1
|
He Z, Zhao X, Gao WY, Bao G, Li Y, Zuo Q, Song X, Mou LY, Sun W, Wang R. Controlled reversible methionine-selective sulfimidation of peptides. SCIENCE ADVANCES 2025; 11:eadv8712. [PMID: 40397727 PMCID: PMC12094199 DOI: 10.1126/sciadv.adv8712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Accepted: 04/16/2025] [Indexed: 05/23/2025]
Abstract
Site-selective chemical peptide manipulation is an effective strategy to understand and regulate structure and function. However, methionine-selective modification remains one of the most difficult challenges in peptide chemistry, with notable limited strategies. In this study, we report a general reversible modification strategy at methionine sites that uses the ruthenium-catalyzed sulfimidation of peptides. This method provides a convenient and effective strategy for late-stage peptide functionalization. The N═S bonds of the conjugates are reduced in the presence of glutathione, resulting the traceless releasing of corresponding peptides and amides. Practical applications are then demonstrated using precise reversible modifications of bioactive peptides, the stapling and linearization of peptides, peptide-drug conjugates, and split-and-pool synthesis. This on/off strategy through methionine-selective and reversible sulfimidation provides a unique tool for peptide chemistry and peptide-based drug discovery.
Collapse
Affiliation(s)
- Zeyuan He
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences & Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou 730000, China
| | - Xiufang Zhao
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences & Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou 730000, China
| | - Wen-Yan Gao
- School of Life Sciences, Lanzhou University, Lanzhou 730000, China
| | - Guangjun Bao
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences & Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou 730000, China
| | - Yiping Li
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences & Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou 730000, China
| | - Quan Zuo
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Xinyi Song
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences & Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou 730000, China
| | - Ling-Yun Mou
- School of Life Sciences, Lanzhou University, Lanzhou 730000, China
| | - Wangsheng Sun
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences & Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou 730000, China
| | - Rui Wang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences & Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou 730000, China
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| |
Collapse
|
2
|
Pati B, Kumar A, Chowdhury A, Tripathi NM, Gour V, Mukherjee A, Bandyopadhyay A. An Upgraded Solid-Phase Assembly of Chelators (DOTA and NOTA) Enabled Bacterial Uptake Studies of Radiolabeled Peptide. Chembiochem 2025; 26:e202400996. [PMID: 39658876 DOI: 10.1002/cbic.202400996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Accepted: 12/10/2024] [Indexed: 12/12/2024]
Abstract
Among popular radio metal chelators, DOTA and NOTA have been remarkably considered in radionuclide therapy and imaging studies due to several advantages in pharmacology. Here, we developed a practical and general method for assembling DOTA and NOTA in the solid phase peptide (pseudo-dilute conditions) using a wide range of solvents with easily accessible and economical feedstocks, which mitigated unprecedented challenges associated with previously reported methods. This upgraded approach enabled an efficient installation of these two chelators on various bioactive peptide sequences. Finally, we assessed the antimicrobial activity of the DOTA- and NOTA-attached Combi peptides to B. subtilis, which was intact. The authenticity of the assembled DOTA framework was assessed by labeling 177Lu and in vitro bacterial uptake in E. coli and S. aureus. 177Lu-labeled DOTA-Combi peptide exhibited promising uptake for developing a bacterial infection imaging agent while negligible hemolysis activity even at >200 μM. This contribution will be valued for developing peptide radiopharmaceuticals with operational simplicity and economic approaches.
Collapse
Affiliation(s)
- Bibekananda Pati
- Biomimetic Peptide Engineering Laboratory, Department of Chemistry, Indian Institute of Technology Ropar, Rupnagar, Punjab, 140001, India
| | - Anuj Kumar
- Radiopharmaceuticals Division, Bhabha Atomic Research Centre (BARC), Mumbai, 400085, India
| | - Arnab Chowdhury
- Biomimetic Peptide Engineering Laboratory, Department of Chemistry, Indian Institute of Technology Ropar, Rupnagar, Punjab, 140001, India
| | - Nitesh Mani Tripathi
- Biomimetic Peptide Engineering Laboratory, Department of Chemistry, Indian Institute of Technology Ropar, Rupnagar, Punjab, 140001, India
| | - Vinod Gour
- Biomimetic Peptide Engineering Laboratory, Department of Chemistry, Indian Institute of Technology Ropar, Rupnagar, Punjab, 140001, India
| | - Archana Mukherjee
- Radiopharmaceuticals Division, Bhabha Atomic Research Centre (BARC), Mumbai, 400085, India
| | - Anupam Bandyopadhyay
- Biomimetic Peptide Engineering Laboratory, Department of Chemistry, Indian Institute of Technology Ropar, Rupnagar, Punjab, 140001, India
| |
Collapse
|
3
|
Flores-Prieto DE, Stabenfeldt SE. Nanoparticle targeting strategies for traumatic brain injury. J Neural Eng 2024; 21:061007. [PMID: 39622184 DOI: 10.1088/1741-2552/ad995b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 12/02/2024] [Indexed: 12/21/2024]
Abstract
Nanoparticle (NP)-based drug delivery systems hold immense potential for targeted therapy and diagnosis of neurological disorders, overcoming the limitations of conventional treatment modalities. This review explores the design considerations and functionalization strategies of NPs for precise targeting of the brain and central nervous system. This review discusses the challenges associated with drug delivery to the brain, including the blood-brain barrier and the complex heterogeneity of traumatic brain injury. We also examine the physicochemical properties of NPs, emphasizing the role of size, shape, and surface characteristics in their interactions with biological barriers and cellular uptake mechanisms. The review concludes by exploring the options of targeting ligands designed to augment NP affinity and retention to specific brain regions or cell types. Various targeting ligands are discussed for their ability to mimic receptor-ligand interaction, and brain-specific extracellular matrix components. Strategies to mimic viral mechanisms to increase uptake are discussed. Finally, the emergence of antibody, antibody fragments, and antibody mimicking peptides are discussed as promising targeting strategies. By integrating insights from these scientific fields, this review provides an understanding of NP-based targeting strategies for personalized medicine approaches to neurological disorders. The design considerations discussed here pave the way for the development of NP platforms with enhanced therapeutic efficacy and minimized off-target effects, ultimately advancing the field of neural engineering.
Collapse
Affiliation(s)
- David E Flores-Prieto
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, United States of America
| | - Sarah E Stabenfeldt
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, United States of America
| |
Collapse
|
4
|
Sahel DK, Goswami SG, Jatyan R, Tharmatt A, Singh V, Dalela M, Mohanty S, Mittal A, Ramalingam S, Chitkara D. cRGD-modified hybrid lipopolymeric nanoplexes for gene editing in the posterior segment of the eye. Int J Biol Macromol 2024; 271:132426. [PMID: 38820904 DOI: 10.1016/j.ijbiomac.2024.132426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Revised: 04/21/2024] [Accepted: 05/14/2024] [Indexed: 06/02/2024]
Abstract
Eye-related diseases, specifically retinal dystrophy (RD) conditions, are the leading cause of blindness worldwide. Gene addition, regulation, or editing could potentially treat such diseases through gene expression regulation. CRISPR/Cas9 gene editing is one of the most prominent and precise gene editing tools which could be employed to edit genes related to the dystrophic condition. However, CRISPR/Cas9 faces in vivo delivery challenges due to its high molecular weight, negative charge, prone to degradation in the presence of nucleases and proteases, poor cellular degradation, etc., which makes it challenging to adopt for therapeutic applications. We developed cRGD-modified lipopolymeric nanoplexes loaded with Cas9 RNPs with a particle size and zeta potential of 175 ± 20 nm and 2.15 ± 0.9 mV, respectively. The cRGD-modified lipopolymeric nanoplexes were stable for 194 h and able to transfect >70 % ARPE-19 and NIH3T3 cells with an Indel frequency of ~40 % for the VEGF-A gene. The cRGD-modified lipopolymeric nanoplexes found good vitreous mobility and could transfection retinal cells in vivo after 48 h of intravitreal injection in Wistar Rats. Moreover, in vivo VEGFA gene editing was ~10 % with minimal toxicities. Collectively, the cRGD-modified lipopolymeric nanoplexes were found to have extreme potential in delivering CRISPR/Cas9 RNPs payload to the retinal tissues for therapeutic applications.
Collapse
Affiliation(s)
- Deepak Kumar Sahel
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani (BITS Pilani), Pilani Campus, Vidya Vihar, Pilani, Rajasthan, India
| | | | - Reena Jatyan
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani (BITS Pilani), Pilani Campus, Vidya Vihar, Pilani, Rajasthan, India
| | - Abhay Tharmatt
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani (BITS Pilani), Pilani Campus, Vidya Vihar, Pilani, Rajasthan, India
| | - Vivek Singh
- Prof. Brien Holden Eye Research Center, Champalimaud Translational Centre for Eye Research, L.V. Prasad Eye Institute, Kallam Anji Reddy Campus, L V Prasad Marg, Hyderabad, India
| | - Manu Dalela
- Stem Cell Facility, DBT-Centre of Excellence for Stem Cell Research, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Sujata Mohanty
- Stem Cell Facility, DBT-Centre of Excellence for Stem Cell Research, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Anupama Mittal
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani (BITS Pilani), Pilani Campus, Vidya Vihar, Pilani, Rajasthan, India
| | | | - Deepak Chitkara
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani (BITS Pilani), Pilani Campus, Vidya Vihar, Pilani, Rajasthan, India.
| |
Collapse
|
5
|
Wan H, Wang S, Li C, Zeng B, Wu H, Liu C, Chen L, Jin M, Huang W, Zang Y, Zhang D, Gao Z, Jin Z. LA67 Liposome-Loaded Thermo-Sensitive Hydrogel with Active Targeting for Efficient Treatment of Keloid via Peritumoral Injection. Pharmaceutics 2023; 15:2157. [PMID: 37631371 PMCID: PMC10457819 DOI: 10.3390/pharmaceutics15082157] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 08/11/2023] [Accepted: 08/15/2023] [Indexed: 08/27/2023] Open
Abstract
A keloid is a benign tumor manifested as abnormal fibroplasia on the surface of the skin. Curing keloids has become a major clinical challenge, and searching for new treatments and medications has become critical. In this study, we developed a LA67 liposome-loaded thermo-sensitive hydrogel (LA67-RL-Gel) with active targeting for treating keloids via peritumoral injection and explored the anti-keloid mechanism. Firstly, Arg-Gly-Asp (RGD) peptide-modified liposomes (LA67-RL) loaded with LA67 were prepared with a particle size of 105.9 nm and a Zeta potential of -27.4 mV, and an encapsulation efficiency of 89.6 ± 3.7%. We then constructed a thermo-sensitive hydrogel loaded with LA67-RL by poloxamer 407 and 188. The formulation was optimized through the Box-Behnken design, where the impact of the proportion of the ingredients on the quality of the hydrogel was evaluated entirely. The optimal formulation was 20.7% P407 and 2.1% P188, and the gelation time at 37 °C was 9.5 s. LA67-RL-Gel slowly released 92.2 ± 0.8% of LA67 at pH 6.5 PBS for 72 h. LA67-RL-Gel increased adhesion with KF cells; increased uptake; promoted KF cells apoptosis; inhibited cell proliferation; reduced α-SMA content; decreased collagen I, collagen III, and fibronectin deposition; inhibited angiogenesis; and modulated the keloid microenvironment, ultimately exerting anti-keloid effects. In summary, this simple, low-cost, and highly effective anti-keloid liposome hydrogel provides a novel approach for treating keloids and deserves further development.
Collapse
Affiliation(s)
- Hongshuang Wan
- Keloid Research Center, Yanbian University Hospital, Yanji 133000, China; (H.W.)
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; (S.W.)
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Shuangqing Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; (S.W.)
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Yanji 133000, China
| | - Chuying Li
- Keloid Research Center, Yanbian University Hospital, Yanji 133000, China; (H.W.)
| | - Bowen Zeng
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; (S.W.)
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Hao Wu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; (S.W.)
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Yanji 133000, China
| | - Chao Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; (S.W.)
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Liqing Chen
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; (S.W.)
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Mingji Jin
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; (S.W.)
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Wei Huang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; (S.W.)
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Yingda Zang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; (S.W.)
| | - Dongming Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; (S.W.)
| | - Zhonggao Gao
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; (S.W.)
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Yanji 133000, China
| | - Zhehu Jin
- Keloid Research Center, Yanbian University Hospital, Yanji 133000, China; (H.W.)
| |
Collapse
|
6
|
Zambra M, Ranđelović I, Talarico F, Borbély A, Svajda L, Tóvári J, Mező G, Bodero L, Colombo S, Arrigoni F, Fasola E, Gazzola S, Piarulli U. Optimizing the enzymatic release of MMAE from isoDGR-based small molecule drug conjugate by incorporation of a GPLG-PABC enzymatically cleavable linker. Front Pharmacol 2023; 14:1215694. [PMID: 37492088 PMCID: PMC10363981 DOI: 10.3389/fphar.2023.1215694] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 06/28/2023] [Indexed: 07/27/2023] Open
Abstract
Antibody-Drug Conjugates (ADCs) and Small Molecule-Drug Conjugates (SMDCs) represent successful examples of targeted drug-delivery technologies for overcoming unwanted side effects of conventional chemotherapy in cancer treatment. In both strategies, a cytotoxic payload is connected to the tumor homing moiety through a linker that releases the drug inside or in proximity of the tumor cell, and that represents a key component for the final therapeutic effect of the conjugate. Here, we show that the replacement of the Val-Ala-p-aminobenzyloxycarbamate linker with the Gly-Pro-Leu-Gly-p-aminobenzyloxycarbamate (GPLG-PABC) sequence as enzymatically cleavable linker in the SMDC bearing the cyclo[DKP-isoDGR] αVβ3 integrin ligand as tumor homing moiety and the monomethyl auristatin E (MMAE) as cytotoxic payload led to a 4-fold more potent anti-tumoral effect of the final conjugate on different cancer cell lines. In addition, the synthesized conjugate resulted to be significantly more potent than the free MMAE when tested following the "kiss-and-run" protocol, and the relative potency were clearly consistent with the expression of the αVβ3 integrin receptor in the considered cancer cell lines. In vitro enzymatic cleavage tests showed that the GPLG-PABC linker is cleaved by lysosomal enzymes, and that the released drug is observable already after 15 min of incubation. Although additional data are needed to fully characterize the releasing capacity of GPLG-PABC linker, our findings are of therapeutic significance since we are introducing an alternative to other well-established enzymatically sensitive peptide sequences that might be used in the future for generating more efficient and less toxic drug delivery systems.
Collapse
Affiliation(s)
- Marco Zambra
- Science and High Technology Department, University of Insubria, Como, Italy
| | - Ivan Ranđelović
- The National Tumor Biology Laboratory, Department of Experimental Pharmacology, National Institute of Oncology, Budapest, Hungary
| | - Francesco Talarico
- Science and High Technology Department, University of Insubria, Como, Italy
| | - Adina Borbély
- MTA-ELTE Lendület Ion Mobility Mass Spectrometry Research Group and Faculty of Science, Institute of Chemistry, ELTE Eötvös Loránd University, Budapest, Hungary
| | - Laura Svajda
- KINETO Lab Ltd., Budapest, Hungary
- Doctoral School of Pathological Sciences, Semmelweis University, Budapest, Hungary
| | - József Tóvári
- The National Tumor Biology Laboratory, Department of Experimental Pharmacology, National Institute of Oncology, Budapest, Hungary
| | - Gábor Mező
- ELKH-ELTE Research Group of Peptide Chemistry, Faculty of Science, Eötvös Loránd University, Budapest, Hungary
- Faculty of Science, Institute of Chemistry, Eötvös Loránd University, Budapest, Hungary
| | - Lizeth Bodero
- Science and High Technology Department, University of Insubria, Como, Italy
| | - Sveva Colombo
- Science and High Technology Department, University of Insubria, Como, Italy
- Department of Chemistry Organic and Bioorganic Chemistry, Bielefeld University, Bielefeld, Germany
| | - Federico Arrigoni
- Science and High Technology Department, University of Insubria, Como, Italy
| | - Elettra Fasola
- Science and High Technology Department, University of Insubria, Como, Italy
| | - Silvia Gazzola
- Science and High Technology Department, University of Insubria, Como, Italy
| | - Umberto Piarulli
- Science and High Technology Department, University of Insubria, Como, Italy
| |
Collapse
|
7
|
Yin L, Li X, Wang R, Zeng Y, Zeng Z, Xie T. Recent Research Progress of RGD Peptide–Modified Nanodrug Delivery Systems in Tumor Therapy. Int J Pept Res Ther 2023; 29:53. [DOI: 10.1007/s10989-023-10523-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/20/2023] [Indexed: 01/06/2025]
Abstract
AbstractThere have been great advancements in targeted nanodrug delivery systems for tumor therapy. Liposomes, polymeric nanoparticles, and inorganic nanoparticles are commonly employed as nanocarriers for drug delivery, and it has been found that arginine glycine aspartic acid (RGD) peptides and their derivatives can be used as ligands of integrin receptors to enhance the direct targeting ability. In this paper, we review the recent applications of RGD-modified liposomes, polymeric nanoparticles, and inorganic nanocarriers in cancer diagnosis and treatment, discuss the current challenges and prospects, and examine the progress made by the latest research on RGD peptide–modified nano delivery systems in cancer therapy. In recent years, RGD peptide–modified nanodrug delivery systems have been proven to have great potential in tumor therapy. Finally, we provide an overview of the current limitations and future directions of RGD peptide–modified nano-drug delivery systems for cancer therapy. This review aims to elucidate the contribution of RGD peptide–modified nanodrug delivery systems in the field of tumor therapy.
Collapse
|
8
|
Cheung CHP, Chong TH, Wei T, Liu H, Li X. Guanidine Additive Enabled Intermolecular ortho-Phthalaldehyde-Amine-Thiol Three-Component Reactions for Modular Constructions. Angew Chem Int Ed Engl 2023; 62:e202217150. [PMID: 36624047 DOI: 10.1002/anie.202217150] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 01/03/2023] [Accepted: 01/09/2023] [Indexed: 01/11/2023]
Abstract
Recently, ortho-phthalaldehyde (OPA) is experiencing a renascence for the modification of proteins and peptides through OPA-amine two-component reactions for bioconjugation and intramolecular OPA-amine-thiol three-component reactions for cyclization. Historically, small thiol molecules were used in large excess to allow for the intermolecular OPA-amine-thiol reaction forming 1-thio-isoindole derivatives. In this study, we discovered that guanidine could serve as an effective additive to switch the intermolecular OPA-amine-thiol three-component reaction to a stoichiometric process and enable the modular construction of peptide-peptide, and peptide-drug conjugate structures. Thus, 12 model peptide-peptide conjugates have been synthesized from unprotected peptides featuring all proteinogenic residues. Besides, 6 peptide-drug conjugates have been prepared in one step, with excellent conversions and isolated yields. In addition, a conjugate product has been further functionalized by utilizing a premodified OPA derivative, demonstrating the versatility and flexibility of this reaction.
Collapse
Affiliation(s)
- Carina Hey Pui Cheung
- Department of Chemistry, State Key Laboratory of Synthetic Chemistry, The University of Hong Kong, Hong Kong SAR, P. R. China
| | - Tin Hang Chong
- Department of Chemistry, State Key Laboratory of Synthetic Chemistry, The University of Hong Kong, Hong Kong SAR, P. R. China
| | - Tongyao Wei
- Department of Chemistry, State Key Laboratory of Synthetic Chemistry, The University of Hong Kong, Hong Kong SAR, P. R. China
| | - Han Liu
- Department of Chemistry, State Key Laboratory of Synthetic Chemistry, The University of Hong Kong, Hong Kong SAR, P. R. China
| | - Xuechen Li
- Department of Chemistry, State Key Laboratory of Synthetic Chemistry, The University of Hong Kong, Hong Kong SAR, P. R. China.,Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266237, P. R. China
| |
Collapse
|
9
|
Schnepel C, Moritzer A, Gäfe S, Montua N, Minges H, Nieß A, Niemann HH, Sewald N. Enzymatic Late-Stage Halogenation of Peptides. Chembiochem 2023; 24:e202200569. [PMID: 36259362 PMCID: PMC10099709 DOI: 10.1002/cbic.202200569] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 10/18/2022] [Indexed: 01/05/2023]
Abstract
The late-stage site-selective derivatisation of peptides has many potential applications in structure-activity relationship studies and postsynthetic modification or conjugation of bioactive compounds. The development of orthogonal methods for C-H functionalisation is crucial for such peptide derivatisation. Among them, biocatalytic methods are increasingly attracting attention. Tryptophan halogenases emerged as valuable catalysts to functionalise tryptophan (Trp), while direct enzyme-catalysed halogenation of synthetic peptides is yet unprecedented. Here, it is reported that the Trp 6-halogenase Thal accepts a wide range of amides and peptides containing a Trp moiety. Increasing the sequence length and reaction optimisation made bromination of pentapeptides feasible with good turnovers and a broad sequence scope, while regioselectivity turned out to be sequence dependent. Comparison of X-ray single crystal structures of Thal in complex with d-Trp and a dipeptide revealed a significantly altered binding mode for the peptide. The viability of this bioorthogonal approach was exemplified by halogenation of a cyclic RGD peptide.
Collapse
Affiliation(s)
- Christian Schnepel
- Organische und Bioorganische ChemieFakultät für ChemieUniversität BielefeldUniversitätsstraße 2533615BielefeldGermany
- Present address: Department of ChemistryManchester Institute of BiotechnologyThe University of Manchester131 Princess StreetManchesterM1 7DNUK
| | - Ann‐Christin Moritzer
- StrukturbiochemieFakultät für ChemieUniversität BielefeldUniversitätsstraße 2533615BielefeldGermany
| | - Simon Gäfe
- StrukturbiochemieFakultät für ChemieUniversität BielefeldUniversitätsstraße 2533615BielefeldGermany
| | - Nicolai Montua
- Organische und Bioorganische ChemieFakultät für ChemieUniversität BielefeldUniversitätsstraße 2533615BielefeldGermany
| | - Hannah Minges
- Organische und Bioorganische ChemieFakultät für ChemieUniversität BielefeldUniversitätsstraße 2533615BielefeldGermany
| | - Anke Nieß
- Organische und Bioorganische ChemieFakultät für ChemieUniversität BielefeldUniversitätsstraße 2533615BielefeldGermany
| | - Hartmut H. Niemann
- StrukturbiochemieFakultät für ChemieUniversität BielefeldUniversitätsstraße 2533615BielefeldGermany
| | - Norbert Sewald
- Organische und Bioorganische ChemieFakultät für ChemieUniversität BielefeldUniversitätsstraße 2533615BielefeldGermany
| |
Collapse
|
10
|
Mohri K, Nhat KPH, Zouda M, Warashina S, Wada Y, Watanabe Y, Tagami S, Mukai H. Lasso peptide microcin J25 variant containing RGD motif as a PET probe for integrin a v ß 3 in tumor imaging. Eur J Pharm Sci 2023; 180:106339. [PMID: 36414157 DOI: 10.1016/j.ejps.2022.106339] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 11/17/2022] [Accepted: 11/18/2022] [Indexed: 11/21/2022]
Abstract
Microcin J25 (MccJ25), a lasso peptide, has a unique 3-D interlocked structure that provides high stability under acidic conditions, at high temperatures, and in the presence of proteases. In this study, we generated a positron emission tomography (PET) probe based on MccJ25 analog with an RGD motif and investigated their pharmacokinetics and utility for integrin αvβ3 imaging in tumors. The MccJ25 variant with an RGD motif in the loop region and a lysine substitution at the C-terminus (MccJ25(RGDF)GtoK) was produced in E. coli transfected with plasmid DNA containing the MccJ25 biosynthetic gene cluster (mcjABCD). [64Cu]Cu-MccJ25(RGDF)GtoK was synthesized using the C-terminal lysine labeled with copper-64 (t1/2 = 12.7 h) via a bifunctional chelator; it showed stability in 90% mouse plasma for 45 min. Using PET imaging for integrin αvβ3 positive U87MG tumor bearing mice, [64Cu]Cu-MccJ25(RGDF)GtoK could clearly distinguish the tumor, and its accumulation was significantly higher than that of MccJ25(GIGT)GtoK without the binding motif for integrin αvβ3. Furthermore, MccJ25(RGDF)GtoK enabled visualization of only U87MG tumors but not MCF-7 tumors with low integrin αvβ3 expression in double tumor-bearing mice. In ex vivo biodistribution analysis, the integrin αvβ3 non-specific accumulation of [64Cu]Cu-MccJ25(RGDF)GtoK was significantly lower in various tissues, except for the kidneys, as compared to the control probe ([64Cu]Cu-cyclic RGD peptide). These results of the present study indicate that 64Cu-labeling methods are appropriate for the synthesis of MccJ25-based PET probes, and [64Cu]Cu-MccJ25 variants are useful tools for cancer molecular imaging.
Collapse
Affiliation(s)
- Kohta Mohri
- Laboratory for Molecular Delivery and Imaging Technology, RIKEN Center for Biosystems Dynamics Research, 6-7-3 Minatojima-Minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Kim Phuong Huynh Nhat
- Laboratory for Advanced Biomolecular Engineering, RIKEN Center for Biosystems Dynamics Research, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
| | - Maki Zouda
- Laboratory for Molecular Delivery and Imaging Technology, RIKEN Center for Biosystems Dynamics Research, 6-7-3 Minatojima-Minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Shota Warashina
- Laboratory for Molecular Delivery and Imaging Technology, RIKEN Center for Biosystems Dynamics Research, 6-7-3 Minatojima-Minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Yasuhiro Wada
- Laboratory for Pathophysiological and Health Science, RIKEN Center for Biosystems Dynamics Research, 6-7-3 Minatojima-Minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Yasuyoshi Watanabe
- Laboratory for Pathophysiological and Health Science, RIKEN Center for Biosystems Dynamics Research, 6-7-3 Minatojima-Minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Shunsuke Tagami
- Laboratory for Advanced Biomolecular Engineering, RIKEN Center for Biosystems Dynamics Research, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan.
| | - Hidefumi Mukai
- Laboratory for Molecular Delivery and Imaging Technology, RIKEN Center for Biosystems Dynamics Research, 6-7-3 Minatojima-Minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan; Department of Pharmaceutical Informatics, Graduate School of Biomedical Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki-shi, Nagasaki 852-8588, Japan.
| |
Collapse
|
11
|
Al Faruque H, Choi ES, Kim JH, Kim E. Enhanced effect of autologous EVs delivering paclitaxel in pancreatic cancer. J Control Release 2022; 347:330-346. [PMID: 35561870 DOI: 10.1016/j.jconrel.2022.05.012] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 05/02/2022] [Accepted: 05/05/2022] [Indexed: 10/18/2022]
Abstract
We engineered human pancreatic cancer cell (PANC-1)-derived extracellular vesicles (EVs) by conjugating the functional ligand RGD and magnetic nanoparticles (MNPs) onto EV surfaces (rmExo), for pancreatic cancer therapy. Paclitaxel (PTX) loaded into rmExo (rmExo-PTX) was intravenously injected into xenograft mice prepared using PANC-1 cells, which showed a significant reduction in tumor size compared to the free PTX-treated and control groups. The enhanced therapeutic effect was attributed to the modification of the surface of EVs using RGD, which has affinity for αvβ3 that is highly expressed in pancreatic cancer cells. Moreover, autologous EVs seemed to have more benefits in delivering PTX due to an unknown homing property to parent tumor cells, as exemplified by the reduced therapeutic effect of RGD-modified PANC-1 EVs on HT29 xenograft mice and RGD-modified U937 EVs on PANC-1 xenograft mice. The RGD-modified autologous EV vehicles were effective at penetrating and internalizing tumor cells, and eventually regressing the tumors, by mediating spontaneous removal of α-smooth muscle actin and collagen type 1 in the extracellular matrix of xenografts. Our results also identified an important molecule involved in the home-driving properties of PANC-1 EVs, integrin β3, which was expressed both on PANC-1 cells and the EVs derived from them. Additional therapeutic effect by permanent magnet near tumor xenograft was not observed in this study.
Collapse
Affiliation(s)
- Hasan Al Faruque
- Division of Bio-Fusion Research, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Techno-jungangdaero 333, Daegue 42988, Republic of Korea.
| | - Eun-Sook Choi
- Division of Electronic Information System Research, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Techno-jungangdaero 333, Daegue 42988, Republic of Korea.
| | - Jung-Hee Kim
- Division of Electronic Information System Research, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Techno-jungangdaero 333, Daegue 42988, Republic of Korea.
| | - Eunjoo Kim
- Division of Electronic Information System Research, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Techno-jungangdaero 333, Daegue 42988, Republic of Korea.
| |
Collapse
|
12
|
Paulus J, Sewald N. Synthesis and Evaluation of a Non-Peptide Small-Molecule Drug Conjugate Targeting Integrin αVβ3. Front Chem 2022; 10:869639. [PMID: 35480387 PMCID: PMC9035832 DOI: 10.3389/fchem.2022.869639] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 02/28/2022] [Indexed: 01/16/2023] Open
Abstract
An integrin αVβ3-targeting linear RGD mimetic containing a small-molecule drug conjugate (SMDC) was synthesized by combining the antimitotic agent monomethyl auristatin E (MMAE), an enzymatically cleavable Val-Ala-PABC linker with a linear conjugable RGD mimetic. The structure proposal for the conjugable RGD mimetic was suggested upon the DAD mapping analysis of a previously synthesized small-molecule RGD mimetic array based on a tyrosine scaffold. Therefore, a diversifying strategy was developed as well as a novel method for the partial hydrogenation of pyrimidines in the presence of the hydrogenolytically cleavable Cbz group. The small-molecule RGD mimetics were evaluated in an ELISA-like assay, and the structural relationships were analyzed by DAD mapping revealing activity differences induced by structural changes as visualized in dependence on special structural motifs. This provided a lead structure for generation of an SMDC containing the antimitotic drug MMAE. The resulting SMDC containing a linear RGD mimetic was tested in a cell adhesion and an in vitro cell viability assay in comparison to reference SMDCs containing cRGDfK or cRADfK as the homing device. The linear RGD SMDC and the cRGDfK SMDC inhibited adhesion of αVβ3-positive WM115 cells to vitronectin with IC50 values in the low µM range, while no effect was observed for the αVβ3-negative M21-L cell line. The cRADfK SMDC used as a negative control was about 30-fold less active in the cell adhesion assay than the cRGDfK SMDC. Conversely, both the linear RGD SMDC and the cRGDfK SMDC are about 55-fold less cytotoxic than MMAE against the αVβ3-positive WM115 cell line with IC50 values in the nM range, while the cRADfK SMDC is 150-fold less cytotoxic than MMAE. Hence, integrin binding also influences the antiproliferative activity giving a targeting index of 2.8.
Collapse
|
13
|
Aliouat H, Peng Y, Waseem Z, Wang S, Zhou W. Pure DNA scaffolded drug delivery systems for cancer therapy. Biomaterials 2022; 285:121532. [DOI: 10.1016/j.biomaterials.2022.121532] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 04/04/2022] [Accepted: 04/15/2022] [Indexed: 02/07/2023]
|
14
|
Gruß H, Feiner RC, Mseya R, Schröder DC, Jewgiński M, Müller KM, Latajka R, Marion A, Sewald N. Peptide stapling by late-stage Suzuki–Miyaura cross-coupling. Beilstein J Org Chem 2022; 18:1-12. [PMID: 35047078 PMCID: PMC8744458 DOI: 10.3762/bjoc.18.1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 12/09/2021] [Indexed: 12/19/2022] Open
Abstract
The development of peptide stapling techniques to stabilise α-helical secondary structure motifs of peptides led to the design of modulators of protein–protein interactions, which had been considered undruggable for a long time. We disclose a novel approach towards peptide stapling utilising macrocyclisation by late-stage Suzuki–Miyaura cross-coupling of bromotryptophan-containing peptides of the catenin-binding domain of axin. Optimisation of the linker length in order to find a compromise between both sufficient linker rigidity and flexibility resulted in a peptide with an increased α-helicity and enhanced binding affinity to its native binding partner β-catenin. An increased proteolytic stability against proteinase K has been demonstrated.
Collapse
Affiliation(s)
- Hendrik Gruß
- Department of Chemistry, Bielefeld University, Universitätsstr. 25, 33615 Bielefeld, Germany
| | - Rebecca C Feiner
- Department of Technology, Bielefeld University, Universitätsstr. 25, 33615 Bielefeld, Germany
| | - Ridhiwan Mseya
- Department of Chemistry, Middle East Technical University, 06800, Ankara, Turkey
| | - David C Schröder
- Department of Chemistry, Bielefeld University, Universitätsstr. 25, 33615 Bielefeld, Germany
| | - Michał Jewgiński
- Department of Bioorganic Chemistry, Wrocław University of Science and Technology, Wybrzeze Wyspianskiego 27, 50-370 Wrocław, Poland
| | - Kristian M Müller
- Department of Technology, Bielefeld University, Universitätsstr. 25, 33615 Bielefeld, Germany
| | - Rafał Latajka
- Department of Bioorganic Chemistry, Wrocław University of Science and Technology, Wybrzeze Wyspianskiego 27, 50-370 Wrocław, Poland
| | - Antoine Marion
- Department of Chemistry, Middle East Technical University, 06800, Ankara, Turkey
| | - Norbert Sewald
- Department of Chemistry, Bielefeld University, Universitätsstr. 25, 33615 Bielefeld, Germany
| |
Collapse
|
15
|
Mizuno Y, Kimura K, Onoe S, Shukuri M, Kuge Y, Akizawa H. Influence of Linker Molecules in Hexavalent RGD Peptides on Their Multivalent Interactions with Integrin α vβ 3. J Med Chem 2021; 64:16008-16019. [PMID: 34730982 DOI: 10.1021/acs.jmedchem.1c01396] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Multivalent RGD peptides have been used as an excellent targeting vector to integrin αvβ3-positive tumors. However, little attention has been paid to the influence of linker molecules in multivalent RGD peptides on their dissociation kinetics from tumor cells. In this study, we evaluated the dissociation kinetics of 99mTc-labeled hexavalent RGD peptides which have (CH2-CH2-O)n (n = 4, [99mTc][Tc(L1)6]+ and n = 12, [99mTc][Tc(L2)6]+) or (DPro-Gly)n (n = 1, [99mTc][Tc(L3)6]+; n = 6, [99mTc][Tc(L4)6]+; and n = 9, [99mTc][Tc(L5)6]+) as a linker molecule. The results showed that [99mTc][Tc(L4)6]+ and [99mTc][Tc(L5)6]+ displayed slower dissociation kinetics and [99mTc][Tc(L4)6]+ showed exceptionally high in vitro cellular uptake (203.1 ± 16.7% dose/mg protein) and the highest tumor to blood ratio (138.1 ± 26.3 at 4 h p.i.) in tumor bearing nude mice. These findings indicate that the use of appropriate length of (DPro-Gly)n would maximize the binding of multivalent RGD peptides to clustered integrin αvβ3.
Collapse
Affiliation(s)
- Yuki Mizuno
- Laboratory of Physical Chemistry, Showa Pharmaceutical University, Machida 194-8543, Japan.,Central Institute of Isotope Science, Hokkaido University, Sapporo 060-0815, Japan.,Department of Biomedical Imaging, Graduate School of Biomedical Science and Engineering, Hokkaido University, Sapporo 060-8638, Japan
| | - Kohta Kimura
- Laboratory of Physical Chemistry, Showa Pharmaceutical University, Machida 194-8543, Japan
| | - Satoru Onoe
- Laboratory of Physical Chemistry, Showa Pharmaceutical University, Machida 194-8543, Japan
| | - Miho Shukuri
- Laboratory of Physical Chemistry, Showa Pharmaceutical University, Machida 194-8543, Japan
| | - Yuji Kuge
- Central Institute of Isotope Science, Hokkaido University, Sapporo 060-0815, Japan.,Department of Biomedical Imaging, Graduate School of Biomedical Science and Engineering, Hokkaido University, Sapporo 060-8638, Japan
| | - Hiromichi Akizawa
- Laboratory of Physical Chemistry, Showa Pharmaceutical University, Machida 194-8543, Japan
| |
Collapse
|
16
|
|
17
|
Kamalkazemi E, Abedi-Gaballu F, Mohammad Hosseini TF, Mohammadi A, Mansoori B, Dehghan G, Baradaran B, Sheibani N. Glimpse into Cellular Internalization and Intracellular Trafficking of Lipid-Based Nanoparticles in Cancer Cells. Anticancer Agents Med Chem 2021; 22:1897-1912. [PMID: 34488605 DOI: 10.2174/1871520621666210906101421] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 06/14/2021] [Accepted: 06/27/2021] [Indexed: 11/22/2022]
Abstract
Lipid-based nanoparticles as drug delivery carriers have been mainly used for delivery of anti-cancer therapeutic agents. Lipid-based nanoparticles, due to their smaller particle size and similarity to cell membranes, are readily internalized into cancer cells. Interestingly, cancer cells also overexpress receptors for specific ligands including folic acid, hyaluronic acid, and transferrin on their surface. This allows the use of these ligands for surface modification of the lipid-based nanoparticle. These modifications then allow the specific recognition of these ligand-coated nanoparticles by their receptors on cancer cells allowing the targeted gradual intracellular accumulation of the functionalized nanoplatforms. These interactions could eventually enhance the internalization of desired drugs via increasing ligand-receptor mediated cellular uptake of the nanoplatforms. The cellular internalization of the nanoplatforms also varies and depends on their physicochemical properties including particle size, zeta potential, and shape. The cellular uptake is also influenced by the types of ligand internalization pathway utilized by cells such as phagocytosis, macropinocytosis, and multiple endocytosis pathways. In this review, we will classify and discuss lipid based nanoparticles engineered to express specific ligands, and are recognized by their receptors on cancer cell, and their cellular internalization pathways. Moreover, the intracellular fate of nanoparticles decorated with specific ligands and the best internalization pathways (caveolae mediated endocytosis) for safe cargo delivery will be discussed.
Collapse
Affiliation(s)
- Elham Kamalkazemi
- Department of Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz. Iran
| | | | | | - Ali Mohammadi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz. Iran
| | - Behzad Mansoori
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz. Iran
| | - Gholamreza Dehghan
- Department of Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz. Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz. Iran
| | - Nader Sheibani
- Departments of Ophthalmology and Visual Sciences, Biomedical Engineering, and Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI . United States
| |
Collapse
|
18
|
Schneider H, Englert S, Macarrón Palacios A, Lerma Romero JA, Ali A, Avrutina O, Kolmar H. Synthetic Integrin-Targeting Dextran-Fc Hybrids Efficiently Inhibit Tumor Proliferation In Vitro. Front Chem 2021; 9:693097. [PMID: 34368077 PMCID: PMC8339797 DOI: 10.3389/fchem.2021.693097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 06/21/2021] [Indexed: 11/13/2022] Open
Abstract
Herein, we present the design, synthesis, and biological evaluation of novel integrin-targeting molecular hybrids combining RGD peptides and a potent cytotoxin presented on dextran polysaccharides. Based on an aglycosylated Fc as a centerpiece, endosomal-cleavable cytotoxic agent monomethyl auristatin E (MMAE) and dextran as multimerization site were covalently connected by two bioorthogonal enzyme-mediated reactions site-specifically. Decoration of dextran with cyclic RGD peptides, introduced by copper “click” reaction, resulted in the final constructs with the potential to kill integrin-overexpressing tumor cells. We found that these modifications had little impact on the stability of the Fc scaffold and the RGD-bearing construct showed good binding properties of αvβ3-expressing U87MG cells. Furthermore, the construct showed a remarkable antiproliferative activity. These results demonstrate the general capability of our design to provoke receptor-mediated endocytosis upon binding to the cellular surface, followed by endosomal cleavage of the linkage between Fc-dextran and MMAE and its subsequent release. Our approach opens new avenues to transcribe small molecule binders into tailor-made multimeric molecular hybrids with antitumor potential.
Collapse
Affiliation(s)
- Hendrik Schneider
- Institute for Organic Chemistry and Biochemistry, Technical University Darmstadt, Darmstadt, Germany
| | - Simon Englert
- Institute for Organic Chemistry and Biochemistry, Technical University Darmstadt, Darmstadt, Germany
| | - Arturo Macarrón Palacios
- Institute for Organic Chemistry and Biochemistry, Technical University Darmstadt, Darmstadt, Germany
| | | | - Ataurehman Ali
- Institute for Organic Chemistry and Biochemistry, Technical University Darmstadt, Darmstadt, Germany
| | - Olga Avrutina
- Institute for Organic Chemistry and Biochemistry, Technical University Darmstadt, Darmstadt, Germany
| | - Harald Kolmar
- Institute for Organic Chemistry and Biochemistry, Technical University Darmstadt, Darmstadt, Germany
| |
Collapse
|
19
|
Bodero L, Parente S, Arrigoni F, Klimpel A, Neundorf I, Gazzola S, Piarulli U. Synthesis and Biological Evaluation of an
iso
DGR‐Paclitaxel Conjugate Containing a Cell‐Penetrating Peptide to Promote Cellular Uptake. European J Org Chem 2021. [DOI: 10.1002/ejoc.202100241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Affiliation(s)
- Lizeth Bodero
- Dipartimento di Scienza e Alta Tecnologia Università degli Studi dell'Insubria Via Valleggio 11 22100 Como Italy
| | - Sara Parente
- Dipartimento di Scienza e Alta Tecnologia Università degli Studi dell'Insubria Via Valleggio 11 22100 Como Italy
| | - Federico Arrigoni
- Dipartimento di Scienza e Alta Tecnologia Università degli Studi dell'Insubria Via Valleggio 11 22100 Como Italy
| | - Annika Klimpel
- University of Cologne Department of Chemistry Institute for Biochemistry Zuelpicher Str. 47a 50674 Cologne Germany
| | - Ines Neundorf
- University of Cologne Department of Chemistry Institute for Biochemistry Zuelpicher Str. 47a 50674 Cologne Germany
| | - Silvia Gazzola
- Dipartimento di Scienza e Alta Tecnologia Università degli Studi dell'Insubria Via Valleggio 11 22100 Como Italy
| | - Umberto Piarulli
- Dipartimento di Scienza e Alta Tecnologia Università degli Studi dell'Insubria Via Valleggio 11 22100 Como Italy
| |
Collapse
|
20
|
Brennecke B, Wang Q, Haap W, Grether U, Hu HY, Nazaré M. DOTAM-Based, Targeted, Activatable Fluorescent Probes for the Highly Sensitive and Selective Detection of Cancer Cells. Bioconjug Chem 2021; 32:702-712. [PMID: 33691062 DOI: 10.1021/acs.bioconjchem.0c00699] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The utilization of an activatable, substrate-based probe design in combination with a cellular targeting approach has been rarely explored for cancer imaging on a small-molecule basis, although such probes could benefit from advantages of both concepts. Cysteine proteases like cathepsin S are known to be involved in fundamental processes associated with tumor development and progression and thus are valuable cancer markers. We report the development of a combined dual functional DOTAM-based, RGD-targeted internally quenched fluorescent probe that is activated by cathepsin S. The probe exhibits excellent in vitro activation kinetics which can be fully translated to human cancer cell lines. We demonstrate that the targeted, activatable probe is superior to its nontargeted analog, exhibiting improved uptake into ανβ3-integrin expressing human sarcoma cells (HT1080) and significantly higher resultant fluorescence staining. However, profound activation was also found in cancer cells with a lower integrin expression level, whereas in healthy cells almost no probe activation could be observed, highlighting the high selectivity of our probe toward cancer cells. These auspicious results show the outstanding potential of the dual functionality concept combining a substrate-based probe design with a targeting approach, which could form the basis for highly sensitive and selective in vivo imaging probes.
Collapse
Affiliation(s)
- Benjamin Brennecke
- Medicinal Chemistry, Leibniz-Forschungsinstitut für Molekulare Pharmakologie Berlin, 13125 Berlin, Germany
| | - Qinghua Wang
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Institute of Materia Medica, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100050, China
| | - Wolfgang Haap
- Roche Innovation Center Basel, Pharma Research and Early Development, F. Hoffmann-La Roche Ltd., 4070 Basel, Switzerland
| | - Uwe Grether
- Roche Innovation Center Basel, Pharma Research and Early Development, F. Hoffmann-La Roche Ltd., 4070 Basel, Switzerland
| | - Hai-Yu Hu
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Institute of Materia Medica, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100050, China
| | - Marc Nazaré
- Medicinal Chemistry, Leibniz-Forschungsinstitut für Molekulare Pharmakologie Berlin, 13125 Berlin, Germany
| |
Collapse
|
21
|
Kemker I, Schröder DC, Feiner RC, Müller KM, Marion A, Sewald N. Tuning the Biological Activity of RGD Peptides with Halotryptophans†. J Med Chem 2020; 64:586-601. [PMID: 33356253 DOI: 10.1021/acs.jmedchem.0c01536] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
An array of l- and d-halotryptophans with different substituents at the indole moiety was synthesized employing either enzymatic halogenation by halogenases or incorporation of haloindoles using tryptophan synthase. Introduction of these Trp derivatives into RGD peptides as a benchmark system was performed to investigate their influence on bioactivity. Halotryptophan-containing RGD peptides display increased affinity toward integrin αvβ3 and enhanced selectivity over integrin α5β1. In addition, bromotryptophan was exploited as a platform for late-stage diversification by Suzuki-Miyaura cross-coupling (SMC), resulting in new-to-nature biaryl motifs. These peptides show enhanced affinity toward αvβ3, good affinity to αvβ8, and remarkable selectivity over α5β1 and αIIbβ3 while featuring fluorogenic properties. Their feasibility as a probe was demonstrated in vitro. Extensive molecular dynamics simulations were undertaken to elucidate NMR and high-performance liquid chromatography (HPLC) data for these late-stage diversified cyclic RGD peptides and to further characterize their conformational preferences.
Collapse
Affiliation(s)
- Isabell Kemker
- Organic and Bioorganic Chemistry, Department of Chemistry, Bielefeld University, Universitätsstraße 25, 33615 Bielefeld, Germany
| | - David C Schröder
- Organic and Bioorganic Chemistry, Department of Chemistry, Bielefeld University, Universitätsstraße 25, 33615 Bielefeld, Germany
| | - Rebecca C Feiner
- Cellular and Molecular Biotechnology, Faculty of Technology, Bielefeld University, Universitätsstraße 25, 33615 Bielefeld, Germany
| | - Kristian M Müller
- Cellular and Molecular Biotechnology, Faculty of Technology, Bielefeld University, Universitätsstraße 25, 33615 Bielefeld, Germany
| | - Antoine Marion
- Department of Chemistry, Middle East Technical University, Kimya Bölümü Üniversiteler Mah., Çankaya, 06800 Ankara, Turkey
| | - Norbert Sewald
- Organic and Bioorganic Chemistry, Department of Chemistry, Bielefeld University, Universitätsstraße 25, 33615 Bielefeld, Germany.,Department of Chemistry, Middle East Technical University, Kimya Bölümü Üniversiteler Mah., Çankaya, 06800 Ankara, Turkey
| |
Collapse
|
22
|
Dachwitz S, Duwe DH, Wang YH, Gruß H, Hannappel Y, Hellweg T, Sewald N. Suzuki-Miyaura Cross-Coupling of Bromotryptophan Derivatives at Ambient Temperature. Chemistry 2020; 26:16357-16364. [PMID: 32639079 PMCID: PMC7756874 DOI: 10.1002/chem.202002454] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 06/23/2020] [Indexed: 02/06/2023]
Abstract
Mild reaction conditions are highly desirable for bio‐orthogonal side chain derivatizations of amino acids, peptides or proteins due to the sensitivity of these substrates. Transition metal catalysed cross‐couplings such as Suzuki–Miyaura reactions are highly versatile, but usually require unfavourable reaction conditions, in particular, when applied with aryl bromides. Ligand‐free solvent‐stabilised Pd‐nanoparticles represent an efficient and sustainable alternative to conventional phosphine‐based catalysts, because the cross‐coupling can be performed at considerably lower temperature. We report on the application of such a highly reactive heterogeneous catalyst for the Suzuki–Miyaura cross‐coupling of brominated tryptophan derivatives. The solvent‐stabilised Pd‐nanoparticles are even more efficient than the literature‐known ADHP‐Pd precatalyst. Interestingly, the latter also leads to the formation of quasi‐homogeneous Pd‐nanoparticles as the catalytic species. One advantage of our approach is the compatibility with aqueous and aerobic conditions at near‐ambient temperatures and short reaction times of only 2 h. The influence of different Nα‐protecting groups, boronic acids as well as the impact of different amino acid side chains in bromotryptophan‐containing peptides has been studied. Notably, a surprising acceleration of the catalysis was observed when palladium‐coordinating side chains were present in proximal positions.
Collapse
Affiliation(s)
- Steffen Dachwitz
- Department of Chemistry, Organic and Bioorganic Chemistry, Bielefeld University, Universitätsstraße 25, 33615, Bielefeld, Germany
| | - Dario H Duwe
- Department of Chemistry, Organic and Bioorganic Chemistry, Bielefeld University, Universitätsstraße 25, 33615, Bielefeld, Germany
| | - Yating Hong Wang
- Department of Chemistry, Organic and Bioorganic Chemistry, Bielefeld University, Universitätsstraße 25, 33615, Bielefeld, Germany
| | - Hendrik Gruß
- Department of Chemistry, Organic and Bioorganic Chemistry, Bielefeld University, Universitätsstraße 25, 33615, Bielefeld, Germany
| | - Yvonne Hannappel
- Department of Chemistry, Physical Chemistry, Bielefeld University, Universitätsstraße 25, 33615, Bielefeld, Germany
| | - Thomas Hellweg
- Department of Chemistry, Physical Chemistry, Bielefeld University, Universitätsstraße 25, 33615, Bielefeld, Germany
| | - Norbert Sewald
- Department of Chemistry, Organic and Bioorganic Chemistry, Bielefeld University, Universitätsstraße 25, 33615, Bielefeld, Germany
| |
Collapse
|
23
|
Borbély A, Thoreau F, Figueras E, Kadri M, Coll J, Boturyn D, Sewald N. Synthesis and Biological Characterization of Monomeric and Tetrameric RGD-Cryptophycin Conjugates. Chemistry 2020; 26:2602-2605. [PMID: 31943410 PMCID: PMC7064988 DOI: 10.1002/chem.201905437] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Indexed: 01/28/2023]
Abstract
The effective delivery of cytotoxic agents to tumor cells is a key challenge in anticancer therapy. Multivalent integrinspecific ligands are considered a promising tool to increase the binding affinity, selectivity, and internalization efficiency of small-molecule drug conjugates. Herein, we report the synthesis and biological evaluation of a multimeric conjugate containing the high-affinity integrin αv β3 binding ligand RAFT-c(RGDfK)4 , a lysosomally cleavable Val-Cit linker, and cryptophycin-55 glycinate, a potent inhibitor of tubulin polymerization. In vitro cytotoxicity assays verified that the multimeric RGD-cryptophycin conjugate displays improved potency compared to the monomeric analogue in integrin αv β3 overexpressing tumor cell lines, while significantly reduced activity was observed in the integrin-negative cell line.
Collapse
Affiliation(s)
- Adina Borbély
- Organic and Bioorganic ChemistryDepartment of ChemistryBielefeld UniversityUniversitätsstraße 2533615BielefeldGermany
| | - Fabien Thoreau
- CNRS, Department of Molecular ChemistryUniversity Grenoble Alpes, UMR 525038000GrenobleFrance
| | - Eduard Figueras
- Organic and Bioorganic ChemistryDepartment of ChemistryBielefeld UniversityUniversitätsstraße 2533615BielefeldGermany
| | - Malika Kadri
- Institute for Advanced BiosciencesUniversity Grenoble Alpes, INSERM U1209—UMR CNRS 530938700GrenobleFrance
| | - Jean‐Luc Coll
- Institute for Advanced BiosciencesUniversity Grenoble Alpes, INSERM U1209—UMR CNRS 530938700GrenobleFrance
| | - Didier Boturyn
- CNRS, Department of Molecular ChemistryUniversity Grenoble Alpes, UMR 525038000GrenobleFrance
| | - Norbert Sewald
- Organic and Bioorganic ChemistryDepartment of ChemistryBielefeld UniversityUniversitätsstraße 2533615BielefeldGermany
| |
Collapse
|
24
|
Abstract
Monomeric RGD peptides show unspecific fluid-phase uptake in cells, whereas multimeric RGD peptides are thought to be internalized by integrin-mediated endocytosis. However, a potential correlation between uptake mechanism and molecular mass has been neglected so far. A dual derivatization of peptide c(RGDw(7Br)K) was performed to investigate this. A fluorescent probe was installed by chemoselective Suzuki-Miyaura cross-coupling of the 7-bromotryptophan and a poly(ethylene glycol) (PEG) linker was attached to the lysine residue. Flow cytometry and live cell imaging confirmed unspecific uptake of the small, non-PEGylated peptide, whereas the PEG5000 peptide conjugate unveiled a selective internalization by M21 cells overexpressing αv β3 and no uptake in αv -deficient M21L cells.
Collapse
Affiliation(s)
- Isabell Kemker
- Organische und Bioorganische ChemieFakultät für ChemieUniversität BielefeldUniversitätsstrasse 2533615BielefeldGermany
| | - Rebecca C. Feiner
- Zelluläre und Molekulare BiotechnologieTechnische FakultätUniversität BielefeldUniversitätsstrasse 2533615BielefeldGermany
| | - Kristian M. Müller
- Zelluläre und Molekulare BiotechnologieTechnische FakultätUniversität BielefeldUniversitätsstrasse 2533615BielefeldGermany
| | - Norbert Sewald
- Organische und Bioorganische ChemieFakultät für ChemieUniversität BielefeldUniversitätsstrasse 2533615BielefeldGermany
| |
Collapse
|