1
|
Bao K, Yoon JS, Ahn S, Lee JH, Cross CJ, Jeong MY, Frangioni JV, Choi HS. A robotic system for automated chemical synthesis of therapeutic agents. MATERIALS ADVANCES 2024; 5:5290-5297. [PMID: 38894709 PMCID: PMC11181120 DOI: 10.1039/d4ma00099d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 04/29/2024] [Indexed: 06/21/2024]
Abstract
The development of novel compounds for tissue-specific targeting and imaging is often impeded by a lack of lead compounds and the availability of reliable chemistry. Automated chemical synthesis systems provide a potential solution by enabling reliable, repeated access to large compound libraries for screening. Here we report an integrated solid-phase combinatorial chemistry system created using commercial and customized robots. Our goal is to optimize reaction parameters, such as varying temperature, shaking, microwave irradiation, aspirating and dispensing large-sized solid beads, and handling different washing solvents for separation and purification. This automated system accommodates diverse chemical reactions such as peptide synthesis and conventional coupling reactions. To confirm its functionality and reproducibility, 20 nerve-specific contrast agents for biomedical imaging were systematically and repeatedly synthesized and compared to other nerve-targeted agents using molecular fingerprinting and Uniform Manifold Approximation and Projection, which lays the foundation for creating reliable and reproductive chemical libraries in bioimaging and nanomedicine.
Collapse
Affiliation(s)
- Kai Bao
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School Boston MA 02114 USA
- Center for Molecular Imaging, Department of Medicine, Beth Israel Deaconess Medical Center Boston MA 02215 USA
| | - Jong Seo Yoon
- Center for Molecular Imaging, Department of Medicine, Beth Israel Deaconess Medical Center Boston MA 02215 USA
| | - Sung Ahn
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School Boston MA 02114 USA
| | - Jeong Heon Lee
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School Boston MA 02114 USA
- Center for Molecular Imaging, Department of Medicine, Beth Israel Deaconess Medical Center Boston MA 02215 USA
| | - Conor J Cross
- Center for Molecular Imaging, Department of Medicine, Beth Israel Deaconess Medical Center Boston MA 02215 USA
| | - Myung Yung Jeong
- Center for Molecular Imaging, Department of Medicine, Beth Israel Deaconess Medical Center Boston MA 02215 USA
- Department of Cogno-Mechatronics Engineering, Pusan National University Busan 46241 South Korea
| | - John V Frangioni
- Center for Molecular Imaging, Department of Medicine, Beth Israel Deaconess Medical Center Boston MA 02215 USA
- Curadel, LLC Natick MA 01760 USA
| | - Hak Soo Choi
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School Boston MA 02114 USA
- Center for Molecular Imaging, Department of Medicine, Beth Israel Deaconess Medical Center Boston MA 02215 USA
| |
Collapse
|
2
|
Pun MD, Gallazzi F, Ho KV, Watkinson L, Carmack TL, Iweha E, Li L, Anderson CJ. Albumin-Binding Lutetium-177-Labeled LLP2A Derivatives as Theranostics for Melanoma. Mol Pharm 2024; 21:2960-2969. [PMID: 38680059 DOI: 10.1021/acs.molpharmaceut.4c00095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/01/2024]
Abstract
Very late antigen-4 (VLA-4) is a transmembrane integrin protein that is highly expressed in aggressive forms of metastatic melanoma. A small-molecule peptidomimetic, LLP2A, was found to have a low pM affinity binding to VLA-4. Because LLP2A itself does not inhibit cancer cell proliferation and survival, it is an ideal candidate for the imaging and delivery of therapeutic payloads. An analog of [177Lu]Lu-labeled-LLP2A was previously investigated as a therapeutic agent in melanoma tumor-bearing mice, resulting in only a modest improvement in tumor growth inhibition, likely due to rapid clearance of the agent from the tumor. To improve the pharmacokinetic profile, DOTAGA-PEG4-LLP2A with a 4-(p-iodophenyl)butyric acid (pIBA) albumin binding moiety was synthesized. We demonstrate the feasibility of this albumin binding strategy by comparing in vitro cell binding assays and in vivo biodistribution performance of [177Lu]Lu-DOTAGA-PEG4-LLP2A ([177Lu]Lu-1) to the albumin binding [177Lu]Lu-DOTAGA-pIBA-PEG4-LLP2A ([177Lu]Lu-2). In vitro cell binding assay results for [177Lu]Lu-1 and [177Lu]Lu-2 showed Kd values of 0.40 ± 0.07 and 1.75 ± 0.40 nM, with similar Bmax values of 200 ± 6 and 315 ± 15 fmol/mg, respectively. In vivo biodistribution data for both tracers exhibited specific uptake in the tumor, spleen, thymus, and bone due to endogenous expression of VLA-4. Compound [177Lu]Lu-2 exhibited a much longer blood circulation time compared to [177Lu]Lu-1. The tumor uptake for [177Lu]Lu-1 was highest at 1 h (∼15%ID/g) and that for [177Lu]Lu-2 was highest at 4 h (∼23%ID/g). Significant clearance of [177Lu]Lu-1 from the tumor occurs at 24 h (<5%ID/g) while[177Lu]Lu-2 is retained for greater than 96 h (∼10%ID/g). An efficacy study showed that melanoma tumor-bearing mice receiving compound [177Lu]Lu-2 given in two fractions (2 × 14.8 MBq, 14 days apart) had a greater median survival time than mice administered a single 29.6 MBq dose of compound [177Lu]Lu-1, while a single 29.6 MBq dose of [177Lu]Lu-2 imparted hematopoietic toxicity. The in vitro and in vivo data show addition of pIBA to [177Lu]Lu-DOTAGA-PEG4-LLP2A slows blood clearance for a higher tumor uptake, and there is potential of [177Lu]Lu-2 as a theranostic in fractionated administered doses.
Collapse
Affiliation(s)
- Michael D Pun
- Department of Chemistry, University of Missouri, Columbia, Missouri 65211, United States
- Molecular Imaging and Theranostics Center, University of Missouri, Columbia, Missouri 65211, United States
| | - Fabio Gallazzi
- Department of Chemistry, University of Missouri, Columbia, Missouri 65211, United States
- Molecular Interactions Core, University of Missouri, Columbia, Missouri 65211, United States
| | - Khanh-Van Ho
- Department of Chemistry, University of Missouri, Columbia, Missouri 65211, United States
- Molecular Imaging and Theranostics Center, University of Missouri, Columbia, Missouri 65211, United States
| | - Lisa Watkinson
- Molecular Imaging and Theranostics Center, University of Missouri, Columbia, Missouri 65211, United States
- Research Division, Harry S. Truman Memorial Veterans' Hospital, Columbia, Missouri 65211, United States
- University of Missouri Research Reactor Center, University of Missouri, Columbia, Missouri 65211, United States
| | - Terry L Carmack
- Molecular Imaging and Theranostics Center, University of Missouri, Columbia, Missouri 65211, United States
- Research Division, Harry S. Truman Memorial Veterans' Hospital, Columbia, Missouri 65211, United States
- University of Missouri Research Reactor Center, University of Missouri, Columbia, Missouri 65211, United States
| | - Ejike Iweha
- Department of Chemistry, University of Missouri, Columbia, Missouri 65211, United States
- Molecular Imaging and Theranostics Center, University of Missouri, Columbia, Missouri 65211, United States
| | - Longbo Li
- Department of Chemistry, University of Missouri, Columbia, Missouri 65211, United States
- Molecular Imaging and Theranostics Center, University of Missouri, Columbia, Missouri 65211, United States
| | - Carolyn J Anderson
- Department of Chemistry, University of Missouri, Columbia, Missouri 65211, United States
- Molecular Imaging and Theranostics Center, University of Missouri, Columbia, Missouri 65211, United States
- Department of Radiology, University of Missouri, Columbia, Missouri 65212, United States
- Ellis Fischel Cancer Center, University of Missouri, Columbia, Missouri 65212, United States
| |
Collapse
|
3
|
Kim M, Jung KH, Kim JL, Koo HJ, Jung HJ, Lee H, Lee KH. Imaging Very Late Antigen-4 on MOLT4 Leukemia Tumors with Cysteine Site-Specific 89Zr-Labeled Natalizumab Immuno-Positron Emission Tomography. Mol Pharm 2024; 21:1353-1363. [PMID: 38282332 DOI: 10.1021/acs.molpharmaceut.3c01024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2024]
Abstract
Very late antigen-4 (VLA4; CD49d) is a promising immune therapy target in treatment-resistant leukemia and multiple myeloma, and there is growing interest in repurposing the humanized monoclonal antibody (Ab), natalizumab, for this purpose. Positron emission tomography with radiolabeled Abs (immuno-PET) could facilitate this effort by providing information on natalizumab's in vivo pharmacokinetic and target delivery properties. In this study, we labeled natalizumab with 89Zr specifically on sulfhydryl moieties via maleimide-deferoxamine conjugation. High VLA4-expressing MOLT4 human T cell acute lymphoblastic leukemia cells showed specific 89Zr-natalizumab binding that was markedly blocked by excess Ab. In nude mice bearing MOLT4 tumors, 89Zr-natalizumab PET showed high-contrast tumor uptake at 7 days postinjection. Biodistribution studies confirmed that uptake was the highest in MOLT4 tumors (2.22 ± 0.41%ID/g) and the liver (2.33 ± 0.76%ID/g), followed by the spleen (1.51 ± 0.42%ID/g), while blood activity was lower at 1.12 ± 0.21%ID/g. VLA4-specific targeting in vivo was confirmed by a 58.1% suppression of tumor uptake (0.93 ± 0.15%ID/g) when excess Ab was injected 1 h earlier. In cultured MOLT4 cells, short-term 3 day exposure to the proteasome inhibitor bortezomib (BTZ) did not affect the α4 integrin level, but BTZ-resistant cells that survived the treatment showed increased α4 integrin expression. When the effects of BTZ treatment were tested in mice, there was no change of the α4 integrin level or 89Zr-natalizumab uptake in MOLT4 leukemia tumors, which underscores the complexity of tumor VLA4 regulation in vivo. In conclusion, 89Zr-natalizumab PET may be useful for noninvasive monitoring of tumor VLA4 and may assist in a more rational application of Ab-based therapies for hematologic malignancies.
Collapse
Affiliation(s)
- Mina Kim
- Department of Nuclear Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul 06355, Korea
| | - Kyung-Ho Jung
- Department of Nuclear Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul 06355, Korea
| | - Jung Lim Kim
- Department of Nuclear Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea
| | - Hyun-Jung Koo
- Department of Nuclear Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea
| | - Hye Jin Jung
- Department of Nuclear Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea
| | - Hyunjong Lee
- Department of Nuclear Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea
| | - Kyung-Han Lee
- Department of Nuclear Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul 06355, Korea
| |
Collapse
|
4
|
Daneste H, Mohammadzadeh Boukani L, Ramezani N, Asadi F, Zaidan HK, Sadeghzade A, Ehsannia M, Azarashk A, Gholizadeh N. Combination therapy along with mesenchymal stem cells in wound healing; the state of the art. Adv Med Sci 2023; 68:441-449. [PMID: 37924749 DOI: 10.1016/j.advms.2023.10.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 06/23/2023] [Accepted: 10/17/2023] [Indexed: 11/06/2023]
Abstract
Mesenchymal stem cells (MSCs) are being increasingly used in various therapeutic applications including skin tissue repair and wound healing. The positive effects of the MSCs therapy are largely elicited by immunomodulation, increasing angiogenesis, supporting extracellular matrix (ECM) and thus favoring skin structure. However, the therapeutic competences of MSC-based therapies are somewhat hindered by their apparent modest clinical merits, conferring the need for methods that would rise the efficacy of such therapies. A plethora of reports have shown that therapeutic properties of MSCs could be enhanced with other strategies and compounds like biomaterial and platelet-rich plasma (PRP) to target key possessions of MSCs and properties of adjacent tissues concurrently. Manipulation of cellular stress-response mechanisms to improve cell resistance to oxidative stress prior to or during MSC injection could also improve therapeutic efficacy of MSCs. In the current review, we shed light on the recent advances in MSCs combination therapy with other ingredients and procedures to sustain MSCs-mediated effects in wound healing.
Collapse
Affiliation(s)
- Hossein Daneste
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Narges Ramezani
- Department of Biology, Damghan Branch, Islamic Azad University, Damghan, Iran
| | - Fatemeh Asadi
- Department of Genetics, Izeh Branch, Islamic Azad University, Izeh, Iran
| | - Haider Kamil Zaidan
- Department of Medical Laboratories Techniques, Al-Mustaqbal University College, Hillah, Babylon, Iraq
| | - Azita Sadeghzade
- Department of Oral and Maxillofacial Medicine, School of Dentistry, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Maedeh Ehsannia
- Faculty of Basic Sciences, Islamic Azad University, Tehran East Branch, Tehran, Iran
| | - Ali Azarashk
- Department of Pharmaceutics, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran.
| | - Nasim Gholizadeh
- Department of Dermatology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.
| |
Collapse
|
5
|
Zhu Q, Barnes CE, Mannes PZ, Latoche JD, Day KE, Nedrow JR, Novelli EM, Anderson CJ, Tavakoli S. Targeted imaging of very late antigen-4 for noninvasive assessment of lung inflammation-fibrosis axis. EJNMMI Res 2023; 13:55. [PMID: 37273103 PMCID: PMC10240482 DOI: 10.1186/s13550-023-01006-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 06/01/2023] [Indexed: 06/06/2023] Open
Abstract
BACKGROUND The lack of noninvasive methods for assessment of dysregulated inflammation as a major driver of fibrosis (i.e., inflammation-fibrosis axis) has been a major challenge to precision management of fibrotic lung diseases. Here, we determined the potential of very late antigen-4 (VLA-4)-targeted positron emission tomography (PET) to detect inflammation in a mouse model of bleomycin-induced fibrotic lung injury. METHOD Single time-point and longitudinal VLA-4-targeted PET was performed using a high-affinity peptidomimetic radiotracer, 64Cu-LLP2A, at weeks 1, 2, and 4 after bleomycin-induced (2.5 units/kg) lung injury in C57BL/6J mice. The severity of fibrosis was determined by measuring the hydroxyproline content of the lungs and expression of markers of extracellular matrix remodeling. Flow cytometry and histology was performed to determine VLA-4 expression across different leukocyte subsets and their spatial distribution. RESULTS Lung uptake of 64Cu-LLP2A was significantly elevated throughout different stages of the progression of bleomycin-induced injury. High lung uptake of 64Cu-LLP2A at week-1 post-bleomycin was a predictor of poor survival over the 4-week follow up, supporting the prognostic potential of 64Cu-LLP2A PET during the early stage of the disease. Additionally, the progressive increase in 64Cu-LLP2A uptake from week-1 to week-4 post-bleomycin correlated with the ultimate extent of lung fibrosis and ECM remodeling. Flow cytometry revealed that LLP2A binding was restricted to leukocytes. A combination of increased expression of VLA-4 by alveolar macrophages and accumulation of VLA-4-expressing interstitial and monocyte-derived macrophages as well as dendritic cells was noted in bleomycin-injured, compared to control, lungs. Histology confirmed the increased expression of VLA-4 in bleomycin-injured lungs, particularly in inflamed and fibrotic regions. CONCLUSIONS VLA-4-targeted PET allows for assessment of the inflammation-fibrosis axis and prediction of disease progression in a murine model. The potential of 64Cu-LLP2A PET for assessment of the inflammation-fibrosis axis in human fibrotic lung diseases needs to be further investigated.
Collapse
Affiliation(s)
- Qin Zhu
- Department of Radiology, University of Pittsburgh, UPMC Presbyterian Hospital, 200 Lothrop Street, Suite E200, Pittsburgh, PA, 15213, USA
| | - Clayton E Barnes
- Department of Radiology, University of Pittsburgh, UPMC Presbyterian Hospital, 200 Lothrop Street, Suite E200, Pittsburgh, PA, 15213, USA
| | - Philip Z Mannes
- Department of Radiology, University of Pittsburgh, UPMC Presbyterian Hospital, 200 Lothrop Street, Suite E200, Pittsburgh, PA, 15213, USA
- Medical Scientist Training Program, University of Pittsburgh, Pittsburgh, PA, USA
| | - Joseph D Latoche
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Kathryn E Day
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jessie R Nedrow
- Department of Radiology, University of Pittsburgh, UPMC Presbyterian Hospital, 200 Lothrop Street, Suite E200, Pittsburgh, PA, 15213, USA
| | - Enrico M Novelli
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Heart, Lung, Blood, and Vascular Medicine Institute, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Carolyn J Anderson
- Department of Chemistry, University of Missouri, Columbia, MO, USA
- Department of Radiology, University of Missouri, Columbia, MO, USA
| | - Sina Tavakoli
- Department of Radiology, University of Pittsburgh, UPMC Presbyterian Hospital, 200 Lothrop Street, Suite E200, Pittsburgh, PA, 15213, USA.
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA.
- Heart, Lung, Blood, and Vascular Medicine Institute, University of Pittsburgh Medical Center, Pittsburgh, PA, USA.
| |
Collapse
|
6
|
Hao D, Liu R, Fernandez TG, Pivetti C, Jackson JE, Kulubya ES, Jiang HJ, Ju HY, Liu WL, Panitch A, Lam KS, Leach JK, Farmer DL, Wang A. A bioactive material with dual integrin-targeting ligands regulates specific endogenous cell adhesion and promotes vascularized bone regeneration in adult and fetal bone defects. Bioact Mater 2023; 20:179-193. [PMID: 35663336 PMCID: PMC9160290 DOI: 10.1016/j.bioactmat.2022.05.027] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Revised: 05/19/2022] [Accepted: 05/19/2022] [Indexed: 12/18/2022] Open
Abstract
Significant progress has been made in designing bone materials capable of directing endogenous cells to promote vascularized bone regeneration. However, current strategies lack regulation of the specific endogenous cell populations for vascularized bone regeneration, thus leading to adverse tissue formation and decreased regenerative efficiency. Here, we engineered a biomaterial to regulate endogenous cell adhesion and promote vascularized bone regeneration. The biomaterial works by presenting two synthetic ligands, LLP2A and LXW7, explicitly targeting integrins α4β1 and αvβ3, respectively, expressed on the surfaces of the cells related to bone formation and vascularization, such as mesenchymal stem cells (MSCs), osteoblasts, endothelial progenitor cells (EPCs), and endothelial cells (ECs). In vitro, the LLP2A/LXW7 modified biomaterial improved the adhesion of MSCs, osteoblasts, EPCs, and ECs via integrin α4β1 and αvβ3, respectively. In an adult rat calvarial bone defect model, the LLP2A/LXW7 modified biomaterial enhanced bone formation and vascularization by synergistically regulating endogenous cells with osteogenic and angiogenic potentials, such as DLX5+ cells, osteocalcin+ cells, CD34+/CD45- cells and CD31+ cells. In a fetal sheep spinal bone defect model, the LLP2A/LXW7 modified biomaterial augmented bone formation and vascularization without any adverse effects. This innovative biomaterial offers an off-the-shelf, easy-to-use, and biologically safe product suitable for vascularized bone regeneration in both fetal and adult disease environments. Two integrin-binding ligands for constructing vascularized bone biomaterial. Extracellular matrix (ECM)-mimicking collagen-based biomaterial with specific integrin binding sites for cell adhesion. Biomaterial regulates adhesion of endogenous stem cells with osteogenic and angiogenic potentials. Biomaterial promotes vascularized bone formation in adult and fetal bone defects without safety issues. An easy-to-make and off-the-shelf biomaterial for treatment of clinical bone diseases.
Collapse
Affiliation(s)
- Dake Hao
- Department of Surgery, School of Medicine, University of California Davis, Sacramento, CA, 95817, United States
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, CA, 95817, United States
| | - Ruiwu Liu
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California Davis, Sacramento, CA, 95817, United States
| | - Tomas Gonzalez Fernandez
- Department of Biomedical Engineering, University of California Davis, Davis, CA, 95616, United States
| | - Christopher Pivetti
- Department of Surgery, School of Medicine, University of California Davis, Sacramento, CA, 95817, United States
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, CA, 95817, United States
| | - Jordan Elizabeth Jackson
- Department of Surgery, School of Medicine, University of California Davis, Sacramento, CA, 95817, United States
| | - Edwin Samuel Kulubya
- Department of Surgery, School of Medicine, University of California Davis, Sacramento, CA, 95817, United States
| | - Hong-Jiang Jiang
- Wendeng Orthopaedic Hospital, No. 1 Fengshan Road, Wendeng, 264400, Shandong, China
| | - Hai-Yang Ju
- Wendeng Orthopaedic Hospital, No. 1 Fengshan Road, Wendeng, 264400, Shandong, China
| | - Wen-Liang Liu
- Wendeng Orthopaedic Hospital, No. 1 Fengshan Road, Wendeng, 264400, Shandong, China
| | - Alyssa Panitch
- Department of Surgery, School of Medicine, University of California Davis, Sacramento, CA, 95817, United States
- Department of Biomedical Engineering, University of California Davis, Davis, CA, 95616, United States
| | - Kit S. Lam
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California Davis, Sacramento, CA, 95817, United States
| | - J. Kent Leach
- Department of Orthopaedic Surgery, School of Medicine, University of California Davis, Sacramento, CA, 95817, United States
| | - Diana L. Farmer
- Department of Surgery, School of Medicine, University of California Davis, Sacramento, CA, 95817, United States
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, CA, 95817, United States
| | - Aijun Wang
- Department of Surgery, School of Medicine, University of California Davis, Sacramento, CA, 95817, United States
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, CA, 95817, United States
- Department of Biomedical Engineering, University of California Davis, Davis, CA, 95616, United States
- Corresponding author. Center for Surgical Bioengineering, Department of Surgery, School of Medicine, University of California, Davis, 4625 2nd Ave., Research II, Suite 3005, Sacramento, CA, 95817, USA.
| |
Collapse
|
7
|
Yan YQ, Wang JQ, Zhang L, Yang PP, Ye XW, Liu C, Hou DY, Lai WJ, Wang J, Zeng XZ, Xu W, Wang L. Localized Instillation Enables In Vivo Screening of Targeting Peptides Using One-Bead One-Compound Technology. ACS NANO 2023; 17:1381-1392. [PMID: 36596220 DOI: 10.1021/acsnano.2c09894] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
The One-Bead One-Compound (OBOC) library screening is an efficient technique for identifying targeting peptides. However, due to the relatively large bead size, it is challenging for the OBOC method to be applied for in vivo screening. Herein, we report an in vivo Localized Instillation Beads library (LIB) screening method to discover targeting peptides with the OBOC technique. Inspired by localized instillation, we constructed a cavity inside of a transplanted tumor of a mouse. Then, the OBOC heptapeptide library was injected and incubated inside the tumor cavity. After an efficient elution process, the retained beads were gathered, from which three MDA-MB-231 tumor-targeting heptapeptides were discovered. It was verified that the best peptide had 1.9-fold higher tumor accumulation than the commonly used targeting peptide RGD in vivo. Finally, two targeting proteins were discovered as potential targets of our targeting peptide to the MDA-MB-231 tumor. The in vivo LIB screening method expands the scope of OBOC peptide screening applications to discover targeting peptides in vivo feasibly and reliably.
Collapse
Affiliation(s)
- Ya-Qiong Yan
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST)No. 11 Beiyitiao, Zhongguancun, Beijing100190, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing100049, P. R. China
| | - Jia-Qi Wang
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST)No. 11 Beiyitiao, Zhongguancun, Beijing100190, China
- Department of Urology, the Fourth Hospital of Harbin Medical University, Heilongjiang Key Laboratory of Scientific Research in Urology, No. 37 Yi-Yuan Street, Nangang District, Harbin, Heilongjiang Province150001, China
- NHC Key Laboratory of Molecular Probes and Targeted Diagnosis and Therapy, Harbin Medical University, Harbin, 150001, China
| | - Lingze Zhang
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST)No. 11 Beiyitiao, Zhongguancun, Beijing100190, China
| | - Pei-Pei Yang
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST)No. 11 Beiyitiao, Zhongguancun, Beijing100190, China
| | - Xin-Wei Ye
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST)No. 11 Beiyitiao, Zhongguancun, Beijing100190, China
| | - Cong Liu
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST)No. 11 Beiyitiao, Zhongguancun, Beijing100190, China
| | - Da-Yong Hou
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST)No. 11 Beiyitiao, Zhongguancun, Beijing100190, China
- Department of Urology, the Fourth Hospital of Harbin Medical University, Heilongjiang Key Laboratory of Scientific Research in Urology, No. 37 Yi-Yuan Street, Nangang District, Harbin, Heilongjiang Province150001, China
- NHC Key Laboratory of Molecular Probes and Targeted Diagnosis and Therapy, Harbin Medical University, Harbin, 150001, China
| | - Wen-Jia Lai
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST)No. 11 Beiyitiao, Zhongguancun, Beijing100190, China
| | - Jie Wang
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST)No. 11 Beiyitiao, Zhongguancun, Beijing100190, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing100049, P. R. China
| | - Xiang-Zhong Zeng
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST)No. 11 Beiyitiao, Zhongguancun, Beijing100190, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing100049, P. R. China
| | - Wanhai Xu
- Department of Urology, the Fourth Hospital of Harbin Medical University, Heilongjiang Key Laboratory of Scientific Research in Urology, No. 37 Yi-Yuan Street, Nangang District, Harbin, Heilongjiang Province150001, China
- NHC Key Laboratory of Molecular Probes and Targeted Diagnosis and Therapy, Harbin Medical University, Harbin, 150001, China
| | - Lei Wang
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST)No. 11 Beiyitiao, Zhongguancun, Beijing100190, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing100049, P. R. China
| |
Collapse
|
8
|
Yu X, Ruan M, Wang Y, Nguyen A, Xiao W, Ajena Y, Solano LN, Liu R, Lam KS. Site-Specific Albumin-Selective Ligation to Human Serum Albumin under Physiological Conditions. Bioconjug Chem 2022; 33:2332-2340. [PMID: 36350013 PMCID: PMC9782315 DOI: 10.1021/acs.bioconjchem.2c00361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Human serum albumin (HSA) is the most abundant protein in human blood plasma. It plays a critical role in the native transportation of numerous drugs, metabolites, nutrients, and small molecules. HSA has been successfully used clinically as a noncovalent carrier for insulin (e.g., Levemir), GLP-1 (e.g., Liraglutide), and paclitaxel (e.g., Abraxane). Site-specific bioconjugation strategies for HSA only would greatly expand its role as the biocompatible, non-toxic platform for theranostics purposes. Using the enabling one-bead one-compound (OBOC) technology, we generated combinatorial peptide libraries containing myristic acid, a well-known binder to HSA at Sudlow I and II binding pockets, and an acrylamide. We then used HSA as a probe to screen the OBOC myristylated peptide libraries for reactive affinity elements (RAEs) that can specifically and covalently ligate to the lysine residue at the proximity of these pockets. Several RAEs have been identified and confirmed to be able to conjugate to HSA covalently. The conjugation can occur at physiological pH and proceed with a high yield within 1 h at room temperature. Tryptic peptide profiling of derivatized HSA has revealed two lysine residues (K225 and K414) as the conjugation sites, which is much more specific than the conventional lysine labeling strategy with N-hydroxysuccinimide ester. The RAE-driven site-specific ligation to HSA was found to occur even in the presence of other prevalent blood proteins such as immunoglobulin or whole serum. Furthermore, these RAEs are orthogonal to the maleimide-based conjugation strategy for Cys34 of HSA. Together, these attributes make the RAEs the promising leads to further develop in vitro and in vivo HSA bioconjugation strategies for numerous biomedical applications.
Collapse
Affiliation(s)
- Xingjian Yu
- Department
of Chemistry, University of California Davis, Davis, 95616California, United States,Department
of Biochemistry & Molecular Medicine, School of Medicine, University of California Davis, Sacramento, California95817, United States
| | - Ming Ruan
- Department
of Biochemistry & Molecular Medicine, School of Medicine, University of California Davis, Sacramento, California95817, United States,School
of Food Science, Nanjing Xiaozhuang University, Nanjing, 211171, Jiangsu, China
| | - Yongheng Wang
- Department
of Biochemistry & Molecular Medicine, School of Medicine, University of California Davis, Sacramento, California95817, United States,Department
of Biomedical Engineering, University of
California Davis, Davis, California95616, United States
| | - Audrey Nguyen
- Department
of Biochemistry & Molecular Medicine, School of Medicine, University of California Davis, Sacramento, California95817, United States
| | - Wenwu Xiao
- Department
of Biochemistry & Molecular Medicine, School of Medicine, University of California Davis, Sacramento, California95817, United States
| | - Yousif Ajena
- Department
of Biochemistry & Molecular Medicine, School of Medicine, University of California Davis, Sacramento, California95817, United States
| | - Lucas N. Solano
- Department
of Biochemistry & Molecular Medicine, School of Medicine, University of California Davis, Sacramento, California95817, United States
| | - Ruiwu Liu
- Department
of Biochemistry & Molecular Medicine, School of Medicine, University of California Davis, Sacramento, California95817, United States,
| | - Kit S Lam
- Department
of Biochemistry & Molecular Medicine, School of Medicine, University of California Davis, Sacramento, California95817, United States,
| |
Collapse
|
9
|
Zhang L, Bo R, Wu Y, Li L, Zhu Z, Ma AH, Xiao W, Huang Y, Rojalin T, Yin X, Mao C, Wang F, Wang Y, Zhang H, Low KE, Lee K, Ajena Y, Jing D, Zhang D, Baehr CM, Liu R, Wang L, Li Y, Lam KS. Programmable Bispecific Nano-immunoengager That Captures T Cells and Reprograms Tumor Microenvironment. NANO LETTERS 2022; 22:6866-6876. [PMID: 35926215 PMCID: PMC9479133 DOI: 10.1021/acs.nanolett.2c00582] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Revised: 07/12/2022] [Indexed: 06/15/2023]
Abstract
Immune checkpoint blockade (ICB) therapy has revolutionized clinical oncology. However, the efficacy of ICB therapy is limited by the ineffective infiltration of T effector (Teff) cells to tumors and the immunosuppressive tumor microenvironment (TME). Here, we report a programmable tumor cells/Teff cells bispecific nano-immunoengager (NIE) that can circumvent these limitations to improve ICB therapy. The peptidic nanoparticles (NIE-NPs) bind tumor cell surface α3β1 integrin and undergo in situ transformation into nanofibrillar network nanofibers (NIE-NFs). The prolonged retained nanofibrillar network at the TME captures Teff cells via the activatable α4β1 integrin ligand and allows sustained release of resiquimod for immunomodulation. This bispecific NIE eliminates syngeneic 4T1 breast cancer and Lewis lung cancer models in mice, when given together with anti-PD-1 antibody. The in vivo structural transformation-based supramolecular bispecific NIE represents an innovative class of programmable receptor-mediated targeted immunotherapeutics to greatly enhance ICB therapy against cancers.
Collapse
Affiliation(s)
- Lu Zhang
- Department
of Biochemistry and Molecular Medicine, UC Davis NCI-designated Comprehensive
Cancer Center, University of California
Davis, Sacramento, California 95817, United States
- Department
of Biomedical Engineering, Southern University
of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Ruonan Bo
- Department
of Biochemistry and Molecular Medicine, UC Davis NCI-designated Comprehensive
Cancer Center, University of California
Davis, Sacramento, California 95817, United States
| | - Yi Wu
- Department
of Biochemistry and Molecular Medicine, UC Davis NCI-designated Comprehensive
Cancer Center, University of California
Davis, Sacramento, California 95817, United States
| | - Longmeng Li
- Department
of Biochemistry and Molecular Medicine, UC Davis NCI-designated Comprehensive
Cancer Center, University of California
Davis, Sacramento, California 95817, United States
| | - Zheng Zhu
- Department
of Biochemistry and Molecular Medicine, UC Davis NCI-designated Comprehensive
Cancer Center, University of California
Davis, Sacramento, California 95817, United States
| | - Ai-Hong Ma
- Department
of Biochemistry and Molecular Medicine, UC Davis NCI-designated Comprehensive
Cancer Center, University of California
Davis, Sacramento, California 95817, United States
| | - Wenwu Xiao
- Department
of Biochemistry and Molecular Medicine, UC Davis NCI-designated Comprehensive
Cancer Center, University of California
Davis, Sacramento, California 95817, United States
| | - Yanyu Huang
- Department
of Biochemistry and Molecular Medicine, UC Davis NCI-designated Comprehensive
Cancer Center, University of California
Davis, Sacramento, California 95817, United States
| | - Tatu Rojalin
- Department
of Biochemistry and Molecular Medicine, UC Davis NCI-designated Comprehensive
Cancer Center, University of California
Davis, Sacramento, California 95817, United States
| | - Xingbin Yin
- Department
of Biochemistry and Molecular Medicine, UC Davis NCI-designated Comprehensive
Cancer Center, University of California
Davis, Sacramento, California 95817, United States
| | - Chunping Mao
- Department
of Biomedical Engineering, Southern University
of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Fengyi Wang
- Department
of Biomedical Engineering, Southern University
of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Yongheng Wang
- Department
of Biochemistry and Molecular Medicine, UC Davis NCI-designated Comprehensive
Cancer Center, University of California
Davis, Sacramento, California 95817, United States
| | - Hongyong Zhang
- Department
of Biochemistry and Molecular Medicine, UC Davis NCI-designated Comprehensive
Cancer Center, University of California
Davis, Sacramento, California 95817, United States
| | - Kelmen E. Low
- Department
of Biochemistry and Molecular Medicine, UC Davis NCI-designated Comprehensive
Cancer Center, University of California
Davis, Sacramento, California 95817, United States
| | - Kiana Lee
- Department
of Biochemistry and Molecular Medicine, UC Davis NCI-designated Comprehensive
Cancer Center, University of California
Davis, Sacramento, California 95817, United States
| | - Yousif Ajena
- Department
of Biochemistry and Molecular Medicine, UC Davis NCI-designated Comprehensive
Cancer Center, University of California
Davis, Sacramento, California 95817, United States
| | - Di Jing
- Department
of Biochemistry and Molecular Medicine, UC Davis NCI-designated Comprehensive
Cancer Center, University of California
Davis, Sacramento, California 95817, United States
| | - Dalin Zhang
- Department
of Biochemistry and Molecular Medicine, UC Davis NCI-designated Comprehensive
Cancer Center, University of California
Davis, Sacramento, California 95817, United States
| | - Christopher M. Baehr
- Department
of Biochemistry and Molecular Medicine, UC Davis NCI-designated Comprehensive
Cancer Center, University of California
Davis, Sacramento, California 95817, United States
| | - Ruiwu Liu
- Department
of Biochemistry and Molecular Medicine, UC Davis NCI-designated Comprehensive
Cancer Center, University of California
Davis, Sacramento, California 95817, United States
| | - Lei Wang
- CAS
Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical
Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, Beijing 100190, China
| | - Yuanpei Li
- Department
of Biochemistry and Molecular Medicine, UC Davis NCI-designated Comprehensive
Cancer Center, University of California
Davis, Sacramento, California 95817, United States
| | - Kit S. Lam
- Department
of Biochemistry and Molecular Medicine, UC Davis NCI-designated Comprehensive
Cancer Center, University of California
Davis, Sacramento, California 95817, United States
- Division
of Hematology and Oncology, Department of Internal Medicine, School
of Medicine, University of California Davis, Sacramento, California 95817, United States
| |
Collapse
|
10
|
Vondrakova J, Frolikova M, Ded L, Cerny J, Postlerova P, Palenikova V, Simonik O, Nahacka Z, Basus K, Valaskova E, Machan R, Pacey A, Holubcova Z, Koubek P, Ezrova Z, Park S, Liu R, Partha R, Clark N, Neuzil J, Ikawa M, Erickson K, Lam KS, Moore H, Komrskova K. MAIA, Fc receptor-like 3, supersedes JUNO as IZUMO1 receptor during human fertilization. SCIENCE ADVANCES 2022; 8:eabn0047. [PMID: 36070373 PMCID: PMC9451160 DOI: 10.1126/sciadv.abn0047] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 07/23/2022] [Indexed: 05/17/2023]
Abstract
Gamete fusion is a critical event of mammalian fertilization. A random one-bead one-compound combinatorial peptide library represented synthetic human egg mimics and identified a previously unidentified ligand as Fc receptor-like 3, named MAIA after the mythological goddess intertwined with JUNO. This immunoglobulin super family receptor was expressed on human oolemma and played a major role during sperm-egg adhesion and fusion. MAIA forms a highly stable interaction with the known IZUMO1/JUNO sperm-egg complex, permitting specific gamete fusion. The complexity of the MAIA isotype may offer a cryptic sexual selection mechanism to avoid genetic incompatibility and achieve favorable fitness outcomes.
Collapse
Affiliation(s)
- Jana Vondrakova
- Laboratory of Reproductive Biology, Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Prumyslova 595, 252 50 Vestec, Czech Republic
| | - Michaela Frolikova
- Laboratory of Reproductive Biology, Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Prumyslova 595, 252 50 Vestec, Czech Republic
| | - Lukas Ded
- Laboratory of Reproductive Biology, Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Prumyslova 595, 252 50 Vestec, Czech Republic
| | - Jiri Cerny
- Laboratory of Structural Bioinformatics of Proteins, Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Prumyslova 595, 252 50 Vestec, Czech Republic
| | - Pavla Postlerova
- Laboratory of Reproductive Biology, Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Prumyslova 595, 252 50 Vestec, Czech Republic
- Department of Veterinary Sciences, Faculty of Agrobiology, Food, and Natural Resources, Czech University of Life Sciences Prague, Kamycka 129, 165 00 Prague, Czech Republic
| | - Veronika Palenikova
- Laboratory of Reproductive Biology, Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Prumyslova 595, 252 50 Vestec, Czech Republic
| | - Ondrej Simonik
- Laboratory of Reproductive Biology, Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Prumyslova 595, 252 50 Vestec, Czech Republic
| | - Zuzana Nahacka
- Laboratory of Molecular Therapy, Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Prumyslova 595, 252 50 Vestec, Czech Republic
| | - Krystof Basus
- Laboratory of Reproductive Biology, Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Prumyslova 595, 252 50 Vestec, Czech Republic
| | - Eliska Valaskova
- Laboratory of Reproductive Biology, Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Prumyslova 595, 252 50 Vestec, Czech Republic
| | - Radek Machan
- Imaging Methods Core Facility at BIOCEV, Faculty of Science, Charles University, Prumyslova 595, 252 50 Vestec, Czech Republic
| | - Allan Pacey
- Department of Oncology and Metabolism, University of Sheffield, Medical School, Sheffield S10 2RX, UK
| | - Zuzana Holubcova
- Reprofit International, Clinic of Reproductive Medicine, Brno, Czech Republic
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Pavel Koubek
- ProCrea Swiss IVF Center, Prague, Czech Republic
| | - Zuzana Ezrova
- Laboratory of Molecular Therapy, Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Prumyslova 595, 252 50 Vestec, Czech Republic
| | - Soojin Park
- Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Ruiwu Liu
- Department of Biochemistry and Molecular Medicine, University of California, Davis, School of Medicine, Davis, CA, USA
| | - Raghavendran Partha
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Nathan Clark
- Department of Human Genetics, University of Utah, Salt Lake City, UT, USA
| | - Jiri Neuzil
- Laboratory of Molecular Therapy, Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Prumyslova 595, 252 50 Vestec, Czech Republic
- School of Pharmacy and Medical Science, Griffith University, Parklands Avenue, Southport, Qld 4222, Australia
| | - Masahito Ikawa
- Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Kent Erickson
- Department of Cell Biology and Human Anatomy, University of California, Davis, School of Medicine, Davis, CA, USA
| | - Kit S. Lam
- Department of Biochemistry and Molecular Medicine, University of California, Davis, School of Medicine, Davis, CA, USA
| | - Harry Moore
- Centre for Stem Cell Biology, University of Sheffield, Sheffield S10 2TN, UK
| | - Katerina Komrskova
- Laboratory of Reproductive Biology, Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Prumyslova 595, 252 50 Vestec, Czech Republic
- Department of Zoology, Faculty of Science, Charles University, Vinicna 7, 128 44 Prague, Czech Republic
| |
Collapse
|
11
|
Visualizing γδ T cells by very late antigen-4-targeted positron emission tomography. Eur J Nucl Med Mol Imaging 2022; 49:4156-4170. [DOI: 10.1007/s00259-022-05886-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 06/17/2022] [Indexed: 11/28/2022]
|
12
|
Bellavia MC, Nyiranshuti L, Latoche JD, Ho KV, Fecek RJ, Taylor JL, Day KE, Nigam S, Pun M, Gallazzi F, Edinger RS, Storkus WJ, Patel RB, Anderson CJ. PET Imaging of VLA-4 in a New BRAF V600E Mouse Model of Melanoma. Mol Imaging Biol 2022; 24:425-433. [PMID: 34694528 PMCID: PMC9183947 DOI: 10.1007/s11307-021-01666-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 09/28/2021] [Accepted: 10/05/2021] [Indexed: 01/05/2023]
Abstract
PURPOSE Despite unprecedented responses to immune checkpoint inhibitors and targeted therapy in melanoma, a major subset of patients progresses and have few effective salvage options. We have previously demonstrated robust, selective uptake of the peptidomimetic LLP2A labeled with Cu-64 ([64Cu]-LLP2A) for positron emission tomography (PET) imaging in subcutaneous and metastatic models of B16F10 murine melanoma. LLP2A binds with high affinity to very late antigen-4 (VLA-4, integrin α4β1), a transmembrane protein overexpressed in melanoma and other cancers that facilitates tumor growth and metastasis. Yet B16F10 fails to faithfully reflect human melanoma biology, as it lacks certain oncogenic driver mutations, including BRAF mutations found in ≥ 50 % of clinical specimens. Here, we evaluated the PET tracer [64Cu]-CB-TE1A1P-PEG4-LLP2A ([64Cu]-LLP2A) in novel, translational BRAFV600E mutant melanoma models differing in VLA-4 expression-BPR (VLA-4-) and BPRα (VLA-4+). PROCEDURES BPR cells were transduced with α4 (CD49d) to overexpress intact cell surface VLA-4 (BPRα). The binding affinity of [64Cu]-LLP2A to BPR and BPRα cells was determined by saturation binding assays. [64Cu]-LLP2A internalization into B16F10, BPR, and BPRα cells was quantified via a plate-based assay. Tracer biodistribution and PET/CT imaging were evaluated in mice bearing subcutaneous BPR and BPRα tumors. RESULTS [64Cu]-LLP2A demonstrated high binding affinity to BPRα (Kd = 1.4 nM) but indeterminate binding to BPR cells. VLA-4+ BPRα and B16F10 displayed comparable time-dependent [64Cu]-LLP2A internalization, whereas BPR internalization was undetectable. PET/CT showed increased tracer uptake in BPRα tumors vs. BPR tumors in vivo, which was validated by significantly greater (p < 0.0001) BPRα tumor uptake in biodistribution analyses. CONCLUSIONS [64Cu]-LLP2A discriminates BPRα (VLA-4+) vs. BPR (VLA-4-) melanomas in vivo, supporting translation of these BRAF-mutated melanoma models via prospective imaging and theranostic studies. These results extend the utility of LLP2A to selectively target clinically relevant and therapy-resistant tumor variants toward its use for therapeutic patient care.
Collapse
Affiliation(s)
- Michael C Bellavia
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Lea Nyiranshuti
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, CA, 90095, USA
- RayzeBio Inc., San Diego, CA, 92121, USA
| | - Joseph D Latoche
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Khanh-Van Ho
- Department of Chemistry, University of Missouri, Columbia, MO, 65211, USA
| | - Ronald J Fecek
- Department of Dermatology, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Department of Microbiology and Immunology, Lake Erie College of Osteopathic Medicine at Seton Hill, Greensburg, PA, 15601, USA
| | - Jennifer L Taylor
- Department of Dermatology, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Kathryn E Day
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Shubhanchi Nigam
- Department of Radiology, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Eurofins Scientific, Philadelphia, PA, 19355, USA
| | - Michael Pun
- Department of Chemistry, University of Missouri, Columbia, MO, 65211, USA
| | - Fabio Gallazzi
- Department of Chemistry, University of Missouri, Columbia, MO, 65211, USA
| | - Robert S Edinger
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Department of Radiation Oncology, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Walter J Storkus
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Department of Dermatology, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Ravi B Patel
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, 15213, USA.
- Department of Radiation Oncology, University of Pittsburgh, Pittsburgh, PA, 15213, USA.
| | - Carolyn J Anderson
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, 15213, USA.
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA.
- Department of Chemistry, University of Missouri, Columbia, MO, 65211, USA.
- Department of Radiology, University of Pittsburgh, Pittsburgh, PA, 15213, USA.
- Department of Radiology, University of Missouri, Columbia, MO, 65211, USA.
| |
Collapse
|
13
|
Samec T, Boulos J, Gilmore S, Hazelton A, Alexander-Bryant A. Peptide-based delivery of therapeutics in cancer treatment. Mater Today Bio 2022; 14:100248. [PMID: 35434595 PMCID: PMC9010702 DOI: 10.1016/j.mtbio.2022.100248] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 03/14/2022] [Accepted: 03/27/2022] [Indexed: 11/09/2022] Open
Abstract
Current delivery strategies for cancer therapeutics commonly cause significant systemic side effects due to required high doses of therapeutic, inefficient cellular uptake of drug, and poor cell selectivity. Peptide-based delivery systems have shown the ability to alleviate these issues and can significantly enhance therapeutic loading, delivery, and cancer targetability. Peptide systems can be tailor-made for specific cancer applications. This review describes three peptide classes, targeting, cell penetrating, and fusogenic peptides, as stand-alone nanoparticle systems, conjugations to nanoparticle systems, or as the therapeutic modality. Peptide nanoparticle design, characteristics, and applications are discussed as well as peptide applications in the clinical space.
Collapse
Affiliation(s)
- Timothy Samec
- Nanobiotechnology Laboratory, Clemson University, Department of Bioengineering, Clemson, SC, USA
| | - Jessica Boulos
- Nanobiotechnology Laboratory, Clemson University, Department of Bioengineering, Clemson, SC, USA
| | - Serena Gilmore
- Nanobiotechnology Laboratory, Clemson University, Department of Bioengineering, Clemson, SC, USA
| | - Anthony Hazelton
- Nanobiotechnology Laboratory, Clemson University, Department of Bioengineering, Clemson, SC, USA
| | - Angela Alexander-Bryant
- Nanobiotechnology Laboratory, Clemson University, Department of Bioengineering, Clemson, SC, USA
| |
Collapse
|
14
|
Chu Y, Sun T, Jiang C. Emerging landscapes of nanosystems based on pre-metastatic microenvironment for cancer theranostics. CHINESE CHEM LETT 2022. [DOI: 10.1016/j.cclet.2022.02.051] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
15
|
Ge P, Yang M, Bouchard JL, Dzamko N, Lewis SJG, Halliday GM, Doran TM. Chemoselective Bioconjugation of Amyloidogenic Protein Antigens to PEGylated Microspheres Enables Detection of α-Synuclein Autoantibodies in Human Plasma. Bioconjug Chem 2022; 33:301-310. [PMID: 35020392 DOI: 10.1021/acs.bioconjchem.1c00530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The misfolding and subsequent aggregation of amyloidogenic proteins is a classic pathological hallmark of neurodegenerative diseases. Aggregates of the α-synuclein protein (αS) are implicated in Parkinson's disease (PD) pathogenesis, and naturally occurring autoantibodies to these aggregates are proposed to be potential early-stage biomarkers to facilitate the diagnosis of PD. However, upon misfolding, αS forms a multitude of quaternary structures of varying functions that are unstable ex vivo. Thus, when used as a capture agent in enzyme-linked immunosorbent assays (ELISAs), significant variance among laboratories has prevented the development of these valuable diagnostic tests. We reasoned that those conflicting results arise due to the high nonspecific binding and amyloid nucleation that are typical of ELISA platforms. In this work, we describe a multiplexed, easy-to-operate immunoassay that is generally applicable to quantify the levels of amyloid proteins and their binding partners, named Oxaziridine-Assisted Solid-phase Immunosorbent (OASIS) assay. The assay is built on a hydrophilic poly(ethylene glycol) scaffold that inhibits aggregate nucleation, which we show reduces assay variance when compared to similar ELISA measurements. To validate our OASIS assay in patient-derived samples, we measured the levels of naturally occurring antibodies against the αS monomer and oligomers in a cohort of donor plasma from patients diagnosed with PD. Using OASIS assays, we observed significantly higher titers of immunoglobulin G antibody recognizing αS oligomers in PD patients compared to those in healthy controls, while there was no significant difference in naturally occurring antibodies against the αS monomer. In addition to its development into a blood test to potentially predict or monitor PD, we anticipate that the OASIS assay will be of high utility for studies aimed at understanding protein misfolding, its pathology and symptomology in PD, and other neurodegenerative diseases.
Collapse
Affiliation(s)
- Peng Ge
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Mu Yang
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Jacob L Bouchard
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Nicolas Dzamko
- Brain and Mind Centre and Faculty of Medicine and Health, School of Medical Sciences, The University of Sydney, Camperdown, NSW 2050, Australia
| | - Simon J G Lewis
- Parkinson's Disease Research Clinic, Brain and Mind Centre, The University of Sydney, Camperdown, NSW 2050, Australia
| | - Glenda M Halliday
- Brain and Mind Centre and Faculty of Medicine and Health, School of Medical Sciences, The University of Sydney, Camperdown, NSW 2050, Australia
| | - Todd M Doran
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States.,Institute for Translational Neuroscience, University of Minnesota, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
16
|
Hathi D, Chanswangphuwana C, Cho N, Fontana F, Maji D, Ritchey J, O'Neal J, Ghai A, Duncan K, Akers WJ, Fiala M, Vij R, DiPersio JF, Rettig M, Shokeen M. Ablation of VLA4 in multiple myeloma cells redirects tumor spread and prolongs survival. Sci Rep 2022; 12:30. [PMID: 34996933 PMCID: PMC8741970 DOI: 10.1038/s41598-021-03748-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Accepted: 12/08/2021] [Indexed: 12/11/2022] Open
Abstract
Multiple myeloma (MM) is a cancer of bone marrow (BM) plasma cells, which is increasingly treatable but still incurable. In 90% of MM patients, severe osteolysis results from pathological interactions between MM cells and the bone microenvironment. Delineating specific molecules and pathways for their role in cancer supportive interactions in the BM is vital for developing new therapies. Very Late Antigen 4 (VLA4, integrin α4β1) is a key player in cell–cell adhesion and signaling between MM and BM cells. We evaluated a VLA4 selective near infrared fluorescent probe, LLP2A-Cy5, for in vitro and in vivo optical imaging of VLA4. Furthermore, two VLA4-null murine 5TGM1 MM cell (KO) clones were generated by CRISPR/Cas9 knockout of the Itga4 (α4) subunit, which induced significant alterations in the transcriptome. In contrast to the VLA4+ 5TGM1 parental cells, C57Bl/KaLwRij immunocompetent syngeneic mice inoculated with the VLA4-null clones showed prolonged survival, reduced medullary disease, and increased extramedullary disease burden. The KO tumor foci showed significantly reduced uptake of LLP2A-Cy5, confirming in vivo specificity of this imaging agent. This work provides new insights into the pathogenic role of VLA4 in MM, and evaluates an optical tool to measure its expression in preclinical models.
Collapse
Affiliation(s)
- Deep Hathi
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA
| | - Chantiya Chanswangphuwana
- Department of Medicine, Division of Molecular Oncology, Washington University School of Medicine, St. Louis, MO, USA.,Department of Medicine, Division of Hematology, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Bangkok, Thailand
| | - Nicholas Cho
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA
| | - Francesca Fontana
- Department of Medicine, Division of Cardiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Dolonchampa Maji
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA
| | - Julie Ritchey
- Department of Medicine, Division of Molecular Oncology, Washington University School of Medicine, St. Louis, MO, USA
| | - Julie O'Neal
- Department of Medicine, Division of Molecular Oncology, Washington University School of Medicine, St. Louis, MO, USA
| | - Anchal Ghai
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Kathleen Duncan
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Walter J Akers
- Center for In Vivo Imaging and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Mark Fiala
- Department of Medicine, Division of Molecular Oncology, Washington University School of Medicine, St. Louis, MO, USA
| | - Ravi Vij
- Department of Medicine, Division of Molecular Oncology, Washington University School of Medicine, St. Louis, MO, USA
| | - John F DiPersio
- Department of Medicine, Division of Molecular Oncology, Washington University School of Medicine, St. Louis, MO, USA
| | - Michael Rettig
- Department of Medicine, Division of Molecular Oncology, Washington University School of Medicine, St. Louis, MO, USA
| | - Monica Shokeen
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA. .,Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
17
|
Sartori A, Bugatti K, Portioli E, Baiula M, Casamassima I, Bruno A, Bianchini F, Curti C, Zanardi F, Battistini L. New 4-Aminoproline-Based Small Molecule Cyclopeptidomimetics as Potential Modulators of α 4β 1 Integrin. Molecules 2021; 26:molecules26196066. [PMID: 34641610 PMCID: PMC8512764 DOI: 10.3390/molecules26196066] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 10/01/2021] [Accepted: 10/04/2021] [Indexed: 02/01/2023] Open
Abstract
Integrin α4β1 belongs to the leukocyte integrin family and represents a therapeutic target of relevant interest given its primary role in mediating inflammation, autoimmune pathologies and cancer-related diseases. The focus of the present work is the design, synthesis and characterization of new peptidomimetic compounds that are potentially able to recognize α4β1 integrin and interfere with its function. To this aim, a collection of seven new cyclic peptidomimetics possessing both a 4-aminoproline (Amp) core scaffold grafted onto key α4β1-recognizing sequences and the (2-methylphenyl)ureido-phenylacetyl (MPUPA) appendage, was designed, with the support of molecular modeling studies. The new compounds were synthesized through SPPS procedures followed by in-solution cyclization maneuvers. The biological evaluation of the new cyclic ligands in cell adhesion assays on Jurkat cells revealed promising submicromolar agonist activity in one compound, namely, the c[Amp(MPUPA)Val-Asp-Leu] cyclopeptide. Further investigations will be necessary to complete the characterization of this class of compounds.
Collapse
Affiliation(s)
- Andrea Sartori
- Department of Food and Drug, University of Parma, Parco Area delle Scienze 27/A, 43124 Parma, Italy; (A.S.); (K.B.); (E.P.); (A.B.); (C.C.); (F.Z.)
| | - Kelly Bugatti
- Department of Food and Drug, University of Parma, Parco Area delle Scienze 27/A, 43124 Parma, Italy; (A.S.); (K.B.); (E.P.); (A.B.); (C.C.); (F.Z.)
| | - Elisabetta Portioli
- Department of Food and Drug, University of Parma, Parco Area delle Scienze 27/A, 43124 Parma, Italy; (A.S.); (K.B.); (E.P.); (A.B.); (C.C.); (F.Z.)
| | - Monica Baiula
- Department of Pharmacy and Biotechnology, University of Bologna, Via Irnerio 48, 40126 Bologna, Italy; (M.B.); (I.C.)
| | - Irene Casamassima
- Department of Pharmacy and Biotechnology, University of Bologna, Via Irnerio 48, 40126 Bologna, Italy; (M.B.); (I.C.)
| | - Agostino Bruno
- Department of Food and Drug, University of Parma, Parco Area delle Scienze 27/A, 43124 Parma, Italy; (A.S.); (K.B.); (E.P.); (A.B.); (C.C.); (F.Z.)
| | - Francesca Bianchini
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale G.B. Morgagni 50, 50134 Firenze, Italy;
| | - Claudio Curti
- Department of Food and Drug, University of Parma, Parco Area delle Scienze 27/A, 43124 Parma, Italy; (A.S.); (K.B.); (E.P.); (A.B.); (C.C.); (F.Z.)
| | - Franca Zanardi
- Department of Food and Drug, University of Parma, Parco Area delle Scienze 27/A, 43124 Parma, Italy; (A.S.); (K.B.); (E.P.); (A.B.); (C.C.); (F.Z.)
| | - Lucia Battistini
- Department of Food and Drug, University of Parma, Parco Area delle Scienze 27/A, 43124 Parma, Italy; (A.S.); (K.B.); (E.P.); (A.B.); (C.C.); (F.Z.)
- Correspondence: ; Tel.: +39-0521-906040
| |
Collapse
|
18
|
Ghai A, Fettig N, Fontana F, DiPersio J, Rettig M, Neal JO, Achilefu S, Shoghi KI, Shokeen M. In vivo quantitative assessment of therapeutic response to bortezomib therapy in disseminated animal models of multiple myeloma with [ 18F]FDG and [ 64Cu]Cu-LLP2A PET. EJNMMI Res 2021; 11:97. [PMID: 34586539 PMCID: PMC8481408 DOI: 10.1186/s13550-021-00840-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 09/13/2021] [Indexed: 02/06/2023] Open
Abstract
Background Multiple myeloma (MM) is a disease of cancerous plasma cells in the bone marrow. Imaging-based timely determination of therapeutic response is critical for improving outcomes in MM patients. Very late antigen-4 (VLA4, CD49d/CD29) is overexpressed in MM cells. Here, we evaluated [18F]FDG and VLA4 targeted [64Cu]Cu-LLP2A for quantitative PET imaging in disseminated MM models of variable VLA4 expression, following bortezomib therapy. Methods In vitro and ex vivo VLA4 expression was evaluated by flow cytometry. Human MM cells, MM.1S-CG and U266-CG (C: luciferase and G: green fluorescent protein), were injected intravenously in NOD-SCID gamma mice. Tumor progression was monitored by bioluminescence imaging (BLI). Treatment group received bortezomib (1 mg/kg, twice/week) intraperitoneally. All cohorts (treated, untreated and no tumor) were longitudinally imaged with [18F]FDG (7.4–8.0 MBq) and [64Cu]Cu-LLP2A (2–3 MBq; Molar Activity: 44.14 ± 1.40 MBq/nmol) PET, respectively. Results Flow cytometry confirmed high expression of CD49d in U266 cells (> 99%) and moderate expression in MM.1S cells (~ 52%). BLI showed decrease in total body flux in treated mice. In MM.1S-CG untreated versus treated mice, [64Cu]Cu-LLP2A localized with a significantly higher SUVmean in spine (0.58 versus 0.31, p < 0.01) and femur (0.72 versus 0.39, p < 0.05) at week 4 post-tumor inoculation. There was a four-fold higher uptake of [64Cu]Cu-LLP2A (SUVmean) in untreated U266-CG mice compared to treated mice at 3 weeks post-treatment. Compared to [64Cu]Cu-LLP2A, [18F]FDG PET detected treatment-related changes at later time points. Conclusion [64Cu]Cu-LLP2A is a promising tracer for timely in vivo assessment of therapeutic response in disseminated models of MM. Supplementary Information The online version contains supplementary material available at 10.1186/s13550-021-00840-4.
Collapse
Affiliation(s)
- Anchal Ghai
- Department of Radiology, Mallinckrodt Institute of Radiology, Washington University School of Medicine, 4515 McKinley Avenue, 2nd floor, St. Louis, MO, 63110, USA
| | - Nikki Fettig
- Department of Radiology, Mallinckrodt Institute of Radiology, Washington University School of Medicine, 4515 McKinley Avenue, 2nd floor, St. Louis, MO, 63110, USA
| | - Francesca Fontana
- Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - John DiPersio
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Mike Rettig
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Julie O Neal
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Samuel Achilefu
- Department of Radiology, Mallinckrodt Institute of Radiology, Washington University School of Medicine, 4515 McKinley Avenue, 2nd floor, St. Louis, MO, 63110, USA.,Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA.,Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO, USA
| | - Kooresh I Shoghi
- Department of Radiology, Mallinckrodt Institute of Radiology, Washington University School of Medicine, 4515 McKinley Avenue, 2nd floor, St. Louis, MO, 63110, USA
| | - Monica Shokeen
- Department of Radiology, Mallinckrodt Institute of Radiology, Washington University School of Medicine, 4515 McKinley Avenue, 2nd floor, St. Louis, MO, 63110, USA. .,Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA.
| |
Collapse
|
19
|
Development and Characterization of a Novel Peptide-Drug Conjugate with DM1 for Treatment of FGFR2-Positive Tumors. Biomedicines 2021; 9:biomedicines9080849. [PMID: 34440055 PMCID: PMC8389697 DOI: 10.3390/biomedicines9080849] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 07/13/2021] [Accepted: 07/15/2021] [Indexed: 11/17/2022] Open
Abstract
A maytansin derivative, DM1, is a promising therapeutic compound for treating tumors, but is also a highly poisonous substance with various side effects. For clinical expansion, we tried to develop novel peptide–drug conjugates (PDCs) with DM1. In the study, a one-bead one-compound (OBOC) platform was used to screen and identify a novel, highly stable, non-natural amino acid peptide targeting the tyrosine receptor FGFR2. Then, the identified peptide, named LLC2B, was conjugated with the cytotoxin DM1. Our results show that LLC2B has high affinity for the FGFR2 protein according to an isothermal titration calorimetry (ITC) test. LLC2B-Cy5.5 binding to FGFR2-positive cancer cells was confirmed by fluorescent microscopic imaging and flow cytometry in vitro. Using xenografted nude mouse models established with breast cancer MCF-7 cells and esophageal squamous cell carcinoma KYSE180 cells, respectively, LLC2B-Cy5.5 was observed to specifically target tumor tissues 24 h after tail vein injection. Incubation assays, both in aqueous solution at room temperature and in human plasma at 37 °C, suggested that LLC2B has high stability and strong anti-proteolytic ability. Then, we used two different linkers, one of molecular disulfide bonds and another of a maleimide group, to couple LLC2B to the toxin DM1. The novel peptide–drug conjugates (PDCs) inhibited tumor growth and significantly increased the maximum tolerated dose of DM1 in xenografted mice. In brief, our results suggest that LLC2B–DM1 can be developed into a potential PDC for tumor treatment in the future.
Collapse
|
20
|
Zheleznyak A, Mixdorf M, Marsala L, Prior J, Yang X, Cui G, Xu B, Fletcher S, Fontana F, Lanza G, Achilefu S. Orthogonal targeting of osteoclasts and myeloma cells for radionuclide stimulated dynamic therapy induces multidimensional cell death pathways. Theranostics 2021; 11:7735-7754. [PMID: 34335961 PMCID: PMC8315072 DOI: 10.7150/thno.60757] [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] [Received: 03/22/2021] [Accepted: 06/09/2021] [Indexed: 12/13/2022] Open
Abstract
Rationale: Multiple myeloma (MM) is a multifocal malignancy of bone marrow plasma cells, characterized by vicious cycles of remission and relapse that eventually culminate in death. The disease remains mostly incurable largely due to the complex interactions between the bone microenvironment (BME) and MM cells (MMC). In the “vicious cycle” of bone disease, abnormal activation of osteoclasts (OCs) by MMC causes severe osteolysis, promotes immune evasion, and stimulates the growth of MMC. Disrupting these cancer-stroma interactions would enhance treatment response. Methods: To disrupt this cycle, we orthogonally targeted nanomicelles (NM) loaded with non-therapeutic doses of a photosensitizer, titanocene (TC), to VLA-4 (α4ß1, CD49d/CD29) expressing MMC (MM1.S) and αvß3 (CD51/CD61) expressing OC. Concurrently, a non-lethal dose of a radiopharmaceutical, 18F-fluorodeoxyglucose ([18F]FDG) administered systemically interacted with TC (radionuclide stimulated therapy, RaST) to generate cytotoxic reactive oxygen species (ROS). The in vitro and in vivo effects of RaST were characterized in MM1.S cell line, as well as in xenograft and isograft MM animal models. Results: Our data revealed that RaST induced non-enzymatic hydroperoxidation of cellular lipids culminating in mitochondrial dysfunction, DNA fragmentation, and caspase-dependent apoptosis of MMC using VLA-4 avid TC-NMs. RaST upregulated the expression of BAX, Bcl-2, and p53, highlighting the induction of apoptosis via the BAK-independent pathway. The enhancement of multicopper oxidase enzyme F5 expression, which inhibits lipid hydroperoxidation and Fenton reaction, was not sufficient to overcome RaST-induced increase in the accumulation of irreversible function-perturbing α,ß-aldehydes that exerted significant and long-lasting damage to both DNA and proteins. In vivo, either VLA-4-TC-NM or αvß3-TC-NMs RaST induced a significant therapeutic effect on immunocompromised but not immunocompetent MM-bearing mouse models. Combined treatment with both VLA-4-TC-NM and αvß3-TC-NMs synergistically inhibited osteolysis, reduced tumor burden, and prevented rapid relapse in both in vivo models of MM. Conclusions: By targeting MM and bone cells simultaneously, combination RaST suppressed MM disease progression through a multi-prong action on the vicious cycle of bone cancer. Instead of using the standard multidrug approach, our work reveals a unique photophysical treatment paradigm that uses nontoxic doses of a single light-sensitive drug directed orthogonally to cancer and bone cells, followed by radionuclide-stimulated generation of ROS to inhibit tumor progression and minimize osteolysis in both immunocompetent murine and immunocompromised human MM models.
Collapse
|
21
|
Xiao K, Liu Q, Suby N, Xiao W, Agrawal R, Vu M, Zhang H, Luo Y, Li Y, Lam KS. LHRH-Targeted Redox-Responsive Crosslinked Micelles Impart Selective Drug Delivery and Effective Chemotherapy in Triple-Negative Breast Cancer. Adv Healthc Mater 2021; 10:e2001196. [PMID: 33200571 PMCID: PMC7858235 DOI: 10.1002/adhm.202001196] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 10/20/2020] [Indexed: 02/05/2023]
Abstract
Systemic chemotherapy is efficacious against triple-negative breast cancer (TNBC), but it is often associated with serious side effects. Here, a luteinizing hormone-releasing hormone (LHRH) receptor-targeted and tumor microenvironment-responsive nanoparticle system to selectively deliver chemotherapeutic drugs to TNBC cells, is reported. This delivery system (termed "LHRH-DCMs") contains poly(ethylene glycol) and dendritic cholic acid as a micellar carrier, reversible intra-micellar disulfide bond as a redox-responsive crosslink, and synthetic high-affinity (D-Lys)-LHRH peptide as a targeting moiety. LHRH-DCMs exhibit high drug loading efficiency, optimal particle size, good colloidal stability, and glutathione-responsive drug release. As expected, LHRH-DCMs are more efficiently internalized into human TNBC cells through receptor-mediated endocytosis, resulting in stronger cytotoxicity against these cancer cells than the non-targeted counterpart when encapsulated with paclitaxel (PTX). Furthermore, near-infrared fluorescence and magnetic resonance imaging demonstrate that LHRH-DCMs facilitate the tumor distribution and penetration of payloads in three different animal models of breast cancer, including cell line-derived xenograft (CDX), patient-derived xenograft (PDX), and transgenic mammary carcinoma. Finally, in vivo therapeutic studies show that PTX-LHRH-DCMs outperform both the corresponding nontargeted PTX-DCMs and the current clinical formulation (Taxol) in an orthotopic TNBC model. These results provide new insights into approaches for precise drug delivery of TNBC.
Collapse
Affiliation(s)
- Kai Xiao
- National Chengdu Center for Safety Evaluation of Drugs and National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, 610041, P. R. China
| | - Qiangqiang Liu
- National Chengdu Center for Safety Evaluation of Drugs and National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, 610041, P. R. China
| | - Nell Suby
- Department of Obstetrics and Gynecology, School of Medicine, University of California, Davis, CA, 95817, USA
| | - Wenwu Xiao
- Department of Biochemistry & Molecular Medicine, UC Davis Cancer Center, University of California, Davis, Sacramento, CA, 95817, USA
| | - Rinki Agrawal
- Department of Obstetrics and Gynecology, School of Medicine, University of California, Davis, CA, 95817, USA
| | - Michael Vu
- Department of Biochemistry & Molecular Medicine, UC Davis Cancer Center, University of California, Davis, Sacramento, CA, 95817, USA
| | - Hongyong Zhang
- Division of Hematology & Oncology, Department of Internal Medicine, School of Medicine, University of California, Davis, CA, 95817, USA
| | - Yan Luo
- Department of Biochemistry & Molecular Medicine, UC Davis Cancer Center, University of California, Davis, Sacramento, CA, 95817, USA
| | - Yuanpei Li
- Department of Biochemistry & Molecular Medicine, UC Davis Cancer Center, University of California, Davis, Sacramento, CA, 95817, USA
| | - Kit S Lam
- Department of Biochemistry & Molecular Medicine, UC Davis Cancer Center, University of California, Davis, Sacramento, CA, 95817, USA
- Division of Hematology & Oncology, Department of Internal Medicine, School of Medicine, University of California, Davis, CA, 95817, USA
| |
Collapse
|
22
|
Fontana F, Scott MJ, Allen JS, Yang X, Cui G, Pan D, Yanaba N, Fiala MA, O'Neal J, Schmieder-Atteberry AH, Ritchey J, Rettig M, Simons K, Fletcher S, Vij R, DiPersio JF, Lanza GM. VLA4-Targeted Nanoparticles Hijack Cell Adhesion-Mediated Drug Resistance to Target Refractory Myeloma Cells and Prolong Survival. Clin Cancer Res 2020; 27:1974-1986. [PMID: 33355244 DOI: 10.1158/1078-0432.ccr-20-2839] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 10/20/2020] [Accepted: 12/15/2020] [Indexed: 11/16/2022]
Abstract
PURPOSE In multiple myeloma, drug-resistant cells underlie relapse or progression following chemotherapy. Cell adhesion-mediated drug resistance (CAM-DR) is an established mechanism used by myeloma cells (MMC) to survive chemotherapy and its markers are upregulated in residual disease. The integrin very late antigen 4 (VLA4; α4β1) is a key mediator of CAM-DR and its expression affects drug sensitivity of MMCs. Rather than trying to inhibit its function, here, we hypothesized that upregulation of VLA4 by resistant MMCs could be exploited for targeted delivery of drugs, which would improve safety and efficacy of treatments. EXPERIMENTAL DESIGN We synthetized 20 nm VLA4-targeted micellar nanoparticles (V-NP) carrying DiI for tracing or a novel camptothecin prodrug (V-CP). Human or murine MMCs, alone or with stroma, and immunocompetent mice with orthotopic multiple myeloma were used to track delivery of NPs and response to treatments. RESULTS V-NPs selectively delivered their payload to MMCs in vitro and in vivo, and chemotherapy increased their uptake by surviving MMCs. V-CP, alone or in combination with melphalan, was well tolerated and prolonged survival in myeloma-bearing mice. V-CP also reduced the dose requirement for melphalan, reducing tumor burden in association with suboptimal dosing without increasing overall toxicity. CONCLUSIONS V-CP may be a safe and effective strategy to prevent or treat relapsing or refractory myeloma. V-NP targeting of resistant cells may suggest a new approach to environment-induced resistance in cancer.
Collapse
Affiliation(s)
- Francesca Fontana
- Division of Cardiology, Washington University School of Medicine, St. Louis, Missouri. .,Divison of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Michael J Scott
- Division of Cardiology, Washington University School of Medicine, St. Louis, Missouri
| | - John S Allen
- Division of Cardiology, Washington University School of Medicine, St. Louis, Missouri
| | - Xiaoxia Yang
- Division of Cardiology, Washington University School of Medicine, St. Louis, Missouri
| | - Grace Cui
- Division of Cardiology, Washington University School of Medicine, St. Louis, Missouri
| | - Dipanjan Pan
- University of Maryland, Baltimore County, University of Maryland School of Medicine, Baltimore, Maryland
| | - Noriko Yanaba
- Division of Cardiology, Washington University School of Medicine, St. Louis, Missouri
| | - Mark A Fiala
- Divison of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Julie O'Neal
- Divison of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | | | - Julie Ritchey
- Divison of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Michael Rettig
- Divison of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Kathleen Simons
- SUNY Downstate Health Sciences University, New York, New York
| | - Steven Fletcher
- University of Maryland School of Pharmacy, Baltimore, Maryland
| | - Ravi Vij
- Divison of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - John F DiPersio
- Divison of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Gregory M Lanza
- Division of Cardiology, Washington University School of Medicine, St. Louis, Missouri.
| |
Collapse
|
23
|
Miao Y, Quinn TP. Advances in Receptor-Targeted Radiolabeled Peptides for Melanoma Imaging and Therapy. J Nucl Med 2020; 62:313-318. [PMID: 33277401 DOI: 10.2967/jnumed.120.243840] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 11/20/2020] [Indexed: 12/29/2022] Open
Abstract
Melanocortin-1 receptor (MC1R) and very late antigen-4 (VLA-4, integrin α4β1) are 2 attractive molecular targets for developing peptide radiopharmaceuticals for melanoma imaging and therapy. MC1R- and VLA-4-targeting peptides and peptide-conjugated Cornell prime dots (C' dots) can serve as delivery vehicles to target both diagnostic and therapeutic radionuclides to melanoma cells for imaging and therapy. This review highlights the advances of MC1R- and VLA-4-targeted radiolabeled peptides and peptide-conjugated C' dots for melanoma imaging and therapy. The promising preclinical and clinical results of these new peptide radiopharmaceuticals present an optimistic outlook for clinical translation into receptor-targeting melanoma imaging and radionuclide therapy in the future.
Collapse
Affiliation(s)
- Yubin Miao
- Department of Radiology, School of Medicine, University of Colorado Denver, Aurora, Colorado; and
| | - Thomas P Quinn
- Department of Biochemistry, University of Missouri-Columbia, and Harry S. Truman Veterans' Hospital, Columbia, Missouri
| |
Collapse
|
24
|
Qi J, Rader C. Redirecting cytotoxic T cells with chemically programmed antibodies. Bioorg Med Chem 2020; 28:115834. [PMID: 33166926 DOI: 10.1016/j.bmc.2020.115834] [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: 09/11/2020] [Revised: 10/20/2020] [Accepted: 10/24/2020] [Indexed: 11/30/2022]
Abstract
T-cell engaging bispecific antibodies (T-biAbs) mediate potent and selective cytotoxicity by combining specificities for target and effector cells in one molecule. Chemically programmed T-biAbs (cp-T-biAbs) are precisely assembled compositions of (i) small molecules that govern cancer cell surface targeting with high affinity and specificity and (ii) antibodies that recruit and activate T cells and equip the small molecule with confined biodistribution and longer circulatory half-life. Conceptually similar to cp-T-biAbs, switchable chimeric antigen receptor T cells (sCAR-Ts) can also be put under the control of small molecules by using a chemically programmed antibody as a bispecific adaptor molecule. As such, cp-T-biAbs and cp-sCAR-Ts can endow small molecules with the power of cancer immunotherapy. We here review the concept of chemically programmed antibodies for recruiting and activating T cells as a promising strategy for broadening the utility of small molecules in cancer therapy.
Collapse
Affiliation(s)
- Junpeng Qi
- Department of Immunology and Microbiology, The Scripps Research Institute, Jupiter, FL 33458, USA.
| | - Christoph Rader
- Department of Immunology and Microbiology, The Scripps Research Institute, Jupiter, FL 33458, USA.
| |
Collapse
|
25
|
Ding Y, Ting JP, Liu J, Al-Azzam S, Pandya P, Afshar S. Impact of non-proteinogenic amino acids in the discovery and development of peptide therapeutics. Amino Acids 2020; 52:1207-1226. [PMID: 32945974 PMCID: PMC7544725 DOI: 10.1007/s00726-020-02890-9] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 09/05/2020] [Indexed: 12/14/2022]
Abstract
With the development of modern chemistry and biology, non-proteinogenic amino acids (NPAAs) have become a powerful tool for developing peptide-based drug candidates. Drug-like properties of peptidic medicines, due to the smaller size and simpler structure compared to large proteins, can be changed fundamentally by introducing NPAAs in its sequence. While peptides composed of natural amino acids can be used as drug candidates, the majority have shown to be less stable in biological conditions. The impact of NPAA incorporation can be extremely beneficial in improving the stability, potency, permeability, and bioavailability of peptide-based therapies. Conversely, undesired effects such as toxicity or immunogenicity should also be considered. The impact of NPAAs in the development of peptide-based therapeutics is reviewed in this article. Further, numerous examples of peptides containing NPAAs are presented to highlight the ongoing development in peptide-based therapeutics.
Collapse
Affiliation(s)
- Yun Ding
- Protein Engineering, Lilly Biotechnology Center, Eli Lilly and Company, San Diego, CA, 92121, USA
| | - Joey Paolo Ting
- Protein Engineering, Lilly Biotechnology Center, Eli Lilly and Company, San Diego, CA, 92121, USA
| | - Jinsha Liu
- Protein Engineering, Lilly Biotechnology Center, Eli Lilly and Company, San Diego, CA, 92121, USA
| | - Shams Al-Azzam
- Professional Scientific Services, Eurofins Lancaster Laboratories, Lancaster, PA, 17605, USA
| | - Priyanka Pandya
- Protein Engineering, Lilly Biotechnology Center, Eli Lilly and Company, San Diego, CA, 92121, USA
| | - Sepideh Afshar
- Protein Engineering, Lilly Biotechnology Center, Eli Lilly and Company, San Diego, CA, 92121, USA.
| |
Collapse
|
26
|
Jiang M, Liu R, Liu L, Kot A, Liu X, Xiao W, Jia J, Li Y, Lam KS, Yao W. Identification of osteogenic progenitor cell-targeted peptides that augment bone formation. Nat Commun 2020; 11:4278. [PMID: 32855388 PMCID: PMC7453024 DOI: 10.1038/s41467-020-17417-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 06/16/2020] [Indexed: 12/02/2022] Open
Abstract
Activation and migration of endogenous mesenchymal stromal cells (MSCs) are critical for bone regeneration. Here, we report a combinational peptide screening strategy for rapid discovery of ligands that not only bind strongly to osteogenic progenitor cells (OPCs) but also stimulate osteogenic cell Akt signaling in those OPCs. Two lead compounds are discovered, YLL3 and YLL8, both of which increase osteoprogenitor osteogenic differentiation in vitro. When given to normal or osteopenic mice, the compounds increase mineral apposition rate, bone formation, bone mass, and bone strength, as well as expedite fracture repair through stimulated endogenous osteogenesis. When covalently conjugated to alendronate, YLLs acquire an additional function resulting in a “tri-functional” compound that: (i) binds to OPCs, (ii) targets bone, and (iii) induces “pro-survival” signal. These bone-targeted, osteogenic peptides are well suited for current tissue-specific therapeutic paradigms to augment the endogenous osteogenic cells for bone regeneration and the treatment of bone loss. Activation of osteogenic cells is essential for bone regeneration. Here, the authors screen a peptide library and identify 2 compounds that promote osteogenic progenitor cell differentiation in vitro, and show that they increase bone formation and fracture repair in mice.
Collapse
Affiliation(s)
- Min Jiang
- Center for Musculoskeletal Health, Department of Internal Medicine, The University of California at Davis Medical Center, Sacramento, CA, 95817, USA.,Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, 200025, Shanghai, China
| | - Ruiwu Liu
- Department of Biochemistry and Molecular Medicine, The University of California at Davis Medical Center, Sacramento, CA, 95817, USA
| | - Lixian Liu
- Center for Musculoskeletal Health, Department of Internal Medicine, The University of California at Davis Medical Center, Sacramento, CA, 95817, USA
| | - Alexander Kot
- Center for Musculoskeletal Health, Department of Internal Medicine, The University of California at Davis Medical Center, Sacramento, CA, 95817, USA
| | - Xueping Liu
- Center for Musculoskeletal Health, Department of Internal Medicine, The University of California at Davis Medical Center, Sacramento, CA, 95817, USA
| | - Wenwu Xiao
- Department of Biochemistry and Molecular Medicine, The University of California at Davis Medical Center, Sacramento, CA, 95817, USA
| | - Junjing Jia
- Center for Musculoskeletal Health, Department of Internal Medicine, The University of California at Davis Medical Center, Sacramento, CA, 95817, USA
| | - Yuanpei Li
- Department of Biochemistry and Molecular Medicine, The University of California at Davis Medical Center, Sacramento, CA, 95817, USA
| | - Kit S Lam
- Department of Biochemistry and Molecular Medicine, The University of California at Davis Medical Center, Sacramento, CA, 95817, USA
| | - Wei Yao
- Center for Musculoskeletal Health, Department of Internal Medicine, The University of California at Davis Medical Center, Sacramento, CA, 95817, USA.
| |
Collapse
|
27
|
Barkat HA, Das SS, Barkat MA, Beg S, Hadi HA. Selective targeting of cancer signaling pathways with nanomedicines: challenges and progress. Future Oncol 2020; 16:2959-2979. [PMID: 32805124 DOI: 10.2217/fon-2020-0198] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Cancer is one of the leading causes of death worldwide. Regardless of advances in understanding the molecular mechanics of cancer, its treatment is still lacking and the death rates for many forms of the disease remain the same as six decades ago. Although a variety of therapeutic agents and strategies have been reported, these therapies often failed to provide efficient therapy to patients as a consequence of the inability to deliver right and adequate chemotherapeutic agents to the right place. However, the situation has started to revolutionize substantially with the advent of novel 'targeted' nanocarrier-based cancer therapies. Such therapies hold great potential in cancer management as they are biocompatible, tailored to specific needs, tolerated and deliver enough drugs at the targeted site. Their use also enhances the delivery of chemotherapeutics by improving biodistribution, lowering toxicity, inhibiting degradation and increasing cellular uptake. However, in some instances, nonselective targeting is not enough and the inclusion of a ligand moiety is required to achieve tumor targeting and enhanced drug accumulation at the tumor site. This contemporary review outlines the targeting potential of nanocarriers, highlighting the essentiality of nanoparticles, tumor-associated molecular signaling pathways, and various biological and pathophysiological barriers.
Collapse
Affiliation(s)
- Harshita Abul Barkat
- Department of Pharmaceutics, College of Pharmacy, University of Hafr Al Batin, Al Jamiah, Hafr Al-Batin, 39524, Saudi Arabia
| | - Sabya Sachi Das
- Department of Pharmaceutical Sciences & Technology, BIT, Mesra, Ranchi, 835215, Jharkhand, India
| | - Md Abul Barkat
- Department of Pharmaceutics, College of Pharmacy, University of Hafr Al Batin, Al Jamiah, Hafr Al-Batin, 39524, Saudi Arabia
| | - Sarwar Beg
- Department of Pharmaceutics, Nanomedicine Research Lab, School of Pharmaceutical Education & Research, Jamia Hamdard, New Delhi, India
| | - Hazrina Ab Hadi
- Department of Pharmaceutical Technology, Faculty of Pharmacy, International Islamic University Malaysia, Kuantan, Pahang, 25200, Malaysia
| |
Collapse
|
28
|
Hao D, Liu R, Gao K, He C, He S, Zhao C, Sun G, Farmer DL, Panitch A, Lam KS, Wang A. Developing an Injectable Nanofibrous Extracellular Matrix Hydrogel With an Integrin αvβ3 Ligand to Improve Endothelial Cell Survival, Engraftment and Vascularization. Front Bioeng Biotechnol 2020; 8:890. [PMID: 32850742 PMCID: PMC7403189 DOI: 10.3389/fbioe.2020.00890] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Accepted: 07/10/2020] [Indexed: 01/01/2023] Open
Abstract
Endothelial cell (EC) transplantation via injectable collagen hydrogel has received much attention as a potential treatment for various vascular diseases. However, the therapeutic effect of transplanted ECs is limited by their poor viability, which partially occurs as a result of cellular apoptosis triggered by the insufficient cell-extracellular matrix (ECM) engagement. Integrin binding to the ECM is crucial for cell anchorage to the surrounding matrix, cell spreading and migration, and further activation of intracellular signaling pathways. Although collagen contains several different types of integrin binding sites, it still lacks sufficient specific binding sites for ECs. Previously, using one-bead one-compound (OBOC) combinatorial technology, we identified LXW7, an integrin αvβ3 ligand, which possessed a strong binding affinity to and enhanced functionality of ECs. In this study, to improve the EC-matrix interaction, we developed an approach to molecularly conjugate LXW7 to the collagen backbone, via a collagen binding peptide SILY, in order to increase EC specific integrin binding sites on the collagen hydrogel. Results showed that in the in vitro 2-dimensional (2D) culture model, the LXW7-treated collagen surface significantly improved EC attachment and survival and decreased caspase 3 activity in an ischemic-mimicking environment. In the in vitro 3-dimensional (3D) culture model, LXW7-modified collagen hydrogel significantly improved EC spreading, proliferation, and survival. In a mouse subcutaneous implantation model, LXW7-modified collagen hydrogel improved the engraftment of transplanted ECs and supported ECs to form vascular network structures. Therefore, LXW7-functionalized collagen hydrogel has shown promising potential to improve vascularization in tissue regeneration and may be used as a novel tool for EC delivery and the treatment of vascular diseases.
Collapse
Affiliation(s)
- Dake Hao
- Department of Surgery, School of Medicine, University of California, Davis, Sacramento, CA, United States
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, CA, United States
| | - Ruiwu Liu
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, Sacramento, CA, United States
| | - Kewa Gao
- Department of Surgery, School of Medicine, University of California, Davis, Sacramento, CA, United States
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, CA, United States
| | - Chuanchao He
- Department of Surgery, School of Medicine, University of California, Davis, Sacramento, CA, United States
| | - Siqi He
- Department of Surgery, School of Medicine, University of California, Davis, Sacramento, CA, United States
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, CA, United States
| | - Cunyi Zhao
- Department of Biological and Agricultural Engineering, University of California, Davis, Davis, CA, United States
| | - Gang Sun
- Department of Biological and Agricultural Engineering, University of California, Davis, Davis, CA, United States
| | - Diana L. Farmer
- Department of Surgery, School of Medicine, University of California, Davis, Sacramento, CA, United States
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, CA, United States
| | - Alyssa Panitch
- Department of Surgery, School of Medicine, University of California, Davis, Sacramento, CA, United States
- Department of Biomedical Engineering, University of California, Davis, Davis, CA, United States
| | - Kit S. Lam
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, Sacramento, CA, United States
| | - Aijun Wang
- Department of Surgery, School of Medicine, University of California, Davis, Sacramento, CA, United States
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, CA, United States
- Department of Biomedical Engineering, University of California, Davis, Davis, CA, United States
| |
Collapse
|
29
|
Haddad J, Latoche JD, Nigam S, Bellavia MC, Day KE, Zhu Q, Edwards WB, Anderson CJ, Tavakoli S. Molecular Imaging of Very Late Antigen-4 in Acute Lung Injury. J Nucl Med 2020; 62:280-286. [DOI: 10.2967/jnumed.120.242347] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 06/23/2020] [Indexed: 11/16/2022] Open
|
30
|
Hao D, Swindell HS, Ramasubramanian L, Liu R, Lam KS, Farmer DL, Wang A. Extracellular Matrix Mimicking Nanofibrous Scaffolds Modified With Mesenchymal Stem Cell-Derived Extracellular Vesicles for Improved Vascularization. Front Bioeng Biotechnol 2020; 8:633. [PMID: 32671037 PMCID: PMC7329993 DOI: 10.3389/fbioe.2020.00633] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Accepted: 05/22/2020] [Indexed: 12/17/2022] Open
Abstract
The network structure and biological components of natural extracellular matrix (ECM) are indispensable for promoting tissue regeneration. Electrospun nanofibrous scaffolds have been widely used in regenerative medicine to provide structural support for cell growth and tissue regeneration due to their natural ECM mimicking architecture, however, they lack biological functions. Extracellular vesicles (EVs) are potent vehicles of intercellular communication due to their ability to transfer RNAs, proteins, and lipids, thereby mediating significant biological functions in different biological systems. Matrix-bound nanovesicles (MBVs) are identified as an integral and functional component of ECM bioscaffolds mediating significant regenerative functions. Therefore, to engineer EVs modified electrospun scaffolds, mimicking the structure of the natural EV-ECM complex and the physiological interactions between the ECM and EVs, will be attractive and promising in tissue regeneration. Previously, using one-bead one-compound (OBOC) combinatorial technology, we identified LLP2A, an integrin α4β1 ligand, which had a strong binding to human placenta-derived mesenchymal stem cells (PMSCs). In this study, we isolated PMSCs derived EVs (PMSC-EVs) and demonstrated they expressed integrin α4β1 and could improve endothelial cell (EC) migration and vascular sprouting in an ex vivo rat aortic ring assay. LLP2A treated culture surface significantly improved PMSC-EV attachment, and the PMSC-EV treated culture surface significantly enhanced the expression of angiogenic genes and suppressed apoptotic activity. We then developed an approach to enable "Click chemistry" to immobilize LLP2A onto the surface of electrospun scaffolds as a linker to immobilize PMSC-EVs onto the scaffold. The PMSC-EV modified electrospun scaffolds significantly promoted EC survival and angiogenic gene expression, such as KDR and TIE2, and suppressed the expression of apoptotic markers, such as caspase 9 and caspase 3. Thus, PMSC-EVs hold promising potential to functionalize biomaterial constructs and improve the vascularization and regenerative potential. The EVs modified biomaterial scaffolds can be widely used for different tissue engineering applications.
Collapse
Affiliation(s)
- Dake Hao
- Department of Surgery, School of Medicine, University of California, Davis, Sacramento, CA, United States
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, CA, United States
| | - Hila Shimshi Swindell
- Department of Surgery, School of Medicine, University of California, Davis, Sacramento, CA, United States
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, CA, United States
| | - Lalithasri Ramasubramanian
- Department of Surgery, School of Medicine, University of California, Davis, Sacramento, CA, United States
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, CA, United States
| | - Ruiwu Liu
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, Sacramento, CA, United States
| | - Kit S. Lam
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, Sacramento, CA, United States
| | - Diana L. Farmer
- Department of Surgery, School of Medicine, University of California, Davis, Sacramento, CA, United States
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, CA, United States
| | - Aijun Wang
- Department of Surgery, School of Medicine, University of California, Davis, Sacramento, CA, United States
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, CA, United States
- Department of Biomedical Engineering, University of California, Davis, Davis, CA, United States
| |
Collapse
|
31
|
Lau J, Rousseau E, Kwon D, Lin KS, Bénard F, Chen X. Insight into the Development of PET Radiopharmaceuticals for Oncology. Cancers (Basel) 2020; 12:E1312. [PMID: 32455729 PMCID: PMC7281377 DOI: 10.3390/cancers12051312] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 05/17/2020] [Accepted: 05/18/2020] [Indexed: 12/20/2022] Open
Abstract
While the development of positron emission tomography (PET) radiopharmaceuticals closely follows that of traditional drug development, there are several key considerations in the chemical and radiochemical synthesis, preclinical assessment, and clinical translation of PET radiotracers. As such, we outline the fundamentals of radiotracer design, with respect to the selection of an appropriate pharmacophore. These concepts will be reinforced by exemplary cases of PET radiotracer development, both with respect to their preclinical and clinical evaluation. We also provide a guideline for the proper selection of a radionuclide and the appropriate labeling strategy to access a tracer with optimal imaging qualities. Finally, we summarize the methodology of their evaluation in in vitro and animal models and the road to clinical translation. This review is intended to be a primer for newcomers to the field and give insight into the workflow of developing radiopharmaceuticals.
Collapse
Affiliation(s)
- Joseph Lau
- Laboratory of Molecular Imaging and Nanomedicine (LOMIN), National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD 20892, USA;
| | - Etienne Rousseau
- Department of Nuclear Medicine and Radiobiology, University of Sherbrooke, Sherbrooke, QC J1H 5N4, Canada;
| | - Daniel Kwon
- Department of Molecular Oncology, BC Cancer, Vancouver, BC V5Z 1L3, Canada; (D.K.); (K.-S.L.); (F.B.)
| | - Kuo-Shyan Lin
- Department of Molecular Oncology, BC Cancer, Vancouver, BC V5Z 1L3, Canada; (D.K.); (K.-S.L.); (F.B.)
| | - François Bénard
- Department of Molecular Oncology, BC Cancer, Vancouver, BC V5Z 1L3, Canada; (D.K.); (K.-S.L.); (F.B.)
| | - Xiaoyuan Chen
- Laboratory of Molecular Imaging and Nanomedicine (LOMIN), National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD 20892, USA;
| |
Collapse
|
32
|
Hao D, Fan Y, Xiao W, Liu R, Pivetti C, Walimbe T, Guo F, Zhang X, Farmer DL, Wang F, Panitch A, Lam KS, Wang A. Rapid endothelialization of small diameter vascular grafts by a bioactive integrin-binding ligand specifically targeting endothelial progenitor cells and endothelial cells. Acta Biomater 2020; 108:178-193. [PMID: 32151698 DOI: 10.1016/j.actbio.2020.03.005] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 02/29/2020] [Accepted: 03/03/2020] [Indexed: 12/31/2022]
Abstract
Establishing and maintaining a healthy endothelium on vascular and intravascular devices is crucial for the prevention of thrombosis and stenosis. Generating a biofunctional surface on vascular devices to recruit endothelial progenitor cells (EPCs) and endothelial cells (ECs) has proven efficient in promoting in situ endothelialization. However, molecules conventionally used for EPC/EC capturing generally lack structural stability, capturing specificity, and biological functionalities, which have limited their applications. Discovery of effective, specific, and structurally stable EPC/EC capturing ligands is desperately needed. Using the high-throughput One-Bead One-Compound combinatorial library screening technology, we recently identified a disulfide cyclic octa-peptide LXW7 (cGRGDdvc), which possesses strong binding affinity and functionality to EPCs/ECs, weak binding to platelets, and no binding to inflammatory cells. Because LXW7 is cyclic and 4 out of the 8 amino acids are unnatural D-amino acids, LXW7 is highly proteolytically stable. In this study, we applied LXW7 to modify small diameter vascular grafts using a Click chemistry approach. In vitro studies demonstrated that LXW7-modified grafts significantly improved EPC attachment, proliferation and endothelial differentiation and suppressed platelet attachment. In a rat carotid artery bypass model, LXW7 modification of the small diameter vascular grafts significantly promoted EPC/EC recruitment and rapidly achieved endothelialization. At 6 weeks after implantation, LXW7-modified grafts retained a high patency of 83%, while the untreated grafts had a low patency of 17%. Our results demonstrate that LXW7 is a potent EPC/EC capturing and platelet suppressing ligand and LXW7-modified vascular grafts rapidly generate a healthy and stable endothelial interface between the graft surface and the circulation to reduce thrombosis and improve patency. STATEMENT OF SIGNIFICANCE: In this study, One-Bead One-Compound (OBOC) technology has been applied for the first time in discovering bioactive ligands for tissue regeneration applications. Current molecules used to modify artificial vascular grafts generally lack EPC/EC capturing specificity, biological functionalities and structural stability. Using OBOC technology, we identified LXW7, a constitutionally stable disulfide cyclic octa-peptide with strong binding affinity and biological functionality to EPCs/ECs, very weak binding to platelets and no binding to inflammatory cells. These characteristics are crucial for promoting rapid endothelialization to prevent thrombosis and improve patency of vascular grafts. LXW7 coating technology could be applied to a wide range of vascular and intravascular devices, including grafts, stents, cardiac valves, and catheters, where a "living" endothelium and healthy blood interface are needed.
Collapse
|
33
|
Sandomenico A, Caporale A, Doti N, Cross S, Cruciani G, Chambery A, De Falco S, Ruvo M. Synthetic Peptide Libraries: From Random Mixtures to In Vivo Testing. Curr Med Chem 2020; 27:997-1016. [PMID: 30009695 DOI: 10.2174/0929867325666180716110833] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2018] [Revised: 06/22/2018] [Accepted: 06/29/2018] [Indexed: 01/13/2023]
Abstract
Combinatorially generated molecular repertoires have been largely used to identify novel bioactive compounds. Ever more sophisticated technological solutions have been proposed to simplify and speed up such process, expanding the chemical diversity space and increasing the prospect to select new molecular entities with specific and potent activities against targets of therapeutic relevance. In this context, random mixtures of oligomeric peptides were originally used and since 25 years they represent a continuous source of bioactive molecules with potencies ranging from the sub-nM to microM concentration. Synthetic peptide libraries are still employed as starting "synthetic broths" of structurally and chemically diversified molecular fragments from which lead compounds can be extracted and further modified. Thousands of studies have been reported describing the application of combinatorial mixtures of synthetic peptides with different complexity and engrafted on diverse structural scaffolds for the identification of new compounds which have been further developed and also tested in in vivo models of relevant diseases. We briefly review some of the most used methodologies for library preparation and screening and the most recent case studies appeared in the literature where compounds have reached at least in vivo testing in animal or similar models. Recent technological advancements in biotechnology, engineering and computer science have suggested new options to facilitate the discovery of new bioactive peptides. In this instance, we anticipate here a new approach for the design of simple but focused tripeptide libraries against druggable cavities of therapeutic targets and its complementation with existing approaches.
Collapse
Affiliation(s)
- Annamaria Sandomenico
- Istituto di Biostrutture e Bioimmagini del CNR and CIRPeB, Universita Federico II di Napoli, via Mezzocannone, 16, 80134 Napoli, Italy
| | - Andrea Caporale
- Istituto di Biostrutture e Bioimmagini del CNR and CIRPeB, Universita Federico II di Napoli, via Mezzocannone, 16, 80134 Napoli, Italy
| | - Nunzianna Doti
- Istituto di Biostrutture e Bioimmagini del CNR and CIRPeB, Universita Federico II di Napoli, via Mezzocannone, 16, 80134 Napoli, Italy
| | - Simon Cross
- Molecular Discovery Ltd, Unit 501 Centennial Park, Centennial Avenue Elstree, Borehamwood, Hertfordshire WD6 3FG, United Kingdom
| | - Gabriele Cruciani
- Molecular Discovery Ltd, Unit 501 Centennial Park, Centennial Avenue Elstree, Borehamwood, Hertfordshire WD6 3FG, United Kingdom.,Dipartimento di Chimica, Biologia e Biotecnologia, Via Elce di Sotto 8, 06123 Perugia, Italy
| | - Angela Chambery
- Dipartimento di Scienze e Tecnologie Ambientali, Biologiche e Farmaceutiche, Università della Campania "Luigi Vanvitelli", via Vivaldi, 43, 81100 Caserta, Italy
| | - Sandro De Falco
- Istituto di Genetica e Biofisica del CNR, via Pietro Castellino, 111, 80131, Napoli, Italy
| | - Menotti Ruvo
- Istituto di Biostrutture e Bioimmagini del CNR and CIRPeB, Universita Federico II di Napoli, via Mezzocannone, 16, 80134 Napoli, Italy
| |
Collapse
|
34
|
Murrell E, Luyt LG. Incorporation of Fluorine into an OBOC Peptide Library by Copper-Free Click Chemistry toward the Discovery of PET Imaging Agents. ACS COMBINATORIAL SCIENCE 2020; 22:109-113. [PMID: 32011850 DOI: 10.1021/acscombsci.9b00146] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
A one-bead one-compound (OBOC) library of peptide-based imaging agents was developed where a 19F-containing moiety was added onto the N-terminus of octamer peptides through copper-free click chemistry prior to screening of the library. This created a library of complete imaging agents that was screened against CXCR4, a receptor of interest for cancer imaging. The screen directly resulted in the discovery of a peptide-based imaging agent with an IC50 of 138 μM. This proof-of-concept study describes a new type of OBOC peptide library design, where hits discovered from screening can be easily translated into their fluorine-18 counterpart for PET imaging without loss of affinity.
Collapse
Affiliation(s)
- Emily Murrell
- Department of Chemistry, University of Western Ontario, 1151 Richmond Street, London, Ontario N6A 5B7, Canada
| | - Leonard G. Luyt
- Department of Chemistry, University of Western Ontario, 1151 Richmond Street, London, Ontario N6A 5B7, Canada
- Departments of Oncology and Medical Imaging, University of Western Ontario, 1151 Richmond Street, London, Ontario N6A 5B7, Canada
- London Regional Cancer Program, Lawson Health Research Institute, 790 Commissioners Road East, London, Ontario N6A 4L6, Canada
| |
Collapse
|
35
|
Hao D, Ma B, He C, Liu R, Farmer DL, Lam KS, Wang A. Surface modification of polymeric electrospun scaffolds via a potent and high-affinity integrin α4β1 ligand improved the adhesion, spreading and survival of human chorionic villus-derived mesenchymal stem cells: a new insight for fetal tissue engineering. J Mater Chem B 2020; 8:1649-1659. [PMID: 32011618 PMCID: PMC7353926 DOI: 10.1039/c9tb02309g] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Cell-biomaterial interactions are primarily governed by cell adhesion, which arises from the binding of cellular integrins to the extracellular matrix (ECM). Integrins drive the assembly of focal contacts that serve as mechanotransducers and signaling nexuses for stem cells, for example integrin α4β1 plays pivotal roles in regulating mesenchymal stem cell (MSC) homing, adhesion, migration and differentiation. The strategy to control the integrin-mediated cell adhesion to bioinspired, ECM-mimicking materials is essential to regulate cell functions and tissue regeneration. Previously, using one-bead one-compound (OBOC) combinatorial technology, we discovered that LLP2A was a high-affinity peptidomimetic ligand (IC50 = 2 pM) against integrin α4β1. In this study, we identified that LLP2A had a strong binding to human early gestation chorionic villi-derived MSCs (CV-MSCs) via integrin α4β1. To improve CV-MSC seeding, expansion and delivery for regenerative applications, we constructed artificial scaffolds simulating the structure of the native ECM by immobilizing LLP2A onto the scaffold surface as cell adhesion sites. LLP2A modification significantly enhanced CV-MSC adhesion, spreading and viability on the polymeric scaffolds via regulating signaling pathways including phosphorylation of focal adhesion kinase (FAK), and AKT, NF-kB and Caspase 9. In addition, we also demonstrated that LLP2A had strong binding to MSCs of other sources, such as bone marrow-derived mesenchymal stem cells (BM-MSCs) and adipose tissue-derived mesenchymal stem cells (AT-MSCs). Therefore, LLP2A and its derivatives not only hold great promise for improving CV-MSC-mediated treatment of fetal diseases, but they can also be widely applied to functionalize various biological and medical materials, which are in need of MSC recruitment, enrichment and survival, for regenerative medicine applications.
Collapse
Affiliation(s)
- Dake Hao
- Department of Surgery, School of Medicine, University of California Davis, Sacramento, CA 95817, USA. and Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, CA 95817, USA
| | - Bowen Ma
- Department of Surgery, School of Medicine, University of California Davis, Sacramento, CA 95817, USA.
| | - Chuanchao He
- Department of Surgery, School of Medicine, University of California Davis, Sacramento, CA 95817, USA.
| | - Ruiwu Liu
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California Davis, Sacramento, CA 95817, USA
| | - Diana L Farmer
- Department of Surgery, School of Medicine, University of California Davis, Sacramento, CA 95817, USA. and Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, CA 95817, USA
| | - Kit S Lam
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California Davis, Sacramento, CA 95817, USA
| | - Aijun Wang
- Department of Surgery, School of Medicine, University of California Davis, Sacramento, CA 95817, USA. and Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, CA 95817, USA and Department of Biomedical Engineering, University of California Davis, Davis, CA 95616, USA
| |
Collapse
|
36
|
Gai Y, Yuan L, Sun L, Li H, Li M, Fang H, Altine B, Liu Q, Zhang Y, Zeng D, Lan X. Comparison of Al 18F- and 68Ga-labeled NOTA-PEG 4-LLP2A for PET imaging of very late antigen-4 in melanoma. J Biol Inorg Chem 2020; 25:99-108. [PMID: 31745667 PMCID: PMC7067668 DOI: 10.1007/s00775-019-01742-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Accepted: 11/06/2019] [Indexed: 12/31/2022]
Abstract
Malignant melanoma is an aggressive cancer with poor prognosis. Very late antigen-4 (VLA-4) is overexpressed in melanoma and many other tumors, making it an attractive target for developing molecular diagnostic and therapeutic agents. We compared Al18F- and 68Ga-labeled LLP2A peptides for PET imaging of VLA-4 expression in melanoma. The peptidomimetic ligand LLP2A was modified with chelator 2-S-(4-isothiocyanatobenzyl)-1,4,7-triazacyclononane-1,4,7-triacetic acid (p-SCN-Bn-NOTA), and the resulting NOTA-PEG4-LLP2A peptide was then radiolabeled with Al18F or 68Ga. The two labeled peptides were assayed for in vitro and in vivo VLA-4 targeting efficiency. Good Al18F and 68Ga radiolabeling yields were achieved, and the resulting PET tracers showed good serum stability. In the in vivo evaluation of the B16F10 xenograft mouse model, both tracers exhibited high accumulation with good contrast in static PET images. Compared with 68Ga-NOTA-PEG4-LLP2A, Al18F-NOTA-PEG4-LLP2A resulted in relatively higher background, including higher liver uptake (1 h: 20.1 ± 2.6 vs. 15.3 ± 1.7%ID/g, P < 0.05; 2 h: 11.0 ± 1.2 vs. 8.0 ± 0.8%ID/g, P < 0.05) and lower tumor-to-blood ratios (2.5 ± 0.4 vs. 3.3 ± 0.5 at 1 h, P < 0.05; 5.1 ± 0.9 vs. 7.3 ± 0.6 at 2 h, P < 0.01) at some time points. The results obtained from the mice blocked with unlabeled peptides and VLA-4-negative A375 xenografts groups confirmed the high specificity of the developed tracers. Despite the relatively high liver uptake, both Al18F-NOTA-PEG4-LLP2A and 68Ga-NOTA-PEG4-LLP2A exhibited high VLA-4 targeting efficacy with comparable in vivo performance, rendering them promising candidates for imaging tumors that overexpress VLA-4.
Collapse
Affiliation(s)
- Yongkang Gai
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Lujie Yuan
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Lingyi Sun
- Center for Radiochemistry Research, Department of Diagnostic Radiology, Oregon Health and Science University, Portland, OR, 97239, USA
| | - Huiling Li
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Mengting Li
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Hanyi Fang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Bouhari Altine
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Qingyao Liu
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Yongxue Zhang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Dexing Zeng
- Center for Radiochemistry Research, Department of Diagnostic Radiology, Oregon Health and Science University, Portland, OR, 97239, USA.
| | - Xiaoli Lan
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China.
| |
Collapse
|
37
|
Ramasubramanian L, Kumar P, Wang A. Engineering Extracellular Vesicles as Nanotherapeutics for Regenerative Medicine. Biomolecules 2019; 10:E48. [PMID: 31905611 PMCID: PMC7023093 DOI: 10.3390/biom10010048] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 12/24/2019] [Accepted: 12/26/2019] [Indexed: 01/01/2023] Open
Abstract
Long thought of to be vesicles that primarily recycled waste biomolecules from cells, extracellular vesicles (EVs) have now emerged as a new class of nanotherapeutics for regenerative medicine. Recent studies have proven their potential as mediators of cell proliferation, immunomodulation, extracellular matrix organization and angiogenesis, and are currently being used as treatments for a variety of diseases and injuries. They are now being used in combination with a variety of more traditional biomaterials and tissue engineering strategies to stimulate tissue repair and wound healing. However, the clinical translation of EVs has been greatly slowed due to difficulties in EV isolation and purification, as well as their limited yields and functional heterogeneity. Thus, a field of EV engineering has emerged in order to augment the natural properties of EVs and to recapitulate their function in semi-synthetic and synthetic EVs. Here, we have reviewed current technologies and techniques in this growing field of EV engineering while highlighting possible future applications for regenerative medicine.
Collapse
Affiliation(s)
- Lalithasri Ramasubramanian
- Surgical Bioengineering Laboratory, Department of Surgery, School of Medicine, University of California–Davis, Sacramento, CA 95817, USA (P.K.)
- Department of Biomedical Engineering, University of California–Davis, Davis, CA 95616, USA
| | - Priyadarsini Kumar
- Surgical Bioengineering Laboratory, Department of Surgery, School of Medicine, University of California–Davis, Sacramento, CA 95817, USA (P.K.)
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children–Northern California, Sacramento, CA 95817, USA
| | - Aijun Wang
- Surgical Bioengineering Laboratory, Department of Surgery, School of Medicine, University of California–Davis, Sacramento, CA 95817, USA (P.K.)
- Department of Biomedical Engineering, University of California–Davis, Davis, CA 95616, USA
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children–Northern California, Sacramento, CA 95817, USA
| |
Collapse
|
38
|
Lee SB, Han YR, Jeon HJ, Jun CH, Kim SK, Chin J, Lee SJ, Jeong M, Lee JE, Lee CH, Cho SJ, Kim DS, Jeon YH. Medical fluorophore 1 (MF1), a benzoquinolizinium-based fluorescent dye, as an inflammation imaging agent. J Mater Chem B 2019; 7:7326-7331. [PMID: 31681930 DOI: 10.1039/c9tb01266d] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Structure-based targeting of fluorescent dyes is essential for their use as imaging agents for disease diagnosis. Here, we describe the development of the benzoquinolizinium compound Medical fluorophore 1 (MF1) as a novel biomedical imaging agent that allows the visualization of inflammation by virtue of its unique chemical structure. Lipopolysaccharide treatment stimulated the uptake of MF1 by bone marrow-derived macrophages, with no adverse effects on cell proliferation. In vivo fluorescence lifetime imaging revealed the accumulation of MF1 in carrageenan-induced acute inflammatory lesions in mice, which peaked at 6 h. MF1-based imaging also allowed monitoring of the response to the anti-inflammatory drugs dexamethasone and sulfasalazine. Thus, MF1 can be used to diagnose diseases characterized by inflammation as well as treatment efficacy.
Collapse
Affiliation(s)
- Sang Bong Lee
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, 80 Chembok-ro Dong-gu Daegu, Republic of Korea.
| | - Ye Ri Han
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, 80 Chembok-ro Dong-gu Daegu, Republic of Korea.
| | - Hui-Jeon Jeon
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, 80 Chembok-ro Dong-gu Daegu, Republic of Korea.
| | - Chul-Ho Jun
- Department of Chemistry, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea and Center for NanoMedicine, Institute for Basic Science (IBS), Seoul, Republic of Korea and Yonsei-IBS Institute, Yonsei University, Seoul 03722, Republic of Korea
| | - Sang-Kyoon Kim
- Laboratory Animal Center, Daegu-Gyeongbuk Medical Innovation Foundation, 80 Chembok-ro Dong-gu Daegu, Republic of Korea.
| | - Jungwook Chin
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, 80 Chembok-ro Dong-gu Daegu, Republic of Korea.
| | - Su-Jeong Lee
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, 80 Chembok-ro Dong-gu Daegu, Republic of Korea.
| | - Minseon Jeong
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, 80 Chembok-ro Dong-gu Daegu, Republic of Korea.
| | - Jae-Eon Lee
- Laboratory Animal Center, Daegu-Gyeongbuk Medical Innovation Foundation, 80 Chembok-ro Dong-gu Daegu, Republic of Korea. and Department of Biomaterials Science, College of Natural Resources and Life Science/Life and Industry Convergence Research Institute, Pusan National University, Pusan, Republic of Korea
| | - Chang-Hee Lee
- Department of Chemistry, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea and Center for NanoMedicine, Institute for Basic Science (IBS), Seoul, Republic of Korea and Yonsei-IBS Institute, Yonsei University, Seoul 03722, Republic of Korea
| | - Sung Jin Cho
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, 80 Chembok-ro Dong-gu Daegu, Republic of Korea.
| | - Dong-Su Kim
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, 80 Chembok-ro Dong-gu Daegu, Republic of Korea.
| | - Yong Hyun Jeon
- Laboratory Animal Center, Daegu-Gyeongbuk Medical Innovation Foundation, 80 Chembok-ro Dong-gu Daegu, Republic of Korea.
| |
Collapse
|
39
|
Marusina AI, Merleev AA, Luna JI, Olney L, Haigh NE, Yoon D, Guo C, Ovadia EM, Shimoda M, Luxardi G, Boddu S, Lal NN, Takada Y, Lam KS, Liu R, Isseroff RR, Le S, Nolta JA, Kloxin AM, Maverakis E. Tunable hydrogels for mesenchymal stem cell delivery: Integrin-induced transcriptome alterations and hydrogel optimization for human wound healing. Stem Cells 2019; 38:231-245. [PMID: 31648388 DOI: 10.1002/stem.3105] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 09/08/2019] [Accepted: 09/18/2019] [Indexed: 12/11/2022]
Abstract
Therapeutic applications for mesenchymal stem/stromal cells (MSCs) are growing; however, the successful implementation of these therapies requires the development of appropriate MSC delivery systems. Hydrogels are ideally suited to cultivate MSCs but tuning hydrogel properties to match their specific in vivo applications remains a challenge. Thus, further characterization of how hydrogel-based delivery vehicles broadly influence MSC function and fate will help lead to the next generation of more intelligently designed delivery vehicles. To date, few attempts have been made to comprehensively characterize hydrogel impact on the MSC transcriptome. Herein, we have synthesized cell-degradable hydrogels based on bio-inert poly(ethylene glycol) tethered with specific integrin-binding small molecules and have characterized their resulting effect on the MSC transcriptome when compared with 2D cultured and untethered 3D hydrogel cultured MSCs. The 3D culture systems resulted in alterations in the MSC transcriptome, as is evident by the differential expression of genes related to extracellular matrix production, glycosylation, metabolism, signal transduction, gene epigenetic regulation, and development. For example, genes important for osteogenic differentiation were upregulated in 3D hydrogel cultures, and the expression of these genes could be partially suppressed by tethering an integrin-binding RGD peptide within the hydrogel. Highlighting the utility of tunable hydrogels, when applied to ex vivo human wounds the RGD-tethered hydrogel was able to support wound re-epithelialization, possibly due to its ability to increase PDGF expression and decrease IL-6 expression. These results will aid in future hydrogel design for a broad range of applications.
Collapse
Affiliation(s)
- Alina I Marusina
- Department of Dermatology, University of California Davis School of Medicine, Sacramento, California
| | - Alexander A Merleev
- Department of Dermatology, University of California Davis School of Medicine, Sacramento, California
| | - Jesus I Luna
- Department of Dermatology, University of California Davis School of Medicine, Sacramento, California
| | - Laura Olney
- Department of Dermatology, University of California Davis School of Medicine, Sacramento, California
| | - Nathan E Haigh
- Department of Dermatology, University of California Davis School of Medicine, Sacramento, California
| | - Daniel Yoon
- Department of Dermatology, University of California Davis School of Medicine, Sacramento, California
| | - Chen Guo
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, Delaware
| | - Elisa M Ovadia
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, Delaware
| | - Michiko Shimoda
- Department of Dermatology, University of California Davis School of Medicine, Sacramento, California
| | - Guillaume Luxardi
- Department of Dermatology, University of California Davis School of Medicine, Sacramento, California
| | - Sucharita Boddu
- Department of Dermatology, University of California Davis School of Medicine, Sacramento, California
| | - Nelvish N Lal
- Department of Dermatology, University of California Davis School of Medicine, Sacramento, California
| | - Yoshikazu Takada
- Department of Dermatology, University of California Davis School of Medicine, Sacramento, California
| | - Kit S Lam
- Department of Biochemistry and Molecular Medicine, University of California Davis School of Medicine, Sacramento, California
| | - Ruiwu Liu
- Department of Biochemistry and Molecular Medicine, University of California Davis School of Medicine, Sacramento, California
| | - R Rivkah Isseroff
- Department of Dermatology, University of California Davis School of Medicine, Sacramento, California
| | - Stephanie Le
- Department of Dermatology, University of California Davis School of Medicine, Sacramento, California
| | - Jan A Nolta
- Stem Cell Program and Institute for Regenerative Cures, University of California Davis, Sacramento, California
| | - April M Kloxin
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, Delaware.,Department of Materials Science and Engineering, University of Delaware, Newark, Delaware
| | - Emanual Maverakis
- Department of Dermatology, University of California Davis School of Medicine, Sacramento, California
| |
Collapse
|
40
|
Hwang D, Tsuji K, Park H, Burke TR, Rader C. Site-Specific Lysine Arylation as an Alternative Bioconjugation Strategy for Chemically Programmed Antibodies and Antibody-Drug Conjugates. Bioconjug Chem 2019; 30:2889-2896. [PMID: 31675216 DOI: 10.1021/acs.bioconjchem.9b00609] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
By exploiting a uniquely reactive lysine residue (Lys99) for site-specific attachment of small molecules, the humanized catalytic antibody h38C2 has been used as bioconjugation module in the assembly of chemically programmed antibodies and antibody-drug conjugates. Treatment of h38C2 with β-lactam-functionalized small molecules has been previously shown to result in covalent conjugation by selective formation of a stable amide bond with the ε-amino group of the Lys99 residue. Here we report that heteroaryl methylsulfonyl (MS-PODA)-functionalized small molecules represent an alternative bioconjugation strategy through highly efficient, site-specific, and stable arylation of the Lys99 residue. A set of chemically programmed antibodies and antibody-drug conjugates assembled by Lys99 arylation provided proof-of-concept for the therapeutic utility of this alternative bioconjugation strategy. While being equally effective as β-lactam-functionalized ligands for bioconjugation with catalytic antibody h38C2, the MS-PODA moiety offers distinct synthetic advantages, making it highly attractive.
Collapse
Affiliation(s)
| | - Kohei Tsuji
- Chemical Biology Laboratory, Center for Cancer Research , National Cancer Institute, National Institutes of Health , Frederick , Maryland 21702 , United States
| | | | - Terrence R Burke
- Chemical Biology Laboratory, Center for Cancer Research , National Cancer Institute, National Institutes of Health , Frederick , Maryland 21702 , United States
| | | |
Collapse
|
41
|
Raymond AC, Gao B, Girard L, Minna JD, Gomika Udugamasooriya D. Unbiased peptoid combinatorial cell screen identifies plectin protein as a potential biomarker for lung cancer stem cells. Sci Rep 2019; 9:14954. [PMID: 31628412 PMCID: PMC6802198 DOI: 10.1038/s41598-019-51004-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 09/20/2019] [Indexed: 01/16/2023] Open
Abstract
Tumors often contain a small subset of drug-resisting, self-renewing, and highly metastatic cells called tumor initiating cells or cancer stem cells (CSCs). To develop new approaches to detecting and targeting lung cancer CSCs, we applied an "unbiased" peptoid combinatorial cell screen to identify highly specific ligands that bind a CSC subpopulation of non-small cell lung cancer cells (defined by Aldefluor positivity), but not the remaining aldefluor negative cancer cells from the same preclinical model. One of the 'hit' peptoids bound to plectin, a structural protein, predominantly expressed intracellularly, but whose localization on the cell surface is linked to tumor invasion and metastasis. Our studies show both genotypic and phenotypic correlations between plectin and lung CSCs, as well as association of high plectin mRNA expression with poor patient survival in lung adenocarcinoma, potentially identifying plectin as a biomarker for lung CSCs.
Collapse
Affiliation(s)
- Aaron C Raymond
- Department of Pharmacological & Pharmaceutical Sciences, University of Houston, 4849 Calhoun Rd, Houston, TX, 77204-5037, USA
| | - Boning Gao
- Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd., Dallas, TX, 75390, USA
- Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd., Dallas, TX, 75390, USA
- Departments of Pharmacology, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd., Dallas, TX, 75390, USA
| | - Luc Girard
- Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd., Dallas, TX, 75390, USA
- Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd., Dallas, TX, 75390, USA
- Departments of Pharmacology, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd., Dallas, TX, 75390, USA
| | - John D Minna
- Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd., Dallas, TX, 75390, USA
- Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd., Dallas, TX, 75390, USA
- Departments of Pharmacology, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd., Dallas, TX, 75390, USA
- Internal Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA
| | - D Gomika Udugamasooriya
- Department of Pharmacological & Pharmaceutical Sciences, University of Houston, 4849 Calhoun Rd, Houston, TX, 77204-5037, USA.
- Department of Cancer Systems Imaging, MD Anderson Cancer Center, 1881 East Road, Houston, TX, 77030-4009, USA.
| |
Collapse
|
42
|
Multiplexed Competitive Screening of One-Bead-One-Component Combinatorial Libraries Using a ClonePix 2 Colony Sorter. Int J Mol Sci 2019; 20:ijms20205119. [PMID: 31623061 PMCID: PMC6830312 DOI: 10.3390/ijms20205119] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 10/12/2019] [Accepted: 10/13/2019] [Indexed: 12/24/2022] Open
Abstract
Screening solid-phase combinatorial libraries of bioactive compounds against fluorescently labeled target biomolecules is an established technology in ligand and drug discovery. Rarely, however, do screening methods include comprehensive strategies-beyond mere library blocking and competitive screening-to ensure binding selectivity of selected leads. This work presents a method for multiplexed solid-phase peptide library screening using a ClonePix 2 Colony Picker that integrates (i) orthogonal fluorescent labeling for positive selection against a target protein and negative selection against competitor species with (ii) semi-quantitative tracking of target vs. competitor binding for every library bead. The ClonePix 2 technology enables global at-a-glance evaluation and customization of the parameters for bead selection to ensure high affinity and selectivity of the isolated leads. A case study is presented by screening a peptide library against green-labeled human immunoglobulin G (IgG) and red-labeled host cell proteins (HCPs) using ClonePix 2 to select HCP-binding ligands for flow-through chromatography applications. Using this approach, 79 peptide ligand candidates (6.6% of the total number of ligands screened) were identified as potential HCP-selective ligands, enabling a potential rate of >3,000 library beads screened per hour.
Collapse
|
43
|
Yang F, Xiao W, Liu Y, Liu R, Kramer R, Li X, Ajena Y, Baehr CM, Rojalin T, Zhang H, Lam KS. One-bead one-compound combinatorial library derived targeting ligands for detection and treatment of oral squamous cancer. Oncotarget 2019; 10:5468-5479. [PMID: 31534631 PMCID: PMC6739215 DOI: 10.18632/oncotarget.27189] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Accepted: 08/12/2019] [Indexed: 12/12/2022] Open
Abstract
Oral squamous cancers (OSC) are hallmarked by poor prognosis, delayed clinical detection, and a lack of defined, characteristic biomarkers. By screening combinatorial one-bead one-compound (OBOC) peptide libraries against oral squamous cancer cell lines, two cyclic peptide ligands, LLY12 and LLY13 were previously identified. These ligands are capable of specific binding to the oral cancer cell lines (MOK-101, HSC-3, SCC-4 and SCC-10a) but not non-cancerous keratinocytes, leukocytes, fibroblast, and endothelial cells. These two peptides were synthesized and evaluated for their binding property, cytotoxicity and cell permeability. In vitro studies indicate that both LLY12 and LLY13 were able to bind to oral cancer cells with high specificity but did not show any cytotoxicity against human keratinocytes. Biotinylated LLY13, in complex with streptavidin-alexa488 was taken up by live oral cancer cells, thus rendering it as an excellent candidate vehicle for efficient delivery of drug loaded-nanoparticles. In vivo and ex vivo near infra-red fluorescence imaging studies confirmed the in vivo targeting efficiency and specificity of LLY13 in oral cancer orthotopic murine xenograft model. In vivo studies also showed that LLY13 was able to accumulate in the OSC tumors and demarcate the tumor margins in orthotopic xenograft model. Together, our data supports LLY13 as a promising theranostic agent against OSC.
Collapse
Affiliation(s)
- Fan Yang
- Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Boston, MA, USA
| | - Wenwu Xiao
- Department of Biochemistry and Molecular Medicine, University of California, Davis, CA, USA
| | - Yanlei Liu
- Department of Pathology, University of California Davis Medical Center, Sacramento, CA, USA
| | - Ruiwu Liu
- Department of Biochemistry and Molecular Medicine, University of California, Davis, CA, USA
| | - Randall Kramer
- Department of Cell and Tissue Biology, University of California, San Francisco, CA, USA
| | - Xiaocen Li
- Department of Biochemistry and Molecular Medicine, University of California, Davis, CA, USA
| | - Yousif Ajena
- Department of Biochemistry and Molecular Medicine, University of California, Davis, CA, USA
| | - Christopher M Baehr
- Department of Biochemistry and Molecular Medicine, University of California, Davis, CA, USA
| | - Tatu Rojalin
- Department of Biochemistry and Molecular Medicine, University of California, Davis, CA, USA
| | - Hongyong Zhang
- Department of Biochemistry and Molecular Medicine, University of California, Davis, CA, USA
| | - Kit S Lam
- Department of Biochemistry and Molecular Medicine, University of California, Davis, CA, USA
| |
Collapse
|
44
|
Qi J, Hymel D, Nelson CG, Burke TR, Rader C. Conventional and Chemically Programmed Asymmetric Bispecific Antibodies Targeting Folate Receptor 1. Front Immunol 2019; 10:1994. [PMID: 31497024 PMCID: PMC6712926 DOI: 10.3389/fimmu.2019.01994] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Accepted: 08/07/2019] [Indexed: 12/19/2022] Open
Abstract
T-cell engaging bispecific antibodies (biAbs) can mediate potent and specific tumor cell eradication in liquid cancers. Substantial effort has been invested in expanding this concept to solid cancers. To explore their utility in the treatment of ovarian cancer, we built a set of asymmetric biAbs in IgG1-like format that bind CD3 on T cells with a conventional scFv arm and folate receptor 1 (FOLR1) on ovarian cancer cells with a conventional or a chemically programmed Fab arm. For avidity engineering, we also built an asymmetric biAb format with a tandem Fab arm. We show that both conventional and chemically programmed CD3 × FOLR1 biAbs exert specific in vitro and in vivo cytotoxicity toward FOLR1-expressing ovarian cancer cells by recruiting and activating T cells. While the conventional T-cell engaging biAb was curative in an aggressive mouse model of human ovarian cancer, the potency of the chemically programmed biAb was significantly boosted by avidity engineering. Both conventional and chemically programmed CD3 × FOLR1 biAbs warrant further investigation for ovarian cancer immunotherapy.
Collapse
Affiliation(s)
- Junpeng Qi
- Department of Immunology and Microbiology, The Scripps Research Institute, Jupiter, FL, United States
| | - David Hymel
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, United States
| | - Christopher G Nelson
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, United States
| | - Terrence R Burke
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, United States
| | - Christoph Rader
- Department of Immunology and Microbiology, The Scripps Research Institute, Jupiter, FL, United States
| |
Collapse
|
45
|
Baiula M, Spampinato S, Gentilucci L, Tolomelli A. Novel Ligands Targeting α 4β 1 Integrin: Therapeutic Applications and Perspectives. Front Chem 2019; 7:489. [PMID: 31338363 PMCID: PMC6629825 DOI: 10.3389/fchem.2019.00489] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 06/25/2019] [Indexed: 12/13/2022] Open
Abstract
Among the other members of the adhesion molecules' family, α4β1 integrin, a heterodimeric receptor, plays a crucial role in inflammatory diseases, cancer development, metastasis and stem cell mobilization or retention. In many cases, its function in pathogenesis is not yet completely understood and investigations on ligand binding and related stabilization of active/inactive conformations still represent an important goal. For this reason, starting from the highlight of α4β1 functions in human pathologies, we report an overview of synthetic α4β1 integrin ligands under development as potential therapeutic agents. The small molecule library that we have selected represents a collection of lead compounds. These molecules are the object of future refinement in academic and industrial research, in order to achieve a fine tuning of α4β1 integrin regulation for the development of novel agents against pathologies still eluding an effective solution.
Collapse
Affiliation(s)
- Monica Baiula
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Santi Spampinato
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Luca Gentilucci
- Department of Chemistry “G. Ciamician,” University of Bologna, Bologna, Italy
| | | |
Collapse
|
46
|
Hibbitts A, O’Connor AM, McCarthy J, Forde ÉB, Hessman G, O’Driscoll CM, Cryan SA, Devocelle M. Poly(ethylene glycol)-Based Peptidomimetic "PEGtide" of Oligo-Arginine Allows for Efficient siRNA Transfection and Gene Inhibition. ACS OMEGA 2019; 4:10078-10088. [PMID: 31460100 PMCID: PMC6647993 DOI: 10.1021/acsomega.9b00265] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 05/29/2019] [Indexed: 06/10/2023]
Abstract
While a wide range of experimental and commercial transfection reagents are currently available, persistent problems remain regarding their suitability for continued development. These include the transfection efficiency for difficult-to-transfect cell types and the risks of decreased cell viability that may arise from any transfection that does occur. Therefore, research is now turning toward alternative molecules that improve the toxicity profile of the gene delivery vector (GDV), while maintaining the transfection efficiency. Among them, cell-penetrating peptides, such as octa-arginine, have shown significant potential as GDVs. Their pharmacokinetic and pharmacodynamic properties can be enhanced through peptidomimetic conversion, whereby a peptide is modified into a synthetic analogue that mimics its structure and/or function, but whose backbone is not solely based on α-amino acids. Using this technology, novel peptidomimetics were developed by co- and postpolymerization functionalization of substituted ethylene oxides, producing poly(ethylene glycol) (PEG)-based peptidomimetics termed "PEGtides". Specifically, a PEGtide of the poly(α-amino acid) oligo-arginine [poly(glycidylguanidine)] was assessed for its ability to complex and deliver a small interfering ribonucleic acid (siRNA) using a range of cell assays and high-content analysis. PEGtide-siRNA demonstrated significantly increased internalization and gene inhibition over 24 h in Calu-3 pulmonary epithelial cells compared to commercial controls and octa-arginine-treated samples, with no evidence of toxicity. Furthermore, PEGtide-siRNA nanocomplexes can provide significant levels of gene inhibition in "difficult-to-transfect" mouse embryonic hypothalamic (mHypo N41) cells. Overall, the usefulness of this novel PEGtide for gene delivery was clearly demonstrated, establishing it as a promising candidate for continued translational research.
Collapse
Affiliation(s)
- Alan Hibbitts
- Tissue
Engineering Research Group, Department of Anatomy, Department of Chemistry, and Drug Delivery
& Advanced Materials Team, School of Pharmacy, Royal College of Surgeons in Ireland (RCSI), 123 St. Stephen’s Green, Dublin 2, Ireland
- Trinity Centre for Biomedical Engineering and School of Chemistry, Trinity College Dublin (TCD), The University of Dublin, College Green, Dublin 2, Ireland
- Advanced
Materials and Bioengineering Research (AMBER) Centre, RCSI & TCD, Dublin 2, Ireland
| | - Aoife M. O’Connor
- Tissue
Engineering Research Group, Department of Anatomy, Department of Chemistry, and Drug Delivery
& Advanced Materials Team, School of Pharmacy, Royal College of Surgeons in Ireland (RCSI), 123 St. Stephen’s Green, Dublin 2, Ireland
| | - Joanna McCarthy
- Pharmacodelivery
Group, School of Pharmacy, University College
Cork, Cavanagh Pharmacy Building, College Road, Cork T12 YN60, Ireland
| | - Éanna B. Forde
- Tissue
Engineering Research Group, Department of Anatomy, Department of Chemistry, and Drug Delivery
& Advanced Materials Team, School of Pharmacy, Royal College of Surgeons in Ireland (RCSI), 123 St. Stephen’s Green, Dublin 2, Ireland
| | - Gary Hessman
- Trinity Centre for Biomedical Engineering and School of Chemistry, Trinity College Dublin (TCD), The University of Dublin, College Green, Dublin 2, Ireland
| | - Caitriona M. O’Driscoll
- Pharmacodelivery
Group, School of Pharmacy, University College
Cork, Cavanagh Pharmacy Building, College Road, Cork T12 YN60, Ireland
| | - Sally-Ann Cryan
- Tissue
Engineering Research Group, Department of Anatomy, Department of Chemistry, and Drug Delivery
& Advanced Materials Team, School of Pharmacy, Royal College of Surgeons in Ireland (RCSI), 123 St. Stephen’s Green, Dublin 2, Ireland
- Trinity Centre for Biomedical Engineering and School of Chemistry, Trinity College Dublin (TCD), The University of Dublin, College Green, Dublin 2, Ireland
- Advanced
Materials and Bioengineering Research (AMBER) Centre, RCSI & TCD, Dublin 2, Ireland
- Centre
for Research in Medical Devices (CURAM), NUIG & RCSI, Biomedical Sciences, National University of Ireland
Galway (NUIG), Newcastle
Road, Galway H91 W2TY, Ireland
| | - Marc Devocelle
- Tissue
Engineering Research Group, Department of Anatomy, Department of Chemistry, and Drug Delivery
& Advanced Materials Team, School of Pharmacy, Royal College of Surgeons in Ireland (RCSI), 123 St. Stephen’s Green, Dublin 2, Ireland
| |
Collapse
|
47
|
Cho N, Shokeen M. Changing landscape of optical imaging in skeletal metastases. J Bone Oncol 2019; 17:100249. [PMID: 31316892 PMCID: PMC6611980 DOI: 10.1016/j.jbo.2019.100249] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 06/25/2019] [Accepted: 06/26/2019] [Indexed: 02/08/2023] Open
Abstract
Optical imaging is an emerging strategy for in vitro and in vivo visualization of the molecular mechanisms of cancer over time. An increasing number of optical imaging contrast agents and techniques have been developed in recent years specifically for bone research and skeletal metastases. Visualizing molecular processes in relation to bone remodeling in metastasized cancers provides valuable information for understanding disease mechanisms and monitoring expression of primary molecular targets and therapeutic efficacy. This review is intended to provide an overview of tumor-specific and non-specific contrast agents in the first near-infrared window (NIR-I) window from 650 nm to 950 nm that can be used to study functional and structural aspects of skeletal remodeling of cancer in preclinical animal models. Near-infrared (NIR) optical imaging techniques, specifically NIR spectroscopy and photoacoustic imaging, and their use in skeletal metastases will also be discussed. Perspectives on the promises and challenges facing this exciting field are then given.
Collapse
Affiliation(s)
- Nicholas Cho
- Department of Radiology, Washington University School of Medicine, 4515 McKinley Ave, St. Louis, MO 63110, United States.,Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63110, United States
| | - Monica Shokeen
- Department of Radiology, Washington University School of Medicine, 4515 McKinley Ave, St. Louis, MO 63110, United States.,Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63110, United States.,Alvin J. Siteman Cancer Center at Washington University School of Medicine and Barnes Jewish Hospital, St. Louis, MO 63110, United States
| |
Collapse
|
48
|
Xiao W, Ma W, Wei S, Li Q, Liu R, Carney RP, Yang K, Lee J, Nyugen A, Yoneda KY, Lam KS, Li T. High-affinity peptide ligand LXY30 for targeting α3β1 integrin in non-small cell lung cancer. J Hematol Oncol 2019; 12:56. [PMID: 31182116 PMCID: PMC6558829 DOI: 10.1186/s13045-019-0740-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 05/14/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND α3β1 integrin is a promising cancer biomarker and drug target. We previously identified a 9-amino-acid cyclic peptide LXY30 for detecting α3β1 integrin on the surface of live tumor cells. This study was undertaken to characterize LXY30 in the detection, cellular function, imaging, and targeted delivery of in vitro and in vivo non-small cell lung cancer (NSCLC) models. METHODS The whole-cell binding assay was performed by incubating NSCLC cells, extracellular vesicles (EVs), and peripheral blood mononuclear cells (PBMCs) with TentaGel resin beads coated with LXY30. In this study, we defined the nanosize EVs as exosomes, which were characterized by flow cytometry, transmission electron microscopy, dynamic light scattering, and Western blots. The function of LXY30 was determined by modulating the epidermal growth factor receptor (EGFR) signaling pathway by growth inhibition and Western blots. For in vivo biodistribution, mice bearing subcutaneous and intracranial NSCLC xenograft tumors were administrated intraveneously with LXY30-biotin/streptavidin-Cy5.5 complex and then analyzed for in vivo and ex vivo optical imaging and histopathology. RESULTS We showed that LXY30 specifically and sensitively detected α3β1 integrin-expressing NSCLC cells and tumor-derived exosomes. Tumor DNA isolated from LXY30-enriched plasma exosomes might be used to detect driver oncogenic mutations in patients with metastatic NSCLC. LXY30 only enriches tumor cells but not neutrophils, macrophages, or monocytes in the malignant pleural effusion of NSCLC patients for detecting genomic alterations by next-generation sequencing. LXY30 detected increased α3β1 integrin expression on the EGFR-mutant NSCLC cells with acquired resistance to erlotinib compared to parental erlotinib-sensitive EGFR-mutant NSCLC cells. We further showed that LXY30 modulated the EGFR signaling pathway independently from another peptide ligand LXW64 targeting αvβ3 integrin in erlotinib-resistant, EGFR-mutant H1975 cells. Analysis of The Cancer Genome Atlas (TCGA) revealed high α3 integrin expression was associated with poor prognosis in lung squamous cell carcinoma. LXY30-biotin/streptavidin-Cy5.5 complex had higher uptakes in the subcutaneous and intracranial xenografts of various α3β1 integrin-expressing lung adenocarcinoma and patient-derived lung squamous cell carcinoma xenografts while sparing the surrounding normal tissues. CONCLUSION LXY30 is a promising peptide for the cancer diagnosis and in vivo targeted delivery of imaging agents and cancer drugs in NSCLC, independent of histology and tumor genotype.
Collapse
Affiliation(s)
- Wenwu Xiao
- Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento, CA, 95817, USA
| | - Weijie Ma
- Division of Hematology/Oncology, Department of Internal Medicine, University of California Davis School of Medicine, University of California Davis Comprehensive Cancer Center, 4501 X Street, Suite 3016, Sacramento, CA, 95817, USA
| | - Sixi Wei
- Division of Hematology/Oncology, Department of Internal Medicine, University of California Davis School of Medicine, University of California Davis Comprehensive Cancer Center, 4501 X Street, Suite 3016, Sacramento, CA, 95817, USA
- Present Address: Department of Biochemistry, Hospital Affiliated to Guizhou Medical University, Guiyang, Guizhou, China
| | - Qianping Li
- Division of Hematology/Oncology, Department of Internal Medicine, University of California Davis School of Medicine, University of California Davis Comprehensive Cancer Center, 4501 X Street, Suite 3016, Sacramento, CA, 95817, USA
- Present Address: Department of Cardiothoracic Surgery, Shanghai Jiaotong University Affiliated Sixth People's Hospital, 600 Yi-Shan Road, Shanghai, 200233, China
| | - Ruiwu Liu
- Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento, CA, 95817, USA
| | - Randy P Carney
- Department of Biomedical Engineering, University of California Davis, Davis, CA, USA
| | - Kevin Yang
- Division of Hematology/Oncology, Department of Internal Medicine, University of California Davis School of Medicine, University of California Davis Comprehensive Cancer Center, 4501 X Street, Suite 3016, Sacramento, CA, 95817, USA
- Present Address: Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Joyce Lee
- Department of Pharmacy, University of California Davis Health System, Sacramento, CA, 95817, USA
| | - Alan Nyugen
- Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento, CA, 95817, USA
| | - Ken Y Yoneda
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, University of California Davis School of Medicine, Sacramento, CA, USA
- Department of Internal Medicine, Veterans Affairs Northern California Health Care System, Mather, CA, USA
| | - Kit S Lam
- Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento, CA, 95817, USA.
- Division of Hematology/Oncology, Department of Internal Medicine, University of California Davis School of Medicine, University of California Davis Comprehensive Cancer Center, 4501 X Street, Suite 3016, Sacramento, CA, 95817, USA.
| | - Tianhong Li
- Division of Hematology/Oncology, Department of Internal Medicine, University of California Davis School of Medicine, University of California Davis Comprehensive Cancer Center, 4501 X Street, Suite 3016, Sacramento, CA, 95817, USA.
- Department of Internal Medicine, Veterans Affairs Northern California Health Care System, Mather, CA, USA.
| |
Collapse
|
49
|
Patil NA, Karas JA, Wade JD, Hossain MA, Tailhades J. Rapid Photolysis‐Mediated Folding of Disulfide‐Rich Peptides. Chemistry 2019; 25:8599-8603. [DOI: 10.1002/chem.201901334] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Indexed: 12/11/2022]
Affiliation(s)
- Nitin A. Patil
- The Monash Biomedicine Discovery Institute 15 Innovation Walk Clayton VIC 3800 Australia
| | - John A. Karas
- Department of Pharmacology and TherapeuticsThe University of Melbourne Victoria 3010 Australia
| | - John D. Wade
- Department of Pharmacology and TherapeuticsThe University of Melbourne Victoria 3010 Australia
- The Florey Institute of Neuroscience and Mental HealthUniversity of Melbourne 30 Royal Parade, Parkville Victoria 3052 Australia
| | - Mohammed Akhter Hossain
- Department of Pharmacology and TherapeuticsThe University of Melbourne Victoria 3010 Australia
- The Florey Institute of Neuroscience and Mental HealthUniversity of Melbourne 30 Royal Parade, Parkville Victoria 3052 Australia
| | - Julien Tailhades
- The Monash Biomedicine Discovery Institute 15 Innovation Walk Clayton VIC 3800 Australia
- EMBL AustraliaMonash University Clayton Victoria 3800 Australia
| |
Collapse
|
50
|
Rapid Discovery of Illuminating Peptides for Instant Detection of Opioids in Blood and Body Fluids. Molecules 2019; 24:molecules24091813. [PMID: 31083395 PMCID: PMC6539258 DOI: 10.3390/molecules24091813] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 05/03/2019] [Accepted: 05/05/2019] [Indexed: 01/03/2023] Open
Abstract
The United States is currently experiencing an opioid crisis, with more than 47,000 deaths in 2017 due to opioid overdoses. Current approaches for opioid identification and quantification in body fluids include immunoassays and chromatographic methods (e.g., LC-MS, GC-MS), which require expensive instrumentation and extensive sample preparation. Our aim was to develop a portable point-of-care device that can be used for the instant detection of opioids in body fluids. Here, we reported the development of a morphine-sensitive fluorescence-based sensor chip to sensitively detect morphine in the blood using a homogeneous immunoassay without any washing steps. Morphine-sensitive illuminating peptides were identified using a high throughput one-bead one-compound (OBOC) combinatorial peptide library approach. The OBOC libraries contain a large number of random peptides with a molecular rotor dye, malachite green (MG), that are coupled to the amino group on the side chain of lysine at different positions of the peptides. The OBOC libraries were then screened for fluorescent activation under a confocal microscope, using an anti-morphine monoclonal antibody as the screening probe, in the presence and absence of free morphine. Using this novel three-step fluorescent screening assay, we were able to identify the peptide-beads that fluoresce in the presence of an anti-morphine antibody, but lost fluorescence when the free morphine was present. After the positive beads were decoded using automatic Edman microsequencing, the morphine-sensitive illuminating peptides were then synthesized in soluble form, functionalized with an azido group, and immobilized onto microfabricated PEG-array spots on a glass slide. The sensor chip was then evaluated for the detection of morphine in plasma. We demonstrated that this proof-of-concept platform can be used to develop fluorescence-based sensors against morphine. More importantly, this technology can also be applied to the discovery of other novel illuminating peptidic sensors for the detection of illicit drugs and cancer biomarkers in body fluids.
Collapse
|