Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (394)

Search Parameters:
Keywords = peptide-drug conjugates

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
18 pages, 3441 KiB  
Review
Epidermal Growth Factor Receptor (EGFR)-Targeting Peptides and Their Applications in Tumor Imaging Probe Construction: Current Advances and Future Perspectives
by Lu Huang, Ying Dong, Jinhang Li, Xinyu Yang, Xiaoqiong Li, Jia Wu, Jinhua Huang, Qiaoxuan Zhang, Zemin Wan, Shuzhi Hu, Ruibing Feng, Guodong Li, Xianzhang Huang and Pengwei Zhang
Biology 2025, 14(8), 1011; https://doi.org/10.3390/biology14081011 - 7 Aug 2025
Abstract
The epidermal growth factor receptor (EGFR) is a key target for both cancer diagnosis and therapeutic interventions. Assessing EGFR expression before therapy has become routine in clinical practice, yet current methods like biopsy and immunohistochemistry (IHC) have significant limitations, including invasiveness, limited repeatability, [...] Read more.
The epidermal growth factor receptor (EGFR) is a key target for both cancer diagnosis and therapeutic interventions. Assessing EGFR expression before therapy has become routine in clinical practice, yet current methods like biopsy and immunohistochemistry (IHC) have significant limitations, including invasiveness, limited repeatability, and lack of real-time, whole-body data. EGFR-targeted imaging has emerged as a promising alternative. EGFR-targeting peptides, owing to their favorable physicochemical properties and versatility, are increasingly being explored for a variety of applications, including molecular imaging, drug delivery, and targeted therapy. Recent advances have demonstrated the potential of EGFR-targeting peptides conjugated to imaging probes for non-invasive, real-time in vivo tumor detection, precision therapy, and surgical guidance. Here, we provide a comprehensive overview of the latest progress in EGFR-targeting peptides development, with a particular focus on their application in the development of molecular imaging agents, including fluorescence imaging, PET/CT, magnetic resonance imaging, and multimodal imaging. Furthermore, we examine the challenges and future directions concerning the development and clinical application of EGFR-targeting peptide-based imaging probes. Finally, we highlight emerging technologies such as artificial intelligence, mutation-specific peptides, and multimodal imaging platforms, which offer significant potential for advancing the diagnosis and treatment of EGFR-targeted cancers. Full article
Show Figures

Figure 1

47 pages, 7003 KiB  
Review
Phthalocyanines Conjugated with Small Biologically Active Compounds for the Advanced Photodynamic Therapy: A Review
by Kyrylo Chornovolenko and Tomasz Koczorowski
Molecules 2025, 30(15), 3297; https://doi.org/10.3390/molecules30153297 - 6 Aug 2025
Abstract
Phthalocyanines (Pcs) are well-established photosensitizers in photodynamic therapy, valued for their strong light absorption, high singlet oxygen generation, and photostability. Recent advances have focused on covalently conjugating Pcs, particularly zinc phthalocyanines (ZnPcs), with a wide range of small bioactive molecules to improve selectivity, [...] Read more.
Phthalocyanines (Pcs) are well-established photosensitizers in photodynamic therapy, valued for their strong light absorption, high singlet oxygen generation, and photostability. Recent advances have focused on covalently conjugating Pcs, particularly zinc phthalocyanines (ZnPcs), with a wide range of small bioactive molecules to improve selectivity, efficacy, and multifunctionality. These conjugates combine light-activated reactive oxygen species (ROS) production with targeted delivery and controlled release, offering enhanced treatment precision and reduced off-target toxicity. Chemotherapeutic agent conjugates, including those with erlotinib, doxorubicin, tamoxifen, and camptothecin, demonstrate receptor-mediated uptake, pH-responsive release, and synergistic anticancer effects, even overcoming multidrug resistance. Beyond oncology, ZnPc conjugates with antibiotics, anti-inflammatory drugs, antiparasitics, and antidepressants extend photodynamic therapy’s scope to antimicrobial and site-specific therapies. Targeting moieties such as folic acid, biotin, arginylglycylaspartic acid (RGD) and epidermal growth factor (EGF) peptides, carbohydrates, and amino acids have been employed to exploit overexpressed receptors in tumors, enhancing cellular uptake and tumor accumulation. Fluorescent dye and porphyrinoid conjugates further enrich these systems by enabling imaging-guided therapy, efficient energy transfer, and dual-mode activation through pH or enzyme-sensitive linkers. Despite these promising strategies, key challenges remain, including aggregation-induced quenching, poor aqueous solubility, synthetic complexity, and interference with ROS generation. In this review, the examples of Pc-based conjugates were described with particular interest on the synthetic procedures and optical properties of targeted compounds. Full article
(This article belongs to the Section Organic Chemistry)
Show Figures

Figure 1

18 pages, 814 KiB  
Review
Fighting HER2 in Gastric Cancer: Current Approaches and Future Landscapes
by Margherita Ratti, Chiara Citterio, Elena Orlandi, Stefano Vecchia, Elisa Anselmi, Ilaria Toscani, Martina Rotolo, Massimiliano Salati and Michele Ghidini
Int. J. Mol. Sci. 2025, 26(15), 7285; https://doi.org/10.3390/ijms26157285 - 28 Jul 2025
Viewed by 294
Abstract
Gastric cancer (GC) remains a major cause of cancer-related mortality worldwide, with human epidermal growth factor receptor 2 (HER2)-positive disease representing a clinically relevant subset. Trastuzumab combined with chemotherapy is the standard first-line treatment in advanced settings, following the landmark ToGA trial. However, [...] Read more.
Gastric cancer (GC) remains a major cause of cancer-related mortality worldwide, with human epidermal growth factor receptor 2 (HER2)-positive disease representing a clinically relevant subset. Trastuzumab combined with chemotherapy is the standard first-line treatment in advanced settings, following the landmark ToGA trial. However, resistance to trastuzumab has emerged as a significant limitation, prompting the need for more effective second-line therapies. Trastuzumab deruxtecan, a novel antibody–drug conjugate (ADC) composed of trastuzumab linked to a cytotoxic payload, has demonstrated promising efficacy in trastuzumab-refractory, HER2-positive GC, including cases with heterogeneous HER2 expression. Other HER2-targeted ADCs are also under investigation as potential alternatives. In addition, strategies to overcome resistance include HER2-specific immune-based therapies, such as peptide vaccines and chimeric antigen receptor T cell therapies, as well as antibodies targeting distinct HER2 domains or downstream signaling pathways like PI3K/AKT. These emerging approaches aim to improve efficacy in both HER2-high and HER2-low GC. As HER2-targeted treatments evolve, addressing resistance mechanisms and optimizing therapy for broader patient populations is critical. This review discusses current and emerging HER2-directed strategies in GC, focusing on trastuzumab deruxtecan and beyond, and outlines future directions to improve outcomes for patients with HER2-positive GC across all clinical settings. Full article
(This article belongs to the Section Molecular Oncology)
Show Figures

Figure 1

24 pages, 2043 KiB  
Review
Boosting AMPs’ Power: From Structural Engineering to Nanotechnology-Based Delivery
by Oluwasegun Eric Ajayi, Rosa Bellavita, Lorenzo Emiliano Imbò, Sara Palladino, Simone Braccia, Annarita Falanga and Stefania Galdiero
Molecules 2025, 30(14), 2979; https://doi.org/10.3390/molecules30142979 - 15 Jul 2025
Viewed by 440
Abstract
Antimicrobial peptides (AMPs) represent a powerful support to conventional antibiotics in addressing the global challenge of antimicrobial resistance (AMR). Their broad-spectrum antimicrobial activity and unique mechanisms of action enable diverse potential applications, including combating multidrug-resistant pathogens, immune modulation, and cancer therapy. Their clinical [...] Read more.
Antimicrobial peptides (AMPs) represent a powerful support to conventional antibiotics in addressing the global challenge of antimicrobial resistance (AMR). Their broad-spectrum antimicrobial activity and unique mechanisms of action enable diverse potential applications, including combating multidrug-resistant pathogens, immune modulation, and cancer therapy. Their clinical implementation is hindered by challenges such as toxicity, instability, and high production costs. Recent advances in AMP design, optimization, and delivery mechanisms such as nanoparticle conjugation and rational engineering have enhanced their efficacy, stability, and specificity. Integrating AMPs into precision medicine and combining them with existing therapies promises to overcome current limitations. With ongoing advancements, AMPs have the potential to redefine infection management and possibly other medical problems. Full article
(This article belongs to the Special Issue Women’s Special Issue Series: Molecules)
Show Figures

Figure 1

17 pages, 3065 KiB  
Article
Matrix Metalloproteinase-2-Responsive Peptide-Modified Cleavable PEGylated Liposomes for Paclitaxel Delivery
by Xingyu Zhao and Yinghuan Li
Pharmaceuticals 2025, 18(7), 1042; https://doi.org/10.3390/ph18071042 - 15 Jul 2025
Viewed by 517
Abstract
Background/Objectives: PEGylated liposomes are widely recognized for their biocompatibility and capacity to extend systemic circulation via “stealth” properties. However, the PEG corona often limits tumor penetration and cellular internalization. Targeting matrix metalloproteinase-2 (MMP-2), frequently upregulated in breast cancer stroma, presents an opportunity [...] Read more.
Background/Objectives: PEGylated liposomes are widely recognized for their biocompatibility and capacity to extend systemic circulation via “stealth” properties. However, the PEG corona often limits tumor penetration and cellular internalization. Targeting matrix metalloproteinase-2 (MMP-2), frequently upregulated in breast cancer stroma, presents an opportunity to enhance tissue-specific drug delivery. In this study, we engineered MMP-2-responsive GPLGVRG peptide-modified cleavable PEGylated liposomes for targeted paclitaxel (PTX) delivery. Methods: Molecular docking simulations employed the MMP-2 crystal structure (PDB ID: 7XJO) to assess GPLGVRG peptide binding affinity. A cleavable, enzyme-sensitive peptide-PEG conjugate (Chol-PEG2K-GPLGVRG-PEG5K) was synthesized via small-molecule liquid-phase synthesis and characterized by 1H NMR and MALDI-TOF MS. Liposomes incorporating this conjugate (S-Peps-PEG5K) were formulated to evaluate whether MMP-2-mediated peptide degradation triggers detachment of long-chain PEG moieties, thereby enhancing internalization by 4T1 breast cancer cells. Additionally, the effects of tumor microenvironmental pH (~6.5) and MMP-2 concentration on drug release dynamics were investigated. Results: Molecular docking revealed robust GPLGVRG-MMP-2 interactions, yielding a binding energy of −7.1 kcal/mol. The peptide formed hydrogen bonds with MMP-2 residues Tyr A:23 and Arg A:53 (bond lengths: 2.4–2.5 Å) and engaged in hydrophobic contacts, confirming MMP-2 as the primary recognition site. Formulations containing 5 mol% Chol-PEG2K-GPLGVRG-PEG5K combined with 0.15 µg/mL MMP-2 (S-Peps-PEG5K +MMP) exhibited superior internalization efficiency and significantly reduced clonogenic survival compared to controls. Notably, acidic pH (~6.5) induced MMP-2-mediated cleavage of the GPLGVRG peptide, accelerating S-Peps-PEG5K dissociation and facilitating drug release. Conclusions: MMP-2-responsive, cleavable PEGylated liposomes markedly improve PTX accumulation and controlled release at tumor sites by dynamically modulating their stealth properties, offering a promising strategy to enhance chemotherapy efficacy in breast cancer. Full article
Show Figures

Graphical abstract

17 pages, 1966 KiB  
Article
Development of INER-PP-F11N as the Peptide-Radionuclide Conjugate Drug Against CCK2 Receptor-Overexpressing Tumors
by Ming-Cheng Chang, Chun-Tang Chen, Ping-Fang Chiang, I-Chung Tang, Cheng-Liang Peng, Yuh-Feng Wang, Yi-Jou Tai and Ying-Cheng Chiang
Int. J. Mol. Sci. 2025, 26(14), 6565; https://doi.org/10.3390/ijms26146565 - 8 Jul 2025
Viewed by 426
Abstract
This work aimed to evaluate two albumin affinity structure-containing peptide-radionuclide conjugate drugs, INER-PP-F11N-1 and INER-PP-F11N-2, for the diagnosis/treatment of cholecystokinin receptor subtype 2 (CCK2R)-overexpressing cancers. We developed In-111- and Lu-177-labeled INER-PP-F11N radiopharmaceuticals and compared them with the current PP-F11N to investigate metabolic stability, [...] Read more.
This work aimed to evaluate two albumin affinity structure-containing peptide-radionuclide conjugate drugs, INER-PP-F11N-1 and INER-PP-F11N-2, for the diagnosis/treatment of cholecystokinin receptor subtype 2 (CCK2R)-overexpressing cancers. We developed In-111- and Lu-177-labeled INER-PP-F11N radiopharmaceuticals and compared them with the current PP-F11N to investigate metabolic stability, biodistribution, SPECT/CT imaging, and therapeutic responses in CCK2R-expressing tumor xenograft mice. The metabolic stability of [111In]In/[177Lu]Lu-INER-PP-F11N remained above 90% for up to 144 h after labeling, indicating that the compound is highly stable under in vitro conditions. INER-PP-F11N showed 27% and 11% higher cellular uptake and internalization than PP-F11N, respectively. In vivo SPECT/CT imaging confirmed that INER-PP-F11N could accumulate at the tumor site of mice 24 h after receiving the two radiopharmaceutical agents. Biodistribution analysis revealed a significantly greater tumor uptake and reduced accumulation of INER-PP-F11N in the kidneys compared with PP-F11N. Furthermore, INER-PP-F11N significantly inhibited the growth of the CCK2R-overexpressing tumors in mice. The INER-PP-F11N radiopharmaceutical was superior as a theragnostic agent compared with the current PP-F11N. Our study suggests that INER-PP-F11N may be an innovative radiopharmaceutical agent for CCK2R-overexpressing tumors. Full article
Show Figures

Graphical abstract

19 pages, 2643 KiB  
Article
Applying Unbiased, Functional Criteria Allows Selection of Novel Cyclic Peptides for Effective Targeted Drug Delivery to Malignant Prostate Cancer Cells
by Anna Cohen, Maysoon Kashkoosh, Vipin Sharma, Akash Panja, Sagi A. Shpitzer, Shay Golan, Andrii Bazylevich, Gary Gellerman, Galia Luboshits and Michael A. Firer
Pharmaceutics 2025, 17(7), 866; https://doi.org/10.3390/pharmaceutics17070866 - 1 Jul 2025
Viewed by 1726
Abstract
Background: Metastatic prostate cancer (mPrC), with a median survival of under 2 years, represents an important unmet medical need which may benefit from the development of more effective targeted drug delivery systems. Several cell surface receptors have been identified as candidates for targeted [...] Read more.
Background: Metastatic prostate cancer (mPrC), with a median survival of under 2 years, represents an important unmet medical need which may benefit from the development of more effective targeted drug delivery systems. Several cell surface receptors have been identified as candidates for targeted drug delivery to mPrC cells; however, these receptors were selected for their overabundance on PrC cells rather than for their suitability for targeted delivery and uptake of cytotoxic drug payloads. Methods: We describe a novel, unbiased strategy to isolate peptides that fulfill functional criteria required for effective intracellular drug delivery and the specific cytotoxicity of PrC cells without prior knowledge of the targeted receptor. Phage clones displaying 7-mer cyclic peptides were negatively selected in vivo and then positively biopanned through a series of parent and drug-resistant mPrC cells. Peptides from the internalized clones were then subjected to a panel of biochemical and functional tests that led to the selection of several peptide candidates. Results: The selected peptides do not bind PSMA. Peptide-drug conjugates (PDCs) incorporating one of the peptides selectively killed wild-type and drug-resistant PrC cell lines and patient PrC cells but not normal prostate tissue cells in vitro. The PDC also halted the growth of PC3 tumors in a xenograft model. Conclusions: Our study demonstrates that adding unbiased, functional criteria into drug carrier selection protocols can lead to the discovery of novel peptides with appropriate properties required for effective targeted drug delivery into target cancer cells. Full article
(This article belongs to the Section Drug Delivery and Controlled Release)
Show Figures

Figure 1

1 pages, 129 KiB  
Correction
Correction: Chavda et al. Peptide-Drug Conjugates: A New Hope for Cancer Management. Molecules 2022, 27, 7232
by Vivek P. Chavda, Hetvi K. Solanki, Majid Davidson, Vasso Apostolopoulos and Joanna Bojarska
Molecules 2025, 30(12), 2579; https://doi.org/10.3390/molecules30122579 - 13 Jun 2025
Viewed by 272
Abstract
In the original publication cited above [...] Full article
66 pages, 2196 KiB  
Review
Oleocanthal as a Multifunctional Anti-Cancer Agent: Mechanistic Insights, Advanced Delivery Strategies, and Synergies for Precision Oncology
by Shirin Jannati, Adiba Patel, Rajashree Patnaik and Yajnavalka Banerjee
Int. J. Mol. Sci. 2025, 26(12), 5521; https://doi.org/10.3390/ijms26125521 - 9 Jun 2025
Cited by 3 | Viewed by 1190
Abstract
Oleocanthal (OC), a secoiridoid phenolic compound exclusive to extra virgin olive oil (EVOO), has emerged as a promising nutraceutical with multifaceted anti-cancer properties. Despite its well-characterized anti-inflammatory and antioxidant effects, the mechanistic breadth and translational potential of OC in oncology remain underexplored and [...] Read more.
Oleocanthal (OC), a secoiridoid phenolic compound exclusive to extra virgin olive oil (EVOO), has emerged as a promising nutraceutical with multifaceted anti-cancer properties. Despite its well-characterized anti-inflammatory and antioxidant effects, the mechanistic breadth and translational potential of OC in oncology remain underexplored and fragmented across the literature. This comprehensive review synthesizes and critically analyzes recent advances in the molecular, pharmacological, and translational landscape of OC’s anti-cancer activities, providing an integrative framework to bridge preclinical evidence with future clinical application. We delineate the pleiotropic mechanisms by which OC modulates cancer hallmarks, including lysosomal membrane permeabilization (LMP)-mediated apoptosis, the inhibition of key oncogenic signaling pathways (c-MET/STAT3, PAR-2/TNF-α, COX-2/mPGES-1), the suppression of epithelial-to-mesenchymal transition (EMT), angiogenesis, and metabolic reprogramming. Furthermore, this review uniquely highlights the emerging role of OC in modulating drug resistance mechanisms by downregulating efflux transporters and sensitizing tumors to chemotherapy, targeted therapies, and immunotherapies. We also examine OC’s bidirectional interaction with gut microbiota, underscoring its systemic immunometabolic effects. A major unmet need addressed by this review is the lack of consolidated knowledge regarding OC’s pharmacokinetic limitations and drug–drug interaction potential in the context of polypharmacy in oncology. We provide an in-depth analysis of OC’s poor bioavailability, extensive first-pass metabolism, and pharmacogenomic interactions, and systematically compile preclinical evidence on advanced delivery platforms—including nanocarriers, microneedle systems, and peptide–drug conjugates—designed to overcome these barriers. By critically evaluating the mechanistic, pharmacological, and translational dimensions of OC, this review advances the field beyond isolated mechanistic studies and offers a strategic blueprint for its integration into precision oncology. It also identifies key research gaps and outlines the future directions necessary to transition OC from a nutraceutical of dietary interest to a viable adjunctive therapeutic agent in cancer treatment. Full article
(This article belongs to the Special Issue Bioactive Compounds in Cancers)
Show Figures

Figure 1

14 pages, 1793 KiB  
Article
A Metal–Organic Hybrid Composed of Dual Quenching Cofactors as a Nanoquencher for the Fluorescent Determination of Protease Caspase-3
by Fengli Gao, Lin Liu, Cancan He, Yong Chang and Weiqiang Wang
Biosensors 2025, 15(6), 354; https://doi.org/10.3390/bios15060354 - 4 Jun 2025
Viewed by 549
Abstract
Nanoquenchers with a single quenching cofactor exhibit limited fluorescence quenching efficiency. In this work, a metal–organic hybrid with dual quenching cofactors (Cu2+ and pyrroloquinoline quinone or PQQ) was prepared by metal-coordinated assembly and used as a nanoquencher for a protease assay with [...] Read more.
Nanoquenchers with a single quenching cofactor exhibit limited fluorescence quenching efficiency. In this work, a metal–organic hybrid with dual quenching cofactors (Cu2+ and pyrroloquinoline quinone or PQQ) was prepared by metal-coordinated assembly and used as a nanoquencher for a protease assay with enhanced quenching efficiency. The peptide substrate with an oligohistidine (His6) tag was labeled with a fluorophore. Caspase-3 was determined as a protease example. The substrate was attached onto the surface of the Cu-PQQ nanoquencher by a metal coordination interaction between the unsaturated Cu2+ on the nanoparticle surface and the His6 tag in the peptide. The cleavage of the peptide substrate by enzymatic hydrolysis led to the release of a fluorophore-conjugated segment from the nanoquencher surface, thus turning on the fluorescence. The nanoprobe was used to determine caspase-3 with a linear range of 0.01–5 ng/mL and a detection limit of 7 pg/mL. Furthermore, the method was used to evaluate inhibition efficiency and monitor drug-induced cell apoptosis. In contrast to other means of peptide immobilization, such as physical adsorption and covalent coupling, the strategy based on the metal coordination interaction is simple and powerful, thereby achieving assays of caspase-3 activity in lysates with a satisfactory result. The work should be valuable for the design of nanoquenchers with multiple quenching cofactors and the development of novel biosensors. Full article
Show Figures

Figure 1

22 pages, 3036 KiB  
Article
Synthesis and Characterization of Transferrin Receptor-Targeted Peptide Combination SN-38 and Rucaparib Conjugate for the Treatment of Glioblastoma
by Perpetue Bataille Backer and Simeon Kolawole Adesina
Pharmaceutics 2025, 17(6), 732; https://doi.org/10.3390/pharmaceutics17060732 - 2 Jun 2025
Viewed by 831
Abstract
Background/Objectives: Glioblastoma represents a particularly aggressive and fatal type of brain tumor. Peptide-drug conjugates, which offer the promise of traversing the blood-brain barrier to selectively accumulate in tumor tissues and precisely target cancer cells, are an active area of research. We present the [...] Read more.
Background/Objectives: Glioblastoma represents a particularly aggressive and fatal type of brain tumor. Peptide-drug conjugates, which offer the promise of traversing the blood-brain barrier to selectively accumulate in tumor tissues and precisely target cancer cells, are an active area of research. We present the synthesis and characterization of the T7 peptide (HAIYPRH) as a targeting ligand for the transferrin receptor, which is highly expressed on both the blood-brain barrier and glioma cells. Methods: Using the T7 peptide, the synthesis, characterization, and biological evaluation of a transferrin receptor-targeted, combination SN-38 and rucaparib peptide drug conjugate (T7-SN-38-rucaparib) are described. Results: The T7 peptide drug conjugate readily cleaved in the presence of exogenous cathepsin B, releasing the active drug payloads. In vitro experiments demonstrated potent cytotoxic effects of the T7 peptide drug conjugate on glioblastoma cells (IC50 = 22.27 nM), with reduced toxicity to non-cancerous HEK 293 cells (IC50 = 115.78 nM), indicating selective toxicity toward cancer cells. Further investigations revealed that blocking transferrin receptors with drug-free T7 peptide significantly reduced the conjugate’s cytotoxicity, an effect that could be reversed by introducing exogenous cathepsin B to the cells. Conclusions: These findings highlight the potential of glioblastoma-targeted delivery of SN-38 and rucaparib based on specific recognition of the transferrin receptor for transport across the blood-brain barrier, offering the prospect of reduced toxicity and selective killing of cancer cells. Additionally, since rucaparib does not cross the blood-brain barrier, this work is significant to facilitate the use of rucaparib for the treatment of brain tumors. Full article
(This article belongs to the Special Issue Combination Therapy Approaches for Cancer Treatment)
Show Figures

Figure 1

15 pages, 1908 KiB  
Article
Surface Functionalized Polyhydroxyalkanoate Nanoparticles via SpyTag–SpyCatcher System for Targeted Breast Cancer Treatment
by Jin Young Heo, Min Kyung Sung, Seonhye Jang, Hansol Kim, Youngdo Jeong, Dong-Jin Jang, Sang-Jae Lee, Seong-Bo Kim and Sung Tae Kim
Pharmaceutics 2025, 17(6), 721; https://doi.org/10.3390/pharmaceutics17060721 - 29 May 2025
Viewed by 649
Abstract
Background/Objectives: Biodegradable polymers have emerged as promising platforms for drug delivery. Produced by microbiomes, polyhydroxyalkanoates (PHAs) offer excellent biocompatibility, biodegradability, and environmental sustainability. In this study, we report the surface functionalization of PHA-based nanoparticles (NPs) using the SpyTag–SpyCatcher system to enhance cellular uptake. [...] Read more.
Background/Objectives: Biodegradable polymers have emerged as promising platforms for drug delivery. Produced by microbiomes, polyhydroxyalkanoates (PHAs) offer excellent biocompatibility, biodegradability, and environmental sustainability. In this study, we report the surface functionalization of PHA-based nanoparticles (NPs) using the SpyTag–SpyCatcher system to enhance cellular uptake. Methods: Initial conjugation with mEGFP-SpyTag enabled visualization, followed by decoration with HER2-specific Affibody-SpyCatcher and/or TAT-SpyCatcher peptides. The prepared NPs retained a diameter of <200 nm and a negatively charged surface. Results: Affibody-functionalized NPs significantly enhanced internalization and cytotoxicity in HER2-overexpressing SK-BR-3 cells, whereas TAT-functionalized NPs promoted uptake across various cell types, independently of HER2 expression. Dual-functionalized NPs exhibited synergistic or attenuated effects based on the HER2 expression levels, highlighting the critical role of ligand composition in targeted delivery. Conclusions: The results of this study demonstrate that the SpyTag–SpyCatcher-mediated surface engineering of PHA NPs offers a modular and robust strategy for active targeting in nanomedicine. Full article
Show Figures

Graphical abstract

28 pages, 831 KiB  
Review
Cyanobacterial Peptides in Anticancer Therapy: A Comprehensive Review of Mechanisms, Clinical Advances, and Biotechnological Innovation
by Heayyean Lee, Khuld Nihan and Yale Ryan Kwon
Mar. Drugs 2025, 23(6), 233; https://doi.org/10.3390/md23060233 - 29 May 2025
Cited by 1 | Viewed by 975
Abstract
Cyanobacteria-derived peptides represent a promising class of anticancer agents due to their structural diversity and potent bioactivity. They exert cytotoxic effects through mechanisms including microtubule disruption, histone deacetylase inhibition, and apoptosis induction. Several peptides—most notably the dolastatin-derived auristatins—have achieved clinical success as cytotoxic [...] Read more.
Cyanobacteria-derived peptides represent a promising class of anticancer agents due to their structural diversity and potent bioactivity. They exert cytotoxic effects through mechanisms including microtubule disruption, histone deacetylase inhibition, and apoptosis induction. Several peptides—most notably the dolastatin-derived auristatins—have achieved clinical success as cytotoxic payloads in antibody–drug conjugates (ADCs). However, challenges such as limited tumor selectivity, systemic toxicity, and production scalability remain barriers to broader application. Recent advances in targeted delivery technologies, combination therapy strategies, synthetic biology, and genome mining offer promising solutions. Emerging data from preclinical and clinical studies highlight their therapeutic potential, particularly in treatment-resistant cancers. In this review, we (i) summarize key cyanobacterial peptides and their molecular mechanisms of action, (ii) examine progress toward clinical translation, and (iii) explore biotechnological approaches enabling sustainable production and structural diversification. We also discuss future directions for enhancing specificity and the therapeutic index to fully exploit the potential of these marine-derived peptides in oncology. Full article
(This article belongs to the Special Issue Marine Natural Products as Anticancer Agents, 4th Edition)
Show Figures

Figure 1

14 pages, 1523 KiB  
Article
Regulatory T Cell Function Is Not Affected by Antisense Peptide-Conjugated Phosphorodiamidate Morpholino Oligomer (PPMO)-Mediated TMPRSS2 Truncation
by Sandra Gunne, Fiona K. Sailer, Lucas Keutmann, Marie Schwerdtner, Hong M. Moulton, Eva Böttcher-Friebertshäuser and Susanne Schiffmann
Drugs Drug Candidates 2025, 4(2), 25; https://doi.org/10.3390/ddc4020025 - 27 May 2025
Viewed by 673
Abstract
Background: TMPRSS2 plays an important role in the viral entry mechanisms of influenza viruses and coronaviruses. Therefore, TMPRSS2 seems to be a suitable antiviral drug target. To exclude possible side effects of TMPRSS2 truncation in an early stage of drug in-vitro testing, this [...] Read more.
Background: TMPRSS2 plays an important role in the viral entry mechanisms of influenza viruses and coronaviruses. Therefore, TMPRSS2 seems to be a suitable antiviral drug target. To exclude possible side effects of TMPRSS2 truncation in an early stage of drug in-vitro testing, this study aims to analyze the impact of TMPRSS2 truncation via antisense peptide-conjugated phosphorodiamidate morpholino oligomer (PPMO) targeting immune cells, using the example of regulatory T cells (Treg). Methods: TMPRSS2 was truncated in human Tregs using a splice-modulating PPMO. Effects on Treg function were analyzed by evaluation of surface marker and transcription factor expression, cytokine secretion, and effector cell suppression capability. Results: PPMO treatment led to a slight concentration-dependent toxicity in Tregs. Tregs with truncated TMPRSS2 behave similarly to untreated and control PPMO-treated cells in the analyzed assays. Conclusions: Treg function is not altered after TMPRSS2 truncation and therefore, no unwanted side effects in regard of Tregs are expected when using TMPRSS2-truncating PPMO as an anti-viral drug. Full article
(This article belongs to the Special Issue Fighting SARS-CoV-2 and Related Viruses)
Show Figures

Figure 1

16 pages, 6714 KiB  
Article
Construction of Graphene Oxide Probes Loaded with Antisense Peptide Nucleic Acid and Doxorubicin for Regulating Telomerase Activity and Inducing Apoptosis of Cancer Cells
by Yanyan Zhu, Qinghong Ji and Min Hong
Biosensors 2025, 15(6), 337; https://doi.org/10.3390/bios15060337 - 26 May 2025
Viewed by 593
Abstract
In this study, we developed a multifunctional graphene oxide (GO)-based nanoprobe co-loaded with antisense peptide nucleic acid (PNA) and the chemotherapeutic agent doxorubicin (DOX). The nanoplatform was strategically functionalized with folic acid ligands to enable folate receptor-mediated tumor targeting. Upon cellular internalization, the [...] Read more.
In this study, we developed a multifunctional graphene oxide (GO)-based nanoprobe co-loaded with antisense peptide nucleic acid (PNA) and the chemotherapeutic agent doxorubicin (DOX). The nanoplatform was strategically functionalized with folic acid ligands to enable folate receptor-mediated tumor targeting. Upon cellular internalization, the antisense PNA component selectively hybridized with human telomerase reverse transcriptase (hTERT) mRNA through sequence-specific recognition, inducing structural detachment from the GO surface. This displacement restored the fluorescence signal of previously quenched fluorophores conjugated to the PNA strand, thereby enabling the real-time in situ detection and quantitative fluorescence imaging of intracellular hTERT mRNA dynamics. The antisense PNA component effectively reduced the hTERT mRNA level and downregulated telomerase activity via an antisense gene regulation pathway, while the pH-responsive release of DOX induced potent cancer cell apoptosis through chemotherapeutic action. This combinatorial therapeutic strategy demonstrated enhanced anticancer efficacy compared to single-modality treatments, achieving a 60% apoptosis induction in HeLa cells through coordinated gene silencing and chemotherapy. This study establishes GO as a promising dual-drug nanocarrier platform for developing next-generation theranostic systems that integrate molecular diagnostics with multimodal cancer therapy. Full article
(This article belongs to the Special Issue Fluorescent Probes for Bioimaging and Biosensors)
Show Figures

Figure 1

Back to TopTop