pradyot Nandi
- Pharmaceutical Candidate Optimization, Bristol Myers Squibb, Lawrenceville, NJ, USA
| | - Ekaterina G Deyanova
- Pharmaceutical Candidate Optimization, Bristol Myers Squibb, Lawrenceville, NJ, USA
| | - Ishita Barman
- Therapeutic Discovery, 3T Biosciences, South San Francisco, CA, USA
| | - Olafur Gudmundsson
- Pharmaceutical Candidate Optimization, Bristol Myers Squibb, Lawrenceville, NJ, USA
| | - Gavin Dollinger
- Discovery Biotherapeutics, Bristol Myers Squibb, Redwood City, CA, USA
| | - Tim Sproul
- In Vivo Pharmacology, UNITY Biotechnology, South San Francisco, CA, USA
| | | | - Pavel Strop
- Biologics Discovery, Tallac Therapeutics, Burlingame, CA, USA
| | - Arvind Rajpal
- Large Molecule Drug Discovery, Genentech Research and Early Development, South San Francisco, CA, USA
| |
Collapse
|
41
|
Wei X, Zhao H, Huang G, Liu J, He W, Huang Q. ES-MION-Based Dual-Modality PET/MRI Probes for Acidic Tumor Microenvironment Imaging. ACS OMEGA 2022; 7:3442-3451. [PMID: 35128253 PMCID: PMC8811892 DOI: 10.1021/acsomega.1c05815] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Accepted: 12/31/2021] [Indexed: 06/14/2023]
Abstract
Among all characteristics of the tumor microenvironment (TME), which are caused by abnormal proliferation of solid tumors, extracellular acidity is an important indicator for malignancy grading. pH-low insertion peptides (pHLIPs) are adopted to discern the acidic TME. To date, different imaging agents including fluorescent, positron emission tomography (PET), single photon emission computed tomography (SPECT), and magnetic resonance (MR) contrast agents with pHLIPs to target the acidic TME have been used to image various tumor models successfully. In this article, a PET/MRI dual-modality probe, based on extremely small magnetic iron oxide nanoparticles (ES-MIONs) with pHLIPs as a targeting unit, was proposed for the first time. In the phantom study, the probe showed relatively high r 1 relaxivity (r 1 = 1.03 mM-1 s-1), indicating that it could be used as a T1-weighted MR contrast agent. The 68Ga-radiolabeled probe was further studied in vitro and in vivo to evaluate pHLIP targeting efficacy and feasibility for PET/MRI. PET with intratumoral injection and T1-weighted MRI with intravenous injection both showed pHLIP-specific delivery of the probe. Therefore, we successfully designed and developed a radiolabeled ES-MION-based dual-modality PET/MRI agent to target the acidic tumor microenvironment. Although the accumulation of the probe in tumors with intravenous injection was not high enough to exhibit signals in the PET imaging study, our study still provides further insights into the ES-MION-based PET/MRI strategy.
Collapse
Affiliation(s)
- Xiuyan Wei
- Medical
Chemistry and Bioinformatics Center, College of Basic Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Haitao Zhao
- Department
of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji
Hospital, School of Medicine, Shanghai Jiao
Tong University, Shanghai 200127, China
| | - Gang Huang
- Shanghai
Key Laboratory of Molecular Imaging, Shanghai
University of Medicine and Health Sciences, Shanghai 201318, China
| | - Jianhua Liu
- Medical
Chemistry and Bioinformatics Center, College of Basic Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Weina He
- Medical
Chemistry and Bioinformatics Center, College of Basic Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Qingqing Huang
- Shanghai
Key Laboratory of Molecular Imaging, Shanghai
University of Medicine and Health Sciences, Shanghai 201318, China
| |
Collapse
|
42
|
Ding GB, Zhu C, Wang Q, Cao H, Li BC, Yang P, Stauber RH, Nie G, Li Z. Molecularly engineered tumor acidity-responsive plant toxin gelonin for safe and efficient cancer therapy. Bioact Mater 2022; 18:42-55. [PMID: 35387163 PMCID: PMC8961304 DOI: 10.1016/j.bioactmat.2022.02.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 01/17/2022] [Accepted: 02/07/2022] [Indexed: 12/11/2022] Open
Abstract
Due to the unsatisfactory therapeutic efficacy and inexorable side effects of small molecule antineoplastic agents, extensive efforts have been devoted to the development of more potent macromolecular agents with high specificity. Gelonin is a plant-derived protein toxin that exhibits robust antitumor effect via inactivating ribosomes and inhibiting protein synthesis. Nonetheless, its poor internalization ability to tumor cells has compromised the therapeutic promise of gelonin. In this study, a tumor acidity-responsive intracellular protein delivery system ─ functional gelonin (Trx-pHLIP-Gelonin, TpG) composed of a thioredoxin (Trx) tag, a pH low insertion peptide (pHLIP) and gelonin, was designed and obtained by genetic recombination technique for the first time. TpG could effectively enter into tumor cells under weakly acidic conditions and markedly suppress tumor cell proliferation via triggering cell apoptosis and inhibiting protein synthesis. Most importantly, treatment by intravenous injection into subcutaneous SKOV3 solid tumors in a mouse model showed that TpG was much more effective than gelonin in curtailing tumor growth rates with negligible toxicity. Collectively, our present work suggests that the tumor acidity-targeted delivery manner endowed by pHLIP offers a new avenue for efficient delivery of other bioactive substances to acidic diseased tissues. A pH-responsive gelonin delivery platform — TpG was molecularly engineered. TpG exhibited good thermal stability and excellent serum stability. TpG enabled an efficient intracellular translocation of gelonin at pH 6.5. TpG exerted pronounced anti-proliferative effect via inducing massive apoptosis. TpG significantly delayed tumor growth with favorable in vivo biosafety profile.
Collapse
|
43
|
Chen SY, Xu XX, Li X, Yi NB, Li SZ, Xiang XC, Cheng DB, Sun T. Recent advances in the intracellular delivery of macromolecule therapeutics. Biomater Sci 2022; 10:6642-6655. [DOI: 10.1039/d2bm01348g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
This review summarizes the uptake pathway of intracellular delivery vehicles for macromolecule therapeutics, and provides in-depth discussions and prospects about intracellular delivery of macromolecule therapeutics.
Collapse
Affiliation(s)
- Si-Yi Chen
- School of Chemistry, Chemical Engineering & Life Science, Wuhan University of Technology, No. 122 Luoshi Road, Wuhan, 430070, PR China
| | - Xiao-Xue Xu
- School of Chemistry, Chemical Engineering & Life Science, Wuhan University of Technology, No. 122 Luoshi Road, Wuhan, 430070, PR China
| | - Xin Li
- School of Chemistry, Chemical Engineering & Life Science, Wuhan University of Technology, No. 122 Luoshi Road, Wuhan, 430070, PR China
| | - Ning-Bo Yi
- School of Chemistry, Chemical Engineering & Life Science, Wuhan University of Technology, No. 122 Luoshi Road, Wuhan, 430070, PR China
| | - Shi-Zhuo Li
- School of Chemistry, Chemical Engineering & Life Science, Wuhan University of Technology, No. 122 Luoshi Road, Wuhan, 430070, PR China
| | - Xing-Cheng Xiang
- School of Chemistry, Chemical Engineering & Life Science, Wuhan University of Technology, No. 122 Luoshi Road, Wuhan, 430070, PR China
| | - Dong-Bing Cheng
- School of Chemistry, Chemical Engineering & Life Science, Wuhan University of Technology, No. 122 Luoshi Road, Wuhan, 430070, PR China
| | - Taolei Sun
- School of Chemistry, Chemical Engineering & Life Science, Wuhan University of Technology, No. 122 Luoshi Road, Wuhan, 430070, PR China
| |
Collapse
|
44
|
Sun Y, Hu L, Yang P, Zhang M, Wang X, Xiao H, Qiao C, Wang J, Luo L, Feng J, Zheng Y, Wang Y, Shi Y, Chen G. pH Low Insertion Peptide-Modified Programmed Cell Death-Ligand 1 Potently Suppresses T-Cell Activation Under Acidic Condition. Front Immunol 2021; 12:794226. [PMID: 35003115 PMCID: PMC8733706 DOI: 10.3389/fimmu.2021.794226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 12/01/2021] [Indexed: 11/13/2022] Open
Abstract
Programmed cell death-ligand 1 (PD-L1)/PD-1 axis is critical for maintenance of immune homeostasis by limiting overactivation of effector T-cell responses. The impairment of PD-L1/PD-1 signals play an important role in the pathogenesis of inflammatory diseases, making this pathway an ideal target for novel therapeutics to induce immune tolerance. Given weakly acidic environment as a putative hallmark of inflammation, in this study we designed a new cargo by linking the ectodomain of murine PD-L1 to the N terminus of pHLIPs, a low pH-responding and membrane-insertion peptide, and demonstrated its potent immune-suppressive activity. Specifically, PD-L1-pHLIP spanned the cellular membrane and perfectly recognized its ligand PD-1 in acidic buffer. Immobile PD-L1-pHLIP actively inhibited T-cell proliferation and IFN-γ production. Importantly, soluble PD-L1-pHLIP retained its function to dampen T-cell responses under acidic condition instead of neutral aqueous solution. Overall, these data suggest that PD-L1-pHLIP has potentials to be a novel therapeutic avenue for T-cell-mediated inflammatory diseases.
Collapse
Affiliation(s)
- Ying Sun
- Inner Mongolia Key Lab of Molecular Biology, School of Basic Medical Sciences, Inner Mongolia Medical University, Hohhot, China
- State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and Toxicology, Beijing, China
| | - Linhan Hu
- Inner Mongolia Key Lab of Molecular Biology, School of Basic Medical Sciences, Inner Mongolia Medical University, Hohhot, China
- State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and Toxicology, Beijing, China
| | - Peng Yang
- Inner Mongolia Key Lab of Molecular Biology, School of Basic Medical Sciences, Inner Mongolia Medical University, Hohhot, China
| | - Min Zhang
- State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and Toxicology, Beijing, China
| | - Xinwei Wang
- Inner Mongolia Key Lab of Molecular Biology, School of Basic Medical Sciences, Inner Mongolia Medical University, Hohhot, China
| | - He Xiao
- State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and Toxicology, Beijing, China
| | - Chunxia Qiao
- State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and Toxicology, Beijing, China
| | - Jing Wang
- State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and Toxicology, Beijing, China
| | - Longlong Luo
- State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and Toxicology, Beijing, China
| | - Jiannan Feng
- State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and Toxicology, Beijing, China
| | - Yuanqiang Zheng
- Inner Mongolia Key Lab of Molecular Biology, School of Basic Medical Sciences, Inner Mongolia Medical University, Hohhot, China
| | - Yi Wang
- Department of Hematology, The Fifth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Yanchun Shi
- Inner Mongolia Key Lab of Molecular Biology, School of Basic Medical Sciences, Inner Mongolia Medical University, Hohhot, China
| | - Guojiang Chen
- State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and Toxicology, Beijing, China
| |
Collapse
|
45
|
Berillo D, Yeskendir A, Zharkinbekov Z, Raziyeva K, Saparov A. Peptide-Based Drug Delivery Systems. MEDICINA (KAUNAS, LITHUANIA) 2021; 57:medicina57111209. [PMID: 34833427 PMCID: PMC8617776 DOI: 10.3390/medicina57111209] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 10/29/2021] [Accepted: 11/03/2021] [Indexed: 12/14/2022]
Abstract
Peptide-based drug delivery systems have many advantages when compared to synthetic systems in that they have better biocompatibility, biochemical and biophysical properties, lack of toxicity, controlled molecular weight via solid phase synthesis and purification. Lysosomes, solid lipid nanoparticles, dendrimers, polymeric micelles can be applied by intravenous administration, however they are of artificial nature and thus may induce side effects and possess lack of ability to penetrate the blood-brain barrier. An analysis of nontoxic drug delivery systems and an establishment of prospective trends in the development of drug delivery systems was needed. This review paper summarizes data, mainly from the past 5 years, devoted to the use of peptide-based carriers for delivery of various toxic drugs, mostly anticancer or drugs with limiting bioavailability. Peptide-based drug delivery platforms are utilized as peptide–drug conjugates, injectable biodegradable particles and depots for delivering small molecule pharmaceutical substances (500 Da) and therapeutic proteins. Controlled drug delivery systems that can effectively deliver anticancer and peptide-based drugs leading to accelerated recovery without significant side effects are discussed. Moreover, cell penetrating peptides and their molecular mechanisms as targeting peptides, as well as stimuli responsive (enzyme-responsive and pH-responsive) peptides and peptide-based self-assembly scaffolds are also reviewed.
Collapse
Affiliation(s)
- Dmitriy Berillo
- Department of Pharmaceutical and Toxicological Chemistry, Pharmacognosy and Botany School of Pharmacy, Asfendiyarov Kazakh National Medical University, Almaty 050000, Kazakhstan
- Correspondence: (D.B.); (A.S.)
| | - Adilkhan Yeskendir
- Department of Medicine, School of Medicine, Nazarbayev University, Nur-Sultan 010000, Kazakhstan; (A.Y.); (Z.Z.); (K.R.)
| | - Zharylkasyn Zharkinbekov
- Department of Medicine, School of Medicine, Nazarbayev University, Nur-Sultan 010000, Kazakhstan; (A.Y.); (Z.Z.); (K.R.)
| | - Kamila Raziyeva
- Department of Medicine, School of Medicine, Nazarbayev University, Nur-Sultan 010000, Kazakhstan; (A.Y.); (Z.Z.); (K.R.)
| | - Arman Saparov
- Department of Medicine, School of Medicine, Nazarbayev University, Nur-Sultan 010000, Kazakhstan; (A.Y.); (Z.Z.); (K.R.)
- Correspondence: (D.B.); (A.S.)
| |
Collapse
|
46
|
Otieno SA, Qiang W. Roles of key residues and lipid dynamics reveal pHLIP-membrane interactions at intermediate pH. Biophys J 2021; 120:4649-4662. [PMID: 34624273 PMCID: PMC8595900 DOI: 10.1016/j.bpj.2021.10.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 09/16/2021] [Accepted: 10/01/2021] [Indexed: 11/16/2022] Open
Abstract
The pH-low insertion peptide (pHLIP) and its analogs sense the microenvironmental pH variations in tumorous cells and serve as useful anticancer drug deliveries. The pHLIP binds peripherally to membranes and adopts random coil conformation at the physiological pH. The peptide switches from random coil to α-helical conformation and inserts unidirectionally into membrane bilayers when pH drops below a critical transition value that has been routinely determined by the Trp fluorescence spectroscopy. Recent high-resolution studies using solid-state NMR spectroscopy revealed the presence of thermodynamically stable intermediate states of membrane-associated pHLIP around the fluorescence-based transition pH-value. However, the molecular structural features and their mechanistic roles of these intermediate states in the pH-driven membrane insertion process of pHLIP remain largely unknown. This work utilizes solid-state NMR spectroscopy to explore 1) the mechanistic roles of key proline and arginine residues within the pHLIP sequence at intermediate pH-values, and 2) the changes in lipid dynamics at intermediate pH-values in multiple types of model bilayers with anionic phospholipid and/or cholesterol. Our results demonstrate several molecular structural and dynamics changes at around the transition pH-values, including the isomerization of proline-threonine backbone configuration, breaking of arginine-aspartic acid salt bridge and the formation of arginine-lipid interactions, and a universal decreasing of dynamics in lipid headgroups and alkyl chains. Overall, the outcomes provide important insights on the molecular interactions between pHLIP and membrane bilayers at intermediate pH-values and, therefore, prompt the understanding of pH-driven membrane insertion process of this anticancer drug-delivering peptide.
Collapse
Affiliation(s)
- Sarah A Otieno
- Department of Chemistry, Binghamton University, State University of New York, Binghamton, New York
| | - Wei Qiang
- Department of Chemistry, Binghamton University, State University of New York, Binghamton, New York.
| |
Collapse
|
47
|
Xu W, Wang J, Li Q, Wu C, Wu L, Li K, Li Q, Han Q, Zhu J, Bai Y, Deng J, Lyu J, Wang Z. Cancer cell membrane-coated nanogels as a redox/pH dual-responsive drug carrier for tumor-targeted therapy. J Mater Chem B 2021; 9:8031-8037. [PMID: 34486010 DOI: 10.1039/d1tb00788b] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Nanocarriers have shown great advantages in increasing the efficiency of drug delivery and reducing drug side effects. However, their lack of targeting and on-demand drug release abilities will seriously limit their clinical application. Herein, we report tumor cell membrane coated nanogels (NGs) with redox/pH dual-responsive behavior for enhanced tumor chemotherapy. The cell membrane coating improves the tumor targeting efficiency, and stimuli-responsive drug release enhances the therapeutic effects. These NGs are well dispersed in PBS with an average size of 109.1 ± 5.2 nm and a narrow polydispersity index of 0.12. Both in vitro and in vivo studies indicate that these NGs can responsively release the therapeutic drug DOX under acidic conditions or high GSH concentrations and effectively inhibit tumor growth. Based on the results, this nanogel shows promise as a platform for tumor-targeted chemotherapy for future clinical translation.
Collapse
Affiliation(s)
- Weide Xu
- Research Center of Blood Transfusion Medicine, Ministry of Education Key Laboratory of Laboratory Medicine, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou 310014, China.,School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Jilong Wang
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325000, China
| | - Qinghua Li
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325000, China
| | - Chenghu Wu
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325000, China
| | - Lingling Wu
- Research Center of Blood Transfusion Medicine, Ministry of Education Key Laboratory of Laboratory Medicine, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou 310014, China.,School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325035, China.,Rehabilitation & Sports Medicine Research Institute of Zhejiang Province, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou 310014, China
| | - Kaiqiang Li
- Research Center of Blood Transfusion Medicine, Ministry of Education Key Laboratory of Laboratory Medicine, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou 310014, China.,Rehabilitation & Sports Medicine Research Institute of Zhejiang Province, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou 310014, China
| | - Qin Li
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Qing Han
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Jingjing Zhu
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Yongheng Bai
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Junjie Deng
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325000, China.,Oujiang Laboratory, Wenzhou, Zhejiang 325000, China
| | - Jianxin Lyu
- Research Center of Blood Transfusion Medicine, Ministry of Education Key Laboratory of Laboratory Medicine, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou 310014, China.,School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Zhen Wang
- Research Center of Blood Transfusion Medicine, Ministry of Education Key Laboratory of Laboratory Medicine, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou 310014, China.,School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325035, China
| |
Collapse
|
48
|
Hao L, Rohani N, Zhao RT, Pulver EM, Mak H, Kelada OJ, Ko H, Fleming HE, Gertler FB, Bhatia SN. Microenvironment-triggered multimodal precision diagnostics. NATURE MATERIALS 2021; 20:1440-1448. [PMID: 34267368 DOI: 10.1038/s41563-021-01042-y] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Accepted: 05/26/2021] [Indexed: 05/24/2023]
Abstract
Therapeutic outcomes in oncology may be aided by precision diagnostics that offer early detection, localization and the opportunity to monitor response to therapy. Here, we report a multimodal nanosensor engineered to target tumours through acidosis, respond to proteases in the microenvironment to release urinary reporters and (optionally) carry positron emission tomography probes to enable localization of primary and metastatic cancers in mouse models of colorectal cancer. We present a paradigm wherein this multimodal sensor can be employed longitudinally to assess burden of disease non-invasively, including tumour progression and response to chemotherapy. Specifically, we showed that acidosis-mediated tumour insertion enhanced on-target release of matrix metalloproteinase-responsive reporters in urine. Subsequent on-demand loading of the radiotracer 64Cu allowed pH-dependent tumour visualization, enabling enriched microenvironmental characterization when compared with the conventional metabolic tracer 18F-fluorodeoxyglucose. Through tailored target specificities, this modular platform has the capacity to be engineered as a pan-cancer test that may guide treatment decisions for numerous tumour types.
Collapse
Affiliation(s)
- Liangliang Hao
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Nazanin Rohani
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Renee T Zhao
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Emilia M Pulver
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Howard Mak
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | | | - Henry Ko
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Heather E Fleming
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Howard Hughes Medical Institute, Cambridge, MA, USA
| | - Frank B Gertler
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Sangeeta N Bhatia
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Howard Hughes Medical Institute, Cambridge, MA, USA.
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA.
- Ludwig Center at Massachusetts Institute of Technology's Koch Institute for Integrative Cancer Research, Cambridge, MA, USA.
| |
Collapse
|
49
|
Abstract
The use of PET imaging agents in oncology, cardiovascular disease, and neurodegenerative disease shows the power of this technique in evaluating the molecular and biological characteristics of numerous diseases. These agents provide crucial information for designing therapeutic strategies for individual patients. Novel PET tracers are in continual development and many have potential use in clinical and research settings. This article discusses the potential applications of tracers in diagnostics, the biological characteristics of diseases, the ability to provide prognostic indicators, and using this information to guide treatment strategies including monitoring treatment efficacy in real time to improve outcomes and survival.
Collapse
|
50
|
Moody AS, Dayton PA, Zamboni WC. Imaging methods to evaluate tumor microenvironment factors affecting nanoparticle drug delivery and antitumor response. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2021; 4:382-413. [PMID: 34796317 PMCID: PMC8597952 DOI: 10.20517/cdr.2020.94] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 01/07/2021] [Accepted: 01/28/2021] [Indexed: 11/24/2022]
Abstract
Standard small molecule and nanoparticulate chemotherapies are used for cancer treatment; however, their effectiveness remains highly variable. One reason for this variable response is hypothesized to be due to nonspecific drug distribution and heterogeneity of the tumor microenvironment, which affect tumor delivery of the agents. Nanoparticle drugs have many theoretical advantages, but due to variability in tumor microenvironment (TME) factors, the overall drug delivery to tumors and associated antitumor response are low. The nanotechnology field would greatly benefit from a thorough analysis of the TME factors that create these physiological barriers to tumor delivery and treatment in preclinical models and in patients. Thus, there is a need to develop methods that can be used to reveal the content of the TME, determine how these TME factors affect drug delivery, and modulate TME factors to increase the tumor delivery and efficacy of nanoparticles. In this review, we will discuss TME factors involved in drug delivery, and how biomedical imaging tools can be used to evaluate tumor barriers and predict drug delivery to tumors and antitumor response.
Collapse
Affiliation(s)
- Amber S. Moody
- UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA
- UNC Lineberger Comprehensive Cancer Center, Chapel Hill, NC 27599, USA
- Carolina Institute for Nanomedicine, Chapel Hill, NC 27599, USA
- Joint Department of Biomedical Engineering, University of North Carolina and North Carolina State University, Chapel Hill, NC 27599, USA
| | - Paul A. Dayton
- UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA
- UNC Lineberger Comprehensive Cancer Center, Chapel Hill, NC 27599, USA
- Joint Department of Biomedical Engineering, University of North Carolina and North Carolina State University, Chapel Hill, NC 27599, USA
| | - William C. Zamboni
- UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA
- UNC Lineberger Comprehensive Cancer Center, Chapel Hill, NC 27599, USA
- Carolina Institute for Nanomedicine, Chapel Hill, NC 27599, USA
| |
Collapse
|