1
|
Hamilton AK, Radaoui AB, Tsang M, Martinez D, Conkrite KL, Patel K, Sidoli S, Delaidelli A, Modi A, Rokita JL, Lane MV, Hartnett N, Lopez RD, Zhang B, Zhong C, Ennis B, Miller DP, Brown MA, Rathi KS, Raman P, Pogoriler J, Bhatti T, Pawel B, Glisovic-Aplenc T, Teicher B, Erickson SW, Earley EJ, Bosse KR, Sorensen PH, Krytska K, Mosse YP, Havenith KE, Zammarchi F, van Berkel PH, Smith MA, Garcia BA, Maris JM, Diskin SJ. A proteogenomic surfaceome study identifies DLK1 as an immunotherapeutic target in neuroblastoma. Cancer Cell 2024:S1535-6108(24)00366-0. [PMID: 39454577 DOI: 10.1016/j.ccell.2024.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 08/14/2024] [Accepted: 10/03/2024] [Indexed: 10/28/2024]
Abstract
Cancer immunotherapies produce remarkable results in B cell malignancies; however, optimal cell surface targets for many solid cancers remain elusive. Here, we present an integrative proteomic, transcriptomic, and epigenomic analysis of tumor and normal tissues to identify biologically relevant cell surface immunotherapeutic targets for neuroblastoma, an often-fatal childhood cancer. Proteogenomic analyses reveal sixty high-confidence candidate immunotherapeutic targets, and we prioritize delta-like canonical notch ligand 1 (DLK1) for further study. High expression of DLK1 directly correlates with a super-enhancer. Immunofluorescence, flow cytometry, and immunohistochemistry show robust cell surface expression of DLK1. Short hairpin RNA mediated silencing of DLK1 in neuroblastoma cells results in increased cellular differentiation. ADCT-701, a DLK1-targeting antibody-drug conjugate (ADC), shows potent and specific cytotoxicity in DLK1-expressing neuroblastoma xenograft models. Since high DLK1 expression is found in several adult and pediatric cancers, our study demonstrates the utility of a proteogenomic approach and credentials DLK1 as an immunotherapeutic target.
Collapse
Affiliation(s)
- Amber K Hamilton
- Center for Childhood Cancer Research and Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Alexander B Radaoui
- Center for Childhood Cancer Research and Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Matthew Tsang
- Center for Childhood Cancer Research and Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Daniel Martinez
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Karina L Conkrite
- Center for Childhood Cancer Research and Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Khushbu Patel
- Center for Childhood Cancer Research and Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Simone Sidoli
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Alberto Delaidelli
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada; Department of Molecular Oncology, British Columbia Cancer Research Centre, Vancouver, BC, Canada
| | - Apexa Modi
- Center for Childhood Cancer Research and Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Jo Lynne Rokita
- Center for Data-Driven Discovery in Biomedicine and Division of Neurosurgery, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Maria V Lane
- Center for Childhood Cancer Research and Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Nicholas Hartnett
- Center for Childhood Cancer Research and Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Raphael D Lopez
- Center for Childhood Cancer Research and Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Bo Zhang
- Center for Data-Driven Discovery in Biomedicine and Division of Neurosurgery, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Chuwei Zhong
- Center for Data-Driven Discovery in Biomedicine and Division of Neurosurgery, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Brian Ennis
- Center for Data-Driven Discovery in Biomedicine and Division of Neurosurgery, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Daniel P Miller
- Center for Data-Driven Discovery in Biomedicine and Division of Neurosurgery, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Miguel A Brown
- Center for Data-Driven Discovery in Biomedicine and Division of Neurosurgery, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Komal S Rathi
- Center for Childhood Cancer Research and Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Center for Data-Driven Discovery in Biomedicine and Division of Neurosurgery, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Department of Biomedical and Health Informatics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Pichai Raman
- Center for Childhood Cancer Research and Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Center for Data-Driven Discovery in Biomedicine and Division of Neurosurgery, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Department of Biomedical and Health Informatics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Jennifer Pogoriler
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Tricia Bhatti
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Bruce Pawel
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Tina Glisovic-Aplenc
- Center for Childhood Cancer Research and Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | | | | | - Eric J Earley
- RTI International, Research Triangle Park, Durham, NC 27709, USA
| | - Kristopher R Bosse
- Center for Childhood Cancer Research and Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Poul H Sorensen
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada; Department of Molecular Oncology, British Columbia Cancer Research Centre, Vancouver, BC, Canada
| | - Kateryna Krytska
- Center for Childhood Cancer Research and Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Yael P Mosse
- Center for Childhood Cancer Research and Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | | | | | | | - Benjamin A Garcia
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - John M Maris
- Center for Childhood Cancer Research and Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Sharon J Diskin
- Center for Childhood Cancer Research and Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
2
|
Weiner AK, Radaoui AB, Tsang M, Martinez D, Sidoli S, Conkrite KL, Delaidelli A, Modi A, Rokita JL, Patel K, Lane MV, Zhang B, Zhong C, Ennis B, Miller DP, Brown MA, Rathi KS, Raman P, Pogoriler J, Bhatti T, Pawel B, Glisovic-Aplenc T, Teicher B, Erickson SW, Earley EJ, Bosse KR, Sorensen PH, Krytska K, Mosse YP, Havenith KE, Zammarchi F, van Berkel PH, Smith MA, Garcia BA, Maris JM, Diskin SJ. A proteogenomic surfaceome study identifies DLK1 as an immunotherapeutic target in neuroblastoma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.12.06.570390. [PMID: 38106022 PMCID: PMC10723418 DOI: 10.1101/2023.12.06.570390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
Cancer immunotherapies have produced remarkable results in B-cell malignancies; however, optimal cell surface targets for many solid cancers remain elusive. Here, we present an integrative proteomic, transcriptomic, and epigenomic analysis of tumor specimens along with normal tissues to identify biologically relevant cell surface proteins that can serve as immunotherapeutic targets for neuroblastoma, an often-fatal childhood cancer of the developing nervous system. We apply this approach to human-derived cell lines (N=9) and cell/patient-derived xenograft (N=12) models of neuroblastoma. Plasma membrane-enriched mass spectrometry identified 1,461 cell surface proteins in cell lines and 1,401 in xenograft models, respectively. Additional proteogenomic analyses revealed 60 high-confidence candidate immunotherapeutic targets and we prioritized Delta-like canonical notch ligand 1 (DLK1) for further study. High expression of DLK1 directly correlated with the presence of a super-enhancer spanning the DLK1 locus. Robust cell surface expression of DLK1 was validated by immunofluorescence, flow cytometry, and immunohistochemistry. Short hairpin RNA mediated silencing of DLK1 in neuroblastoma cells resulted in increased cellular differentiation. ADCT-701, a DLK1-targeting antibody-drug conjugate (ADC), showed potent and specific cytotoxicity in DLK1-expressing neuroblastoma xenograft models. Moreover, DLK1 is highly expressed in several adult cancer types, including adrenocortical carcinoma (ACC), pheochromocytoma/paraganglioma (PCPG), hepatoblastoma, and small cell lung cancer (SCLC), suggesting potential clinical benefit beyond neuroblastoma. Taken together, our study demonstrates the utility of comprehensive cancer surfaceome characterization and credentials DLK1 as an immunotherapeutic target. Highlights Plasma membrane enriched proteomics defines surfaceome of neuroblastomaMulti-omic data integration prioritizes DLK1 as a candidate immunotherapeutic target in neuroblastoma and other cancersDLK1 expression is driven by a super-enhancer DLK1 silencing in neuroblastoma cells results in cellular differentiation ADCT-701, a DLK1-targeting antibody-drug conjugate, shows potent and specific cytotoxicity in DLK1-expressing neuroblastoma preclinical models.
Collapse
|
3
|
Sasaki I, Tada M, Liu Z, Tatsuta M, Okura T, Aoki M, Takahashi K, Ishioka NS, Watanabe S, Tanaka H. 1-( N, N-Dialkylcarbamoyl)-1,1-difluoromethanesulfonyl ester as a stable and effective precursor for a neopentyl labeling group with astatine-211. Org Biomol Chem 2023; 21:7467-7472. [PMID: 37670575 DOI: 10.1039/d3ob00944k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/07/2023]
Abstract
Radiohalogens with a short half-life are useful radioisotopes for radiotheranostics. Astatine-211 is an α-emitting radiohalogen and is expected to be applicable to targeted α therapy. A neopentyl labeling group is an effective hydrophilic labeling unit for various radiohalogens, which includes 211At. In this study, a 1-(N,N-dialkylcarbamoyl)-1,1-difluoromethanesulfonyl (CDf) ester was developed as a stable precursor for labeling with 211At, 77Br and 125I through a neopentyl labeling group. The CDf ester remained stable in an acetonitrile solution at room temperature and enabled the successful syntheses of 211At-labeled compounds in a highly radiochemical conversion in the presence of K2CO3. 77Br- and 125I-labeled compounds can be prepared from the CDf ester without a base. The utility of the CDf ester was demonstrated in the synthesis of a benzylguanidine with a neopentyl 211At-labeling group. The developed method afforded a 32% radiochemical yield of 211At-labeled benzylguanidine. However, a partial deastatination was observed under acidic conditions during the removal of an N-Boc protecting group. Deprotecting these groups under milder acidic conditions may improve the radiochemical yield. In conclusion, the CDf ester facilitates the syntheses of 211At, 125I and 77Br-labeled compounds that use a neopentyl labeling group for radiotheranostic applications. Further optimization of protecting groups and reaction conditions should enhance the total radiochemical yield of the 211At-labeled compounds.
Collapse
Affiliation(s)
- Ichiro Sasaki
- Department of Quantum-Applied Biosciences, Takasaki Institute for Advanced Quantum Science National Institutes for Quantum Science and Technology, 1233 Watanuki-machi, Takasaki, Gunma 370-1292, Japan
| | - Masatoshi Tada
- Department of Chemical Science and Engineering, Tokyo Institute of Technology, 12-12-1-H101 Ookayama, Meguro, Tokyo, 152-8552, Japan.
| | - Ziyun Liu
- Department of Chemical Science and Engineering, Tokyo Institute of Technology, 12-12-1-H101 Ookayama, Meguro, Tokyo, 152-8552, Japan.
| | - Maho Tatsuta
- Department of Chemical Science and Engineering, Tokyo Institute of Technology, 12-12-1-H101 Ookayama, Meguro, Tokyo, 152-8552, Japan.
| | - Takeru Okura
- Department of Chemical Science and Engineering, Tokyo Institute of Technology, 12-12-1-H101 Ookayama, Meguro, Tokyo, 152-8552, Japan.
| | - Miho Aoki
- Advanced Clinical Research Center, Fukushima Global Medical Science Center, Fukushima Medical University, 1 Hikariga-oka, Fukushima, Fukushima 960-1295, Japan
| | - Kazuhiro Takahashi
- Advanced Clinical Research Center, Fukushima Global Medical Science Center, Fukushima Medical University, 1 Hikariga-oka, Fukushima, Fukushima 960-1295, Japan
| | - Noriko S Ishioka
- Department of Quantum-Applied Biosciences, Takasaki Institute for Advanced Quantum Science National Institutes for Quantum Science and Technology, 1233 Watanuki-machi, Takasaki, Gunma 370-1292, Japan
| | - Shigeki Watanabe
- Department of Quantum-Applied Biosciences, Takasaki Institute for Advanced Quantum Science National Institutes for Quantum Science and Technology, 1233 Watanuki-machi, Takasaki, Gunma 370-1292, Japan
| | - Hiroshi Tanaka
- Department of Chemical Science and Engineering, Tokyo Institute of Technology, 12-12-1-H101 Ookayama, Meguro, Tokyo, 152-8552, Japan.
| |
Collapse
|
4
|
Liu F, Wu Q, Dong Z, Liu K. Integrins in cancer: Emerging mechanisms and therapeutic opportunities. Pharmacol Ther 2023:108458. [PMID: 37245545 DOI: 10.1016/j.pharmthera.2023.108458] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 05/10/2023] [Accepted: 05/22/2023] [Indexed: 05/30/2023]
Abstract
Integrins are vital surface adhesion receptors that mediate the interactions between the extracellular matrix (ECM) and cells and are essential for cell migration and the maintenance of tissue homeostasis. Aberrant integrin activation promotes initial tumor formation, growth, and metastasis. Recently, many lines of evidence have indicated that integrins are highly expressed in numerous cancer types and have documented many functions of integrins in tumorigenesis. Thus, integrins have emerged as attractive targets for the development of cancer therapeutics. In this review, we discuss the underlying molecular mechanisms by which integrins contribute to most of the hallmarks of cancer. We focus on recent progress on integrin regulators, binding proteins, and downstream effectors. We highlight the role of integrins in the regulation of tumor metastasis, immune evasion, metabolic reprogramming, and other hallmarks of cancer. In addition, integrin-targeted immunotherapy and other integrin inhibitors that have been used in preclinical and clinical studies are summarized.
Collapse
Affiliation(s)
- Fangfang Liu
- Research Center of Basic Medicine, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China; China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan 450008, China
| | - Qiong Wu
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan 450008, China; Department of Pathophysiology, School of Basic Medical Sciences, College of Medicine, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Zigang Dong
- Research Center of Basic Medicine, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China; China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan 450008, China; Department of Pathophysiology, School of Basic Medical Sciences, College of Medicine, Zhengzhou University, Zhengzhou, Henan 450001, China; State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou, Henan 450000, China; Tianjian Advanced Biomedical Laboratory, Zhengzhou University, Zhengzhou, Henan 450001, China.
| | - Kangdong Liu
- Research Center of Basic Medicine, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China; China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan 450008, China; Department of Pathophysiology, School of Basic Medical Sciences, College of Medicine, Zhengzhou University, Zhengzhou, Henan 450001, China; State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou, Henan 450000, China; Tianjian Advanced Biomedical Laboratory, Zhengzhou University, Zhengzhou, Henan 450001, China; Cancer Chemoprevention International Collaboration Laboratory, Zhengzhou, Henan 450000, China.
| |
Collapse
|
5
|
Zhao S, Liu J, Lv Z, Zhang G, Xu Z. Recent updates on 1,2,3-triazole-containing hybrids with in vivo therapeutic potential against cancers: A mini-review. Eur J Med Chem 2023; 251:115254. [PMID: 36893627 DOI: 10.1016/j.ejmech.2023.115254] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 03/01/2023] [Accepted: 03/04/2023] [Indexed: 03/08/2023]
Abstract
1,2,3-Triazole moiety which is usually constructed by highly versatile, efficacious and selective copper-catalyzed azide-alkyne cycloaddition not only can act as a linker to connect different pharmacophores, but also is a useful pharmacophore with diverse biological properties. 1,2,3-Triazoles are readily interact with diverse enzymes and receptors in cancer cells through non-covalent interactions and can inhibit cancer cell proliferation, arrest cell cycle and induce apoptosis. In particular, 1,2,3-triazole-containing hybrids have the potential to exert dual or multiple anticancer mechanisms of action, representing useful scaffolds in expediting development of novel anticancer agents. The current review summarizes the in vivo anticancer efficacy and mechanisms of action of 1,2,3-triazole-containing hybrids reported in the last decade to continuously open up a map for the remarkable exploration of more effective candidates.
Collapse
Affiliation(s)
- Shijia Zhao
- School of Chemistry and Chemical Engineering, Wuhan University of Science and Technology, Wuhan, PR China
| | - Jie Liu
- School of Chemistry and Chemical Engineering, Wuhan University of Science and Technology, Wuhan, PR China
| | - Zaosheng Lv
- School of Chemistry and Chemical Engineering, Wuhan University of Science and Technology, Wuhan, PR China
| | - Guangde Zhang
- Engineering Training Center, Wuhan University of Science and Technology, Wuhan, PR China.
| | - Zhi Xu
- School of Chemistry and Chemical Engineering, Wuhan University of Science and Technology, Wuhan, PR China.
| |
Collapse
|
6
|
Fujioka K, Hay BA, Godugu K, Mousa SA. Pharmacokinetics of fluorobenzyl polyethylene glycol conjugated tetraiodothyroacetic acid (NP751), a novel anticancer thyrointegrin α vβ 3 antagonist. Front Pharmacol 2022; 13:902141. [PMID: 36518666 PMCID: PMC9742531 DOI: 10.3389/fphar.2022.902141] [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: 03/22/2022] [Accepted: 11/02/2022] [Indexed: 08/30/2023] Open
Abstract
We have recently reported on the development of fb-PMT (NP751), a conjugate of the thyroid hormone metabolite tetraiodothyroacetic acid (tetrac) and monodisperse polyethylene glycol 36. It exhibited high affinity for thyrointegrin αvβ3 receptor and potent anti-angiogenic and anticancer activity in vivo. The objective of the current study is to determine the pharmacokinetics (PK) of fb-PMT in experimental animals, such as mice, rats, and monkeys. NP751 was quantified using a propylene diamine-modified tetraiodothyroacetic acid (DAT) as an internal standard. The limit of quantification (LOQ) for fb-PMT was 1.5 ng/μL and the recovery efficiency was 93.9% with the developed method. The peak plasma concentration (Cmax) and the area under the curve (AUC) results at different doses in mice, rats and monkeys suggest that pharmacokinetics of NP751 is dose-dependent within the dose ranges administered. Results indicate that NP751 has comparable PK parameters that provides enough exposure as a molecularly tumor targeted molecule in multiple species and is a promising anticancer therapeutic.
Collapse
Affiliation(s)
| | | | | | - Shaker A. Mousa
- Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences and Nanopharmaceuticals, LLC, Rensselaer, NY, United States
| |
Collapse
|
7
|
Karakus OO, Godugu K, Salaheldin T, Fujioka K, Mousa SA. Norepinephrine transporter analog benzylguanidine-conjugated nanoparticles for the delivery of paclitaxel in neuroblastoma. Nanomedicine (Lond) 2021; 16:2331-2342. [PMID: 34651508 DOI: 10.2217/nnm-2021-0230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: We previously synthesized a polyethylene glycol-based norepinephrine transporter-targeted agent, BG-P-TAT, which has a benzylguanidine and a triazolyl-tetrac group. This targeted conjugate showed suppression of neuroblastoma tumor progression. In this study we aimed to synthesize nanoparticles to encapsulate the chemotherapeutic agent paclitaxel for targeting neuroblastoma tumors by using benzylguanidine so that it can compete with norepinephrine for uptake by neuroendocrine cells. Methods: Biocompatible poly(lactide-co-glycolic acid)-polyethylene glycol was chosen to prepare targeted nanoparticles for safe delivery of the chemotherapy agent paclitaxel. Result: Paclitaxel concentration was 60% higher in neuroblastoma tumors of mice treated with paclitaxel encapsulated in targeted nanoparticles than with non-targeted nanoparticles. Conclusion: These findings support the targeted delivery of paclitaxel as a chemotherapeutic agent for neuroblastoma.
Collapse
Affiliation(s)
- Ozlem Ozen Karakus
- Pharmaceutical Research Institute, Albany College of Pharmacy & Health Sciences, Rensselaer, NY 12144, USA
| | - Kavitha Godugu
- Pharmaceutical Research Institute, Albany College of Pharmacy & Health Sciences, Rensselaer, NY 12144, USA
| | - Taher Salaheldin
- Pharmaceutical Research Institute, Albany College of Pharmacy & Health Sciences, Rensselaer, NY 12144, USA
| | - Kazutoshi Fujioka
- Pharmaceutical Research Institute, Albany College of Pharmacy & Health Sciences, Rensselaer, NY 12144, USA
| | - Shaker A Mousa
- Pharmaceutical Research Institute, Albany College of Pharmacy & Health Sciences, Rensselaer, NY 12144, USA
| |
Collapse
|
8
|
Sudha T, Godugu K, Darwish NHE, Nazeer T, Mousa SA. Novel Polyethylene Glycol-Conjugated Triazole Derivative with High Thyrointegrin αvβ3 Affinity in Acute Myeloid Leukemia Management. Cancers (Basel) 2021; 13:cancers13164070. [PMID: 34439224 PMCID: PMC8392871 DOI: 10.3390/cancers13164070] [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: 07/26/2021] [Revised: 08/05/2021] [Accepted: 08/10/2021] [Indexed: 02/08/2023] Open
Abstract
(1) Background: Acute myeloid leukemia (AML) accounts for up to one-third of more than 60,000 leukemia cases diagnosed annually in the U.S. Primary AML cells express membrane αvβ3 integrin, which is associated with adverse prognosis and resistance to chemotherapies. A novel anticancer compound Polyethylene glycol-conjugated bi-TriAzole Tetraiodothyroacetic acid (P-bi-TAT) interacts with high affinity (Ki 0.3 nM) and specificity with the thyrointegrin αvβ3. We evaluated P-bi-TAT activities in two different AML models representing monocytic and myelocytic forms of acute leukemia. (2) Methods and Results: The in vivo AML models were established prior to initiation of treatment protocols by grafting human leukemia cells in immunocompromised mice. IVIS imaging scans revealed that leukemic colonies were extensively established throughout the bone marrow, liver, and lung of the untreated animals. In animals treated with P-bi-TAT at daily doses ranging from 1-10 mg/kg, subcutaneously for 2-3 weeks, IVIS imaging scans revealed 95% reduction in bone marrow colonies and leukemic colonies in liver and lung. Also, the leukemic cells were not detected in bone marrow samples of P-bi-TAT-treated animals. The anti-neoplastic effect of P-bi-TAT administration on leukemic cells was associated with marked inhibition of NF-κB activity. We conclude that experimental P-bi-TAT therapy in vivo appears extraordinarily effective against the two forms of human AML models in mice. Because the P-bi-TAT molecular target, thyrointegrin αvβ3, is consistently expressed in many, if not all, clinical AML samples, P-bi-TAT-based therapy seems to have significant clinical potential in treating most AML sub-types. Hence, P-bi-TAT represents a promising targeted therapeutic agent for AML patients.
Collapse
Affiliation(s)
- Thangirala Sudha
- Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Rensselaer, NY 12144, USA; (T.S.); (K.G.); (N.H.E.D.)
| | - Kavitha Godugu
- Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Rensselaer, NY 12144, USA; (T.S.); (K.G.); (N.H.E.D.)
| | - Noureldien H. E. Darwish
- Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Rensselaer, NY 12144, USA; (T.S.); (K.G.); (N.H.E.D.)
- Hematology Unit, Clinical Pathology Department, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt
| | - Tipu Nazeer
- Albany Medical Center, Pathology Department, AMC Hospital, Albany, NY 12208, USA;
| | - Shaker A. Mousa
- Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Rensselaer, NY 12144, USA; (T.S.); (K.G.); (N.H.E.D.)
- Correspondence:
| |
Collapse
|
9
|
Li X, Li X, Liu F, Li S, Shi D. Rational Multitargeted Drug Design Strategy from the Perspective of a Medicinal Chemist. J Med Chem 2021; 64:10581-10605. [PMID: 34313432 DOI: 10.1021/acs.jmedchem.1c00683] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The development of multitarget-directed ligands (MTDLs) has become a widely focused research topic, but rational design remains as an enormous challenge. This paper reviews and discusses the design strategy of incorporating the second activity into an existing single-active ligand. If the binding sites of both targets share similar endogenous substrates, MTDLs can be designed by merging two lead compounds with similar functional groups. If the binding sites are large or adjacent to the solution, two key pharmacophores can be fused directly. If the binding regions are small and deep inside the proteins, the linked-pharmacophore strategy might be the only way. The added pharmacophores of second targets should not affect the binding mode of the original ones. Moreover, the inhibitory activities of the two targets need to be adjusted to achieve an optimal ratio.
Collapse
Affiliation(s)
- Xiangqian Li
- State Key Laboratory of Microbial Technology, Shandong University, 72 Binhai Road, Qingdao 266237, Shandong, P. R. China
| | - Xiaowei Li
- State Key Laboratory of Microbial Technology, Shandong University, 72 Binhai Road, Qingdao 266237, Shandong, P. R. China
| | - Fang Liu
- State Key Laboratory of Microbial Technology, Shandong University, 72 Binhai Road, Qingdao 266237, Shandong, P. R. China
| | - Shuo Li
- State Key Laboratory of Microbial Technology, Shandong University, 72 Binhai Road, Qingdao 266237, Shandong, P. R. China
| | - Dayong Shi
- State Key Laboratory of Microbial Technology, Shandong University, 72 Binhai Road, Qingdao 266237, Shandong, P. R. China
| |
Collapse
|
10
|
Ozen Karakus O, Godugu K, Mousa SA. Discovery of dual targeting PEGylated BG-P 1600-TAT to norepinephrine transporter (NET) and thyrointegrin αvβ3 in the treatment of neuroblastoma. Bioorg Med Chem 2021; 43:116278. [PMID: 34157571 DOI: 10.1016/j.bmc.2021.116278] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 06/07/2021] [Accepted: 06/11/2021] [Indexed: 12/18/2022]
Abstract
Polymer-drug conjugates are growing in interest as novel anticancer agents for targeted cancer therapy. The aim of this study was to synthesize a poly(ethylene glycol) (PEG) conjugated anticancer drug for neuroblastoma, which is the most common extracranial solid tumor of childhood and the deadliest tumor of infancy. In our previous studies, we designed and synthesized a dual targeting agent using benzylguanidine (BG) conjugated with the high affinity thyrointegrin αvβ3 antagonist TriAzole Tetraiodothyroacetic acid (TAT) via non-cleavable bonding to PEG400 to make BG-P400-TAT and its derivatives as agents against neuroblastoma. Here, we improved the pharmacodynamic properties and increased the solubility by changing the polymer length to 1600 molecular weight. The TAT group, which acts as an integrin αvβ3 antagonist, and the BG group, which can be taken up by neuroblastoma cells through the norepinephrine transporter (NET) system, are conjugated to PEG1600 to make BG-PEG1600-TAT. The binding affinity of BG-PEG1600-TAT was 40-fold higher to integrin αvβ3 versus BG-P400-TAT and was associated with greater anticancer activities against neuroblastoma cells (SK-N-F1 and SKNAS) implanted in SCID mice along with broad spectrum anti-angiogenesis activities versus the FDA approved anti-Vascular Endothelial Growth Factor (VEGF) monoclonal antibody Avastin (bevacizumab). In conclusion, our novel dual targeting of NET and αvβ3 receptor antagonist, BG-P1600-TAT demonstrated broad spectrum anti-angiogenesis and anti-cancer activities in suppressing neuroblastoma tumor progression and metastasis. Thus, BG-PEG1600-TAT represents a potential clinical candidate for targeted therapy in neuroblastoma management.
Collapse
Affiliation(s)
- Ozlem Ozen Karakus
- Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Rensselaer, NY 12144, United States
| | - Kavitha Godugu
- Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Rensselaer, NY 12144, United States
| | - Shaker A Mousa
- Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Rensselaer, NY 12144, United States.
| |
Collapse
|
11
|
Karakus OO, Godugu K, Fujioka K, Mousa SA. Design, synthesis, and biological evaluation of novel bifunctional thyrointegrin antagonists for neuroblastoma. Bioorg Med Chem 2021; 42:116250. [PMID: 34118788 DOI: 10.1016/j.bmc.2021.116250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 05/10/2021] [Accepted: 05/28/2021] [Indexed: 11/18/2022]
Abstract
Receptor-mediated cancer therapy has received much attention in the last few decades. Neuroblastoma and other cancers of the sympathetic nervous system highly express norepinephrine transporter (NET) and cell plasma membrane integrin αvβ3. Dual targeting of the NET and integrin αvβ3 receptors using a Drug-Drug Conjugate (DDC) might provide effective treatment strategy in the fight against neuroblastoma and other neuroendocrine tumors. In this work, we synthesized three dual-targeting BG-P400-TAT derivatives, dI-BG-P400-TAT, dM-BG-P400-TAT, and BG-P400-PAT containing di-iodobenzene, di-methoxybenzene, and piperazine groups, respectively. These derivatives utilize to norepinephrine transporter (NET) and the integrin αvβ3 receptor to simultaneously modulate both targets based on evaluation in a neuroblastoma animal model using the neuroblastoma SK-N-F1 cell line. Among the three synthesized agents, the piperazine substituted BG-P400-PAT exhibited potent integrin αvβ3 antagonism and reduced neuroblastoma tumor growth and cancer cell viability by >90%. In conclusion, BG-P400-PAT and derivatives represent a potential therapeutic approach in the management of neuroblastoma.
Collapse
Affiliation(s)
- Ozlem Ozen Karakus
- Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Rensselaer, NY 12144, United States
| | - Kavitha Godugu
- Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Rensselaer, NY 12144, United States
| | - Kazutoshi Fujioka
- Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Rensselaer, NY 12144, United States
| | - Shaker A Mousa
- Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Rensselaer, NY 12144, United States.
| |
Collapse
|
12
|
Cirillo M, Giacomini D. Molecular Delivery of Cytotoxic Agents via Integrin Activation. Cancers (Basel) 2021; 13:299. [PMID: 33467465 PMCID: PMC7830197 DOI: 10.3390/cancers13020299] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 01/11/2021] [Accepted: 01/12/2021] [Indexed: 12/16/2022] Open
Abstract
Integrins are cell adhesion receptors overexpressed in tumor cells. A direct inhibition of integrins was investigated, but the best inhibitors performed poorly in clinical trials. A gained attention towards these receptors arouse because they could be target for a selective transport of cytotoxic agents. Several active-targeting systems have been developed to use integrins as a selective cell entrance for some antitumor agents. The aim of this review paper is to report on the most recent results on covalent conjugates between integrin ligands and antitumor drugs. Cytotoxic drugs thus conjugated through specific linker to integrin ligands, mainly RGD peptides, demonstrated that the covalent conjugates were more selective against tumor cells and hopefully with fewer side effects than the free drugs.
Collapse
Affiliation(s)
| | - Daria Giacomini
- Department of Chemistry “Giacomo Ciamician”, Alma Mater Studiorum University of Bologna, Via Selmi 2, 40126 Bologna, Italy;
| |
Collapse
|