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

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (728)

Search Parameters:
Keywords = patient-derived xenograft

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
20 pages, 12298 KiB  
Article
Impact of Metastatic Microenvironment on Physiology and Metabolism of Small Cell Neuroendocrine Prostate Cancer Patient-Derived Xenografts
by Shubhangi Agarwal, Deepti Upadhyay, Jinny Sun, Emilie Decavel-Bueff, Robert A. Bok, Romelyn Delos Santos, Said Al Muzhahimi, Rosalie Nolley, Jason Crane, John Kurhanewicz, Donna M. Peehl and Renuka Sriram
Cancers 2025, 17(14), 2385; https://doi.org/10.3390/cancers17142385 - 18 Jul 2025
Viewed by 419
Abstract
Background: Potent androgen receptor pathway inhibitors induce small cell neuroendocrine prostate cancer (SCNC), a highly aggressive subtype of metastatic androgen deprivation-resistant prostate cancer (ARPC) with limited treatment options and poor survival rates. Patients with metastases in the liver have a poor prognosis relative [...] Read more.
Background: Potent androgen receptor pathway inhibitors induce small cell neuroendocrine prostate cancer (SCNC), a highly aggressive subtype of metastatic androgen deprivation-resistant prostate cancer (ARPC) with limited treatment options and poor survival rates. Patients with metastases in the liver have a poor prognosis relative to those with bone metastases alone. The mechanisms that underlie the different behavior of ARPC in bone vs. liver may involve factors intrinsic to the tumor cell, tumor microenvironment, and/or systemic factors, and identifying these factors is critical to improved diagnosis and treatment of SCNC. Metabolic reprogramming is a fundamental strategy of tumor cells to colonize and proliferate in microenvironments distinct from the primary site. Understanding the metabolic plasticity of cancer cells may reveal novel approaches to imaging and treating metastases more effectively. Methods: Using magnetic resonance (MR) imaging and spectroscopy, we interrogated the physiological and metabolic characteristics of SCNC patient-derived xenografts (PDXs) propagated in the bone and liver, and used correlative biochemical, immunohistochemical, and transcriptomic measures to understand the biological underpinnings of the observed imaging metrics. Results: We found that the influence of the microenvironment on physiologic measures using MRI was variable among PDXs. However, the MR measure of glycolytic capacity in the liver using hyperpolarized 13C pyruvic acid recapitulated the enzyme activity (lactate dehydrogenase), cofactor (nicotinamide adenine dinucleotide), and stable isotope measures of fractional enrichment of lactate. While in the bone, the congruence of the glycolytic components was lost and potentially weighted by the interaction of cancer cells with osteoclasts/osteoblasts. Conclusion: While there was little impact of microenvironmental factors on metabolism, the physiological measures (cellularity and perfusion) are highly variable and necessitate the use of combined hyperpolarized 13C MRI and multiparametric (anatomic, diffusion-, and perfusion- weighted) 1H MRI to better characterize pre-treatment tumor characteristics, which will be crucial to evaluate treatment response. Full article
(This article belongs to the Special Issue Magnetic Resonance in Cancer Research)
Show Figures

Figure 1

21 pages, 2524 KiB  
Article
The Relevance of G-Quadruplexes in Gene Promoters and the First Introns Associated with Transcriptional Regulation in Breast Cancer
by Huiling Shu, Ke Xiao, Wenyong Zhu, Rongxin Zhang, Tiantong Tao and Xiao Sun
Int. J. Mol. Sci. 2025, 26(14), 6874; https://doi.org/10.3390/ijms26146874 - 17 Jul 2025
Viewed by 252
Abstract
The role of G-quadruplexes (G4s) in gene regulation has been widely documented, especially in gene promoters. However, the transcriptional mechanisms involving G4s in other regulatory regions remain largely unexplored. In this study, we integrated the G4-DNA data derived from 22 breast cancer patient-derived [...] Read more.
The role of G-quadruplexes (G4s) in gene regulation has been widely documented, especially in gene promoters. However, the transcriptional mechanisms involving G4s in other regulatory regions remain largely unexplored. In this study, we integrated the G4-DNA data derived from 22 breast cancer patient-derived tumor xenograft (PDTX) models and MCF7 cell line as potential breast cancer-associated G4s (BC-G4s). Genome-wide analysis showed that BC-G4s are more prevalent in gene promoters and the first introns. The genes accommodating promoter or intronic BC-G4s show significantly higher transcriptional output than their non-G4 counterparts. The biased distribution of BC-G4s in close proximity to the transcription start site (TSS) is associated with an enrichment of transcription factor (TF) interactions. A significant negative correlation was detected between the G4–TF interactions within the first introns and their cognate promoters. These different interactions are complementary rather than redundant. Furthermore, the differentially expressed genes (DEGs) harboring promoter and first intron BC-G4s are significantly enriched in the cell cycle pathway. Notably, promoter BC-G4s of DEGs could be a central hub for TF–TF co-occurrence. Our analysis also revealed that G4-related single nucleotide variants (SNVs) affect the stability of G4 structures and the transcription of disease-related genes. Collectively, our results shed light on how BC-G4s within promoters and first introns regulate gene expression and reinforce the critical role of G4s and G4-related genes in breast cancer-associated processes. Full article
(This article belongs to the Special Issue Molecular Research of Multi-omics in Cancer)
Show Figures

Figure 1

14 pages, 2956 KiB  
Article
Long-Term Results of Autologous Tooth Bone Grafting in Alveolar Cleft Reconstruction: A Retrospective Cohort Study
by Tamás Würsching, Bence Mészáros, Eleonóra Sólyom, Bálint Molnár, Sándor Bogdán, Zsolt Németh and Krisztián Nagy
Biomedicines 2025, 13(7), 1735; https://doi.org/10.3390/biomedicines13071735 - 16 Jul 2025
Viewed by 319
Abstract
Background/Objectives: During alveolar cleft grafting, the use of autogenous cancellous bone harvested from the iliac crest is still considered the gold standard. Due to the risk of donor-site morbidity and excessive graft resorption, alternative grafting materials (e.g., intraoral bone, xenografts) are being [...] Read more.
Background/Objectives: During alveolar cleft grafting, the use of autogenous cancellous bone harvested from the iliac crest is still considered the gold standard. Due to the risk of donor-site morbidity and excessive graft resorption, alternative grafting materials (e.g., intraoral bone, xenografts) are being tested. The aim of this study was to compare the efficacy of using an autologous tooth-derived graft material and iliac crest cancellous bone in the reconstruction of the alveolar cleft in patients with a unilateral cleft lip and palate. Methods: A total of 21 patients with a unilateral cleft lip and palate, who underwent alveolar bone grafting between 2020 and 2023 were included in the study. In 11 cases, the donor site was the iliac crest; in the rest of the cases, deciduous teeth were harvested, processed, and used as an autologous particulate graft material for alveolar reconstruction. The mean follow-up time was 30.0 months, CBCT scans were taken, and the results were compared based on the ranking system published by Stasiak et al. Results: The Wilcoxon signed-rank test showed that the amount of bone on the cleft side was significantly less than that on the contralateral non-cleft side (ATB: p = 0.002, iliac crest: p = 0.005). The Mann–Whitney U test showed that there were no significant differences in bone quantity on the cleft side between the two groups (U = 47.5, p = 0.617). Conclusions: The use of ATB might be a feasible alternative to autologous bone during alveolar cleft reconstruction. This type of graft shows long-term stability, which is comparable to the bone harvested from the iliac crest. Full article
Show Figures

Figure 1

20 pages, 960 KiB  
Review
Zebrafish as a Model for Translational Immuno-Oncology
by Gabriela Rodrigues Barbosa, Augusto Monteiro de Souza, Priscila Fernandes Silva, Caroline Santarosa Fávero, José Leonardo de Oliveira, Hernandes F. Carvalho, Ana Carolina Luchiari and Leonardo O. Reis
J. Pers. Med. 2025, 15(7), 304; https://doi.org/10.3390/jpm15070304 - 11 Jul 2025
Viewed by 562
Abstract
Despite remarkable progress in cancer immunotherapy, many agents that show efficacy in murine or in vitro models fail to translate clinically. Zebrafish (Danio rerio) have emerged as a powerful complementary model that addresses several limitations of traditional systems. Their optical transparency, [...] Read more.
Despite remarkable progress in cancer immunotherapy, many agents that show efficacy in murine or in vitro models fail to translate clinically. Zebrafish (Danio rerio) have emerged as a powerful complementary model that addresses several limitations of traditional systems. Their optical transparency, genetic tractability, and conserved immune and oncogenic signaling pathways enable high-resolution, real-time imaging of tumor–immune interactions in vivo. Importantly, zebrafish offer a unique opportunity to study the core mechanisms of health and sickness, complementing other models and expanding our understanding of fundamental processes in vivo. This review provides an overview of zebrafish immune system development, highlighting tools for tracking innate and adaptive responses. We discuss their application in modeling immune evasion, checkpoint molecule expression, and tumor microenvironment dynamics using transgenic and xenograft approaches. Platforms for high-throughput drug screening and personalized therapy assessment using patient-derived xenografts (“zAvatars”) are evaluated, alongside limitations, such as temperature sensitivity, immature adaptive immunity in larvae, and interspecies differences in immune responses, tumor complexity, and pharmacokinetics. Emerging frontiers include humanized zebrafish, testing of next-generation immunotherapies, such as CAR T/CAR NK and novel checkpoint inhibitors (LAG-3, TIM-3, and TIGIT). We conclude by outlining the key challenges and future opportunities for integrating zebrafish into the immuno-oncology pipeline to accelerate clinical translation. Full article
(This article belongs to the Special Issue Advances in Animal Models and Precision Medicine for Cancer Research)
Show Figures

Figure 1

15 pages, 1833 KiB  
Article
Comparative Analysis of Gut Microbiota Responses to New SN-38 Derivatives, Irinotecan, and FOLFOX in Mice Bearing Colorectal Cancer Patient-Derived Xenografts
by Katarzyna Unrug-Bielawska, Zuzanna Sandowska-Markiewicz, Magdalena Piątkowska, Paweł Czarnowski, Krzysztof Goryca, Natalia Zeber-Lubecka, Michalina Dąbrowska, Ewelina Kaniuga, Magdalena Cybulska-Lubak, Aneta Bałabas, Małgorzata Statkiewicz, Izabela Rumieńczyk, Kazimiera Pyśniak, Michał Mikula and Jerzy Ostrowski
Cancers 2025, 17(13), 2263; https://doi.org/10.3390/cancers17132263 - 7 Jul 2025
Viewed by 471
Abstract
Background: Symbiotic gut microbiota can enhance cancer therapy efficacy, while treatment-induced dysbiosis may reduce effectiveness or increase toxicity. Our preclinical study compared the anticancer effects and impact on fecal microbiota and metabolites of two water-soluble SN-38 derivatives (BN-MePPR and BN-MOA), with those observed [...] Read more.
Background: Symbiotic gut microbiota can enhance cancer therapy efficacy, while treatment-induced dysbiosis may reduce effectiveness or increase toxicity. Our preclinical study compared the anticancer effects and impact on fecal microbiota and metabolites of two water-soluble SN-38 derivatives (BN-MePPR and BN-MOA), with those observed after treatment with Irinotecan, and the FOLFOX regimen in NOD scid gamma mice bearing patient-derived colon adenocarcinoma xenografts (CRC PDX). Methods: Five individual experiments with Irinotecan and its derivatives and eight individual experiments with FOLFOX were conducted using eight CRC PDX models. Chemotherapeutics were administered intraperitoneally 4–5 times at 5-day intervals. Fecal samples were collected before and after treatment. Microbiota composition was analyzed by 16S rRNA gene (V3–V4 regions) sequencing. Mass spectrometry was used to quantify short-chain fatty acids (SCFAs) and amino acids (AAs). Results: All treatments significantly inhibited tumor growth versus controls. However, no significant changes were observed in gut microbiota α- and β-diversity between treated and untreated groups. Tumor progression in controls was associated with increased abundance of Marvinbryantia, Lactobacillus, Ruminococcus, and [Eubacterium] nodatum group. FOLFOX-treated mice showed increased Marvinbryantia, Bacteroides, and Candidatus Arthromitus, and decreased Akkermansia. No distinct taxa changes were found in the Irinotecan or derivative groups. SCFA levels remained unchanged across groups, while BN-MePPR, BN-MOA, and Irinotecan all increased AA concentrations. Conclusions: Contrary to earlier toxicological data, these findings indicate a relatively limited impact of the tested chemotherapeutics on the gut microbiome and metabolome, emphasizing the importance of research method selection in preclinical studies. Full article
(This article belongs to the Section Cancer Therapy)
Show Figures

Figure 1

54 pages, 3159 KiB  
Review
Biomimetic Tumour Model Systems for Pancreatic Ductal Adenocarcinoma in Relation to Photodynamic Therapy
by Olivia M. Smith, Nicole Lintern, Jiahao Tian, Bárbara M. Mesquita, Sabrina Oliveira, Veronika Vymetalkova, Jai Prakash, Andrew M. Smith, David G. Jayne, Michal Heger and Yazan S. Khaled
Int. J. Mol. Sci. 2025, 26(13), 6388; https://doi.org/10.3390/ijms26136388 - 2 Jul 2025
Viewed by 829
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the most common type of pancreatic cancer and is associated with poor prognosis. Despite years of research and improvements in chemotherapy regimens, the 5-year survival rate of PDAC remains dismal. Therapies for PDAC often face resistance owing in [...] Read more.
Pancreatic ductal adenocarcinoma (PDAC) is the most common type of pancreatic cancer and is associated with poor prognosis. Despite years of research and improvements in chemotherapy regimens, the 5-year survival rate of PDAC remains dismal. Therapies for PDAC often face resistance owing in large part to an extensive desmoplastic stromal matrix. Modelling PDAC ex vivo to investigate novel therapeutics is challenging due to the complex tumour microenvironment and its heterogeneity in native tumours. Development of novel therapies is needed to improve PDAC survival rates, for which disease models that recapitulate the tumour biology are expected to bear utility. This review focuses on the existing preclinical models for human PDAC and discusses advancements in tissue remodelling to guide translational PDAC research. Further emphasis is placed on photodynamic therapy (PDT) due to the ability of this treatment modality to not only directly kill cancer cells by minimally invasive means, but also to perturb the tumour microenvironment and elicit a post-therapeutic anti-tumour immune response. Accordingly, more complex preclinical models that feature multiple biologically relevant PDAC components are needed to develop translatable PDT regimens in a preclinical setting. Full article
(This article belongs to the Special Issue Molecular Advances in Oncologic Photodynamic Therapy)
Show Figures

Figure 1

19 pages, 3401 KiB  
Article
Interleukin 21-Armed EGFR-VHH-CAR-T Cell Therapy for the Treatment of Esophageal Squamous Cell Carcinoma
by Chenglin Zhang, Yanyan Liu, Haoran Guo, Ying Peng, Lei Huang, Shuangshuang Lu and Zhimin Wang
Biomedicines 2025, 13(7), 1598; https://doi.org/10.3390/biomedicines13071598 - 30 Jun 2025
Viewed by 458
Abstract
Background/Objectives: Esophageal squamous cell carcinoma (ESCC) is a common form of esophageal cancer with a poor prognosis and limited treatment options. Epidermal growth factor receptor (EGFR), an overexpressed oncogenic gene in all ESCC patients, is an attractive target for developing therapies against ESCC. [...] Read more.
Background/Objectives: Esophageal squamous cell carcinoma (ESCC) is a common form of esophageal cancer with a poor prognosis and limited treatment options. Epidermal growth factor receptor (EGFR), an overexpressed oncogenic gene in all ESCC patients, is an attractive target for developing therapies against ESCC. There is an extremely urgent need to develop immunotherapy tools targeting EGFR for the treatment of ESCC. Methods: In this study, we developed human Interleukin-21 (hIL-21)-armed, chimeric-antigen-receptor-modified T (CAR-T) cells targeting EGFR as a new therapeutic approach. The CAR contains a variable domain of the llama heavy chain of heavy-chain antibodies (VHHs), also known as nanobodies (Nbs), as a promising substitute for the commonly used single-chain variable fragment (ScFv) for CAR-T development. Results: We show that nanobody-derived, EGFR-targeting CAR-T cells specifically kill EGFR-positive esophageal cancer cells in vitro and in animal models. Human IL-21 expression in CAR-T cells further improved their expansion and antitumor ability and were observed to secrete more interferon-gamma (IFN-γ), tumor necrosis factor alpha (TNF-α), and Interleukin-2 (IL-2) when co-cultured with ESCC cell lines in vitro. More CD8+ CAR-T cells and CD3+CD8+CD45RO+CD62L+ central memory T cells were detected in CAR-T cells expressing hIL-21 cells. Notably, hIL-21-expressing CAR-T cells showed superior antitumor activity in vivo in a KYSE-150 xenograft mouse model. Conclusions: Our results show that hIL-21-armed, nanobody-derived, EGFR-specific CAR-T cell therapy is a highly promising option for treating ESCC patients. Full article
(This article belongs to the Section Cell Biology and Pathology)
Show Figures

Figure 1

15 pages, 4150 KiB  
Article
PRMT5 Identified as a Viable Target for Combination Therapy in Preclinical Models of Pancreatic Cancer
by Xiaolong Wei, William J. Kane, Sara J. Adair, Sarbajeet Nagdas, Denis Liu and Todd W. Bauer
Biomolecules 2025, 15(7), 948; https://doi.org/10.3390/biom15070948 - 30 Jun 2025
Viewed by 458
Abstract
Pancreatic cancer is the third leading cause of cancer-related death in the US. First-line chemotherapy regimens for pancreatic ductal adenocarcinoma (PDAC) include FOLFIRINOX or gemcitabine (Gem) with or without paclitaxel (Ptx); however, 5-year survival with these regimens remains poor. Previous work has demonstrated [...] Read more.
Pancreatic cancer is the third leading cause of cancer-related death in the US. First-line chemotherapy regimens for pancreatic ductal adenocarcinoma (PDAC) include FOLFIRINOX or gemcitabine (Gem) with or without paclitaxel (Ptx); however, 5-year survival with these regimens remains poor. Previous work has demonstrated protein arginine methyltransferase 5 (PRMT5) to be a promising therapeutic target in combination with Gem for the treatment of PDAC; however, these findings have yet to be confirmed in relevant preclinical models of PDAC. To test the possibility of PRMT5 as a viable therapeutic target, clinically relevant orthotopic and metastatic patient-derived xenograft (PDX) mouse models of PDAC growth were utilized to evaluate the effect of PRMT5 knockout (KO) or pharmacologic inhibition on treatment with Gem alone or Gem with Ptx. Primary endpoints included tumor volume, tumor weight, or metastatic tumor burden as appropriate. The results showed that Gem-treated PRMT5 KO tumors exhibited decreased growth and were smaller in size compared to Gem-treated wild-type (WT) tumors. Similarly, the Gem-treated PRMT5 KO metastatic burden was lower than the Gem-treated WT metastatic burden. The addition of a PRMT5 pharmacologic inhibitor to Gem and Ptx therapy resulted in a lower final tumor weight and fewer metastatic tumors. The depletion of PRMT5 results in increased DNA damage in response to Gem and Ptx treatment. Thus, PRMT5 genetic depletion or inhibition in combination with Gem-based therapy improved the response in primary and metastatic PDAC in clinically relevant mouse models, suggesting that PRMT5 is a viable therapeutic target for combination therapy in PDAC. Full article
Show Figures

Figure 1

110 pages, 4617 KiB  
Review
Exploring Experimental Models of Colorectal Cancer: A Critical Appraisal from 2D Cell Systems to Organoids, Humanized Mouse Avatars, Organ-on-Chip, CRISPR Engineering, and AI-Driven Platforms—Challenges and Opportunities for Translational Precision Oncology
by Ahad Al-Kabani, Bintul Huda, Jewel Haddad, Maryam Yousuf, Farida Bhurka, Faika Ajaz, Rajashree Patnaik, Shirin Jannati and Yajnavalka Banerjee
Cancers 2025, 17(13), 2163; https://doi.org/10.3390/cancers17132163 - 26 Jun 2025
Viewed by 2417
Abstract
Background/Objectives: Colorectal cancer (CRC) remains a major global health burden, marked by complex tumor–microenvironment interactions, genetic heterogeneity, and varied treatment responses. Effective preclinical models are essential for dissecting CRC biology and guiding personalized therapeutic strategies. This review aims to critically evaluate current experimental [...] Read more.
Background/Objectives: Colorectal cancer (CRC) remains a major global health burden, marked by complex tumor–microenvironment interactions, genetic heterogeneity, and varied treatment responses. Effective preclinical models are essential for dissecting CRC biology and guiding personalized therapeutic strategies. This review aims to critically evaluate current experimental CRC models, assessing their translational relevance, limitations, and potential for integration into precision oncology. Methods: A systematic literature search was conducted across PubMed, Scopus, and Web of Science, focusing on studies employing defined in vitro, in vivo, and emerging integrative CRC models. Studies were included based on experimental rigor and relevance to therapeutic or mechanistic investigation. Models were compared based on molecular fidelity, tumorigenic capacity, immune interactions, and predictive utility. Results: CRC models were classified into in vitro (2D cell lines, spheroids, patient-derived organoids), in vivo (murine, zebrafish, porcine, canine), and integrative platforms (tumor-on-chip systems, humanized mice, AI-augmented simulations). Traditional models offer accessibility and mechanistic insight, while advanced systems better mimic human tumor complexity, immune landscapes, and treatment response. Tumor-on-chip and AI-driven models show promise in simulating dynamic tumor behavior and predicting clinical outcomes. Cross-platform integration enhances translational validity and enables iterative model refinement. Conclusions: Strategic deployment of complementary CRC models is critical for advancing translational research. This review provides a roadmap for aligning model capabilities with specific research goals, advocating for integrated, patient-relevant systems to improve therapeutic development. Enhancing model fidelity and interoperability is key to accelerating the bench-to-bedside translation in colorectal cancer care. Full article
(This article belongs to the Special Issue Recent Advances in Basic and Clinical Colorectal Cancer Research)
Show Figures

Figure 1

17 pages, 2746 KiB  
Article
Histological Analysis of Sticky Tooth and Sticky Bone
by Robert Dłucik, Marcel Firlej, Katarzyna Bogus, Daniel Dłucik and Bogusława Orzechowska-Wylęgała
J. Funct. Biomater. 2025, 16(7), 233; https://doi.org/10.3390/jfb16070233 - 25 Jun 2025
Viewed by 1391
Abstract
Objective: This study aimed to compare the efficacy of Sticky Tooth (ST) derived from ground teeth and Sticky Bone (SB) based on equine bone and human allograft in maxillary bone defect regeneration through histological examination. Materials and Methods: Forty patients underwent maxillary alveolar [...] Read more.
Objective: This study aimed to compare the efficacy of Sticky Tooth (ST) derived from ground teeth and Sticky Bone (SB) based on equine bone and human allograft in maxillary bone defect regeneration through histological examination. Materials and Methods: Forty patients underwent maxillary alveolar ridge regeneration using four different biomaterials: Sticky Tooth processed with the BonMaker device (n = 10), Sticky Tooth prepared with the Smart Dentin Grinder (n = 10) Sticky Bone derived from an equine xenograft (n = 10), and Sticky Bone derived from human allografts (n = 10). CBCT imaging was performed preoperatively, post-regeneration, and during follow-up. Histological and quantitative statistical evaluation was conducted on biopsy samples obtained four months post-regeneration at the time of implant placement. Results: Successful alveolar ridge regeneration was achieved in all 40 patients. Histological analyses confirmed good integration between the biomaterials and bone tissue without signs of inflammation. Conclusion: Histological comparisons demonstrated that both ST and SB are effective biomaterials for bone regeneration. However, ST exhibited a faster bone healing process compared to xenograft and allograft SB. Full article
(This article belongs to the Section Bone Biomaterials)
Show Figures

Figure 1

16 pages, 20299 KiB  
Article
Biodistribution of a Mucin 4-Selective Monoclonal Antibody: Defining a Potential Therapeutic Agent Against Pancreatic Cancer
by Achyut Dahal, Jerome Schlomer, Laura Bassel, Serguei Kozlov and Joseph J. Barchi
Int. J. Mol. Sci. 2025, 26(13), 6042; https://doi.org/10.3390/ijms26136042 - 24 Jun 2025
Viewed by 441
Abstract
We have previously reported on a novel monoclonal antibody (mAb) we designated F5, which was raised against a glycopeptide derived from the tandem repeat (TR) region of Mucin-4 (MUC4), a heavily O-glycosylated protein that is overexpressed in many pancreatic cancer cells. This mAb [...] Read more.
We have previously reported on a novel monoclonal antibody (mAb) we designated F5, which was raised against a glycopeptide derived from the tandem repeat (TR) region of Mucin-4 (MUC4), a heavily O-glycosylated protein that is overexpressed in many pancreatic cancer cells. This mAb was highly specific for the MUC4 glycopeptide antigen in glycan microarrays, ELISA and SPR assays, selectively stained tissue derived from advanced-stage tumors, and bound MUC4+ tumor cells in flow cytometry assays. The mAb was also unique in that it did not cross-react with other commercial anti-MUC4 mAbs that were raised in a similar but non-glycosylated TR sequence. Here we describe the selective conjugation of a novel near-infrared dye to this mAb and in vivo biodistribution of this labeled mAb to various MUC4-expressing tumors in mice. The labeled mAb were selectively distributed to both cell-derived xenograft (CDX) flank tumors and patient-derived xenograft (PDX) tumors that expressed MUC4 compared to those that were MUC4-negative. Organ distribution analysis showed high uptake in MUC4+ relative to MUC4 tumors. These results suggest that mAb F5 may be used to develop MUC4-targeted, passive antibody-based immunotherapies against Pancreatic Ductal Adenocarcinomas (PDACs) which are notorious for being refractory to many chemo- and radiotherapies Full article
(This article belongs to the Special Issue The Role of Glycans in Immune Regulation)
Show Figures

Graphical abstract

8 pages, 854 KiB  
Communication
Onvansertib-Based Second-Line Therapies in Combination with Gemcitabine and Carboplatin in Patient-Derived Platinum-Resistant Ovarian Carcinomas
by Federica Guffanti, Ilaria Mengoli, Francesca Ricci, Ludovica Perotti, Elena Capellini, Laura Sala, Simone Canesi, Chu-Chiao Wu, Robert Fruscio, Maya Ridinger, Giovanna Damia and Michela Chiappa
Int. J. Mol. Sci. 2025, 26(12), 5708; https://doi.org/10.3390/ijms26125708 - 14 Jun 2025
Viewed by 565
Abstract
Platinum resistance represents an urgent medical need in the management of ovarian cancer. The activity of the combinations of onvansertib, an inhibitor of polo-like kinase 1, with gemcitabine or carboplatin was tested using patient-derived xenografts of high-grade serous ovarian carcinoma resistant to cisplatin [...] Read more.
Platinum resistance represents an urgent medical need in the management of ovarian cancer. The activity of the combinations of onvansertib, an inhibitor of polo-like kinase 1, with gemcitabine or carboplatin was tested using patient-derived xenografts of high-grade serous ovarian carcinoma resistant to cisplatin (DDP). Two PDX models were selected from our xenobank: one with acquired resistance to DDP (#266R) and the other (#315) with intrinsic DDP resistance. Tumor-bearing mice were randomized to receive vehicle, single onvansertib, gemcitabine and carboplatin, and their combinations. Onvansertib/gemcitabine and onvansertib/carboplatin combinations were well tolerated. In the #266R model, single drug treatments were completely inactive, while the combinations of onvansertib/gemcitabine and onvansertib/carboplatin resulted in a significant increase in survival compared to controls and single drugs (p < 0.001 versus control, onvansertib, gemcitabine and carboplatin). Similar efficacy was observed in the s.c. #315 PDX model; indeed, onvansertib and carboplatin monotherapies were inactive, gemcitabine monotherapy was marginally active, while both combinations were highly active. The molecular mechanism underlying the efficacy of the combinations suggests a higher induction of DNA damage which seems plausible considering that, in both cases, gemcitabine and carboplatin, respectively, interfere with DNA metabolism and induce alkylation damage. The results suggest that the combinations of onvansertib/gemcitabine and onvansertib/carboplatin are safe and were shown to be of therapeutic value in the platinum-resistant setting of ovarian carcinoma, strongly supporting their clinical translatability. Full article
(This article belongs to the Special Issue Resistance to Therapy in Ovarian Cancers)
Show Figures

Graphical abstract

22 pages, 30677 KiB  
Article
Mitochondrial Translation Inhibition Uncovers a Critical Metabolic–Epigenetic Interface in Renal Cell Carcinoma
by Kazumi Eckenstein, Beyza Cengiz, Matthew E. K. Chang, Jessie May Cartier, Mark R. Flory and George V. Thomas
Metabolites 2025, 15(6), 393; https://doi.org/10.3390/metabo15060393 - 12 Jun 2025
Viewed by 620
Abstract
Background/Objectives: Renal cell carcinoma (RCC) exhibits distinctive metabolic vulnerabilities that may be therapeutically targeted. This study investigates how tigecycline, an FDA-approved antibiotic that inhibits mitochondrial translation, affects RCC cells and explores potential combinatorial approaches to enhance its efficacy. Methods: We employed comprehensive metabolomic [...] Read more.
Background/Objectives: Renal cell carcinoma (RCC) exhibits distinctive metabolic vulnerabilities that may be therapeutically targeted. This study investigates how tigecycline, an FDA-approved antibiotic that inhibits mitochondrial translation, affects RCC cells and explores potential combinatorial approaches to enhance its efficacy. Methods: We employed comprehensive metabolomic profiling, subcellular proteomics, and functional assays to characterize the effects of tigecycline on RCC cell lines, patient-derived organoids, and xenograft models. The synergistic potential of tigecycline with the histone deacetylase inhibitor entinostat was evaluated using combination index analysis. Results: Tigecycline selectively inhibited mitochondrial translation in RCC cells, reducing mitochondrially-encoded proteins while sparing nuclear-encoded components, profoundly disrupting mitochondrial bioenergetics and reducing tumor growth in xenograft models. Subcellular proteomic analyses revealed that tigecycline treatment triggered a significant accumulation of multiple histone variants concurrent with cell cycle arrest. Based on this discovery, combined treatment with tigecycline and entinostat demonstrated remarkable synergism across RCC cell lines and patient-derived. Conclusions: Our findings identify a promising therapeutic opportunity by targeting the crosstalk between mitochondrial function and epigenetic homeostasis in RCC, with the potential for rapid clinical translation given the established pharmacological profiles of both agents. Full article
Show Figures

Figure 1

26 pages, 6882 KiB  
Article
The Identification of a Key Regulator of Mitochondrial Metabolism, the LRPPRC Protein, as a Novel Therapeutic Target in SDHA-Overexpressing Ovarian Tumors
by Anna Szulta, Lin Wang, Ameera Hasan, Michael Kinter, Atul Pranay, Jie Zhu, Kenneth M. Humphries, Brooke Loveland, Timothy M. Griffin and Magdalena Bieniasz
Cancers 2025, 17(12), 1942; https://doi.org/10.3390/cancers17121942 - 11 Jun 2025
Viewed by 641
Abstract
Background: Ovarian cancer is the deadliest of all gynecologic malignancies due to limited therapeutic options. Our data show that the tumor-specific metabolism of ovarian cancer could be effectively targetable, which highlights a path for new anti-cancer therapies. Methods and Results: Our work shows [...] Read more.
Background: Ovarian cancer is the deadliest of all gynecologic malignancies due to limited therapeutic options. Our data show that the tumor-specific metabolism of ovarian cancer could be effectively targetable, which highlights a path for new anti-cancer therapies. Methods and Results: Our work shows that the upregulation of mitochondrial enzyme SDHA is particularly prevalent in ovarian carcinoma. SDHA overexpression significantly induced orthotopic ovarian tumor growth, reducing mouse survival. We showed that SDHA-overexpressing tumors depend on glutaminolysis and increased activity of the tricarboxylic acid (TCA) cycle coupled with mitochondrial oxidative phosphorylation (OXPHOS), which are essential for high-energy metabolism and cell survival. We identified a distinctive vulnerability of SDHA-overexpressing tumors to agents targeting regulators of the OXPHOS pathway, particularly the LRPPRC protein. LRPPRC is a key regulator of mitochondrial energy metabolism, promoting OXPHOS and ATP generation. However, when overexpressed, the LRPPRC acts as a tumor oncogene. Our analysis of SDHA and LRPPRC gene and protein expression patterns in precursor lesions and established ovarian cancer demonstrated that the upregulation of SDHA is accompanied by LRPPRC overexpression, notably in advanced tumors. Our novel findings highlight for the first time a potential functional interaction between SDHA and LRPPRC in the development and progression of ovarian malignancy. Importantly, our in vivo data showed that pharmacological inhibition of LRPPRC results in a lasting therapeutic benefit and can be an effective therapy in SDHA- and LRPPRC-overexpressing ovarian tumors. Conclusions: Overall, our study underlines an understudied role of concomitant overexpression of SDHA and LRPPRC in ovarian cancer pathogenesis, highlighting new paths for therapeutic development. Full article
(This article belongs to the Section Molecular Cancer Biology)
Show Figures

Figure 1

15 pages, 802 KiB  
Article
Mutational Patterns in Colorectal Cancer: Do PDX Models Retain the Heterogeneity of the Original Tumor?
by Maria El Hage, Zhaoran Su and Michael Linnebacher
Int. J. Mol. Sci. 2025, 26(11), 5111; https://doi.org/10.3390/ijms26115111 - 26 May 2025
Viewed by 538
Abstract
Colorectal cancer (CRC) remains a leading cause of cancer-related mortality worldwide, highlighting the need for a deeper understanding of the genetic mechanisms driving its development and progression. Identifying genetic mutations that affect key molecular pathways is crucial for advancing CRC diagnosis, prognosis, and [...] Read more.
Colorectal cancer (CRC) remains a leading cause of cancer-related mortality worldwide, highlighting the need for a deeper understanding of the genetic mechanisms driving its development and progression. Identifying genetic mutations that affect key molecular pathways is crucial for advancing CRC diagnosis, prognosis, and treatment. Patient-derived xenograft (PDX) models are essential tools in precision medicine and preclinical research, aiding in the development of personalized therapeutic strategies. In this study, a comparative analysis was conducted on the most frequently mutated genes—APC, TP53, KRAS, BRAF, NRAS, and ERBB2—using data from publicly available databases (n = 7894) and models from University Medicine Rostock (n = 139). The aim of this study was to evaluate the accuracy of these models in reflecting the mutational landscape observed in patient-derived samples, with a focus on both individual mutations and co-occurring mutational patterns. Our comparative analysis demonstrated that while the ranking of individual mutations remained consistent, their overall frequencies were slightly lower in the PDX models. Interestingly, we observed a notably higher prevalence of BRAF mutations in the PDX cohort. When examining co-occurring mutations, TP53 and APC mutations—both individually and in combination with other alterations—were the most frequent in both datasets. While the PDX models showed a greater prevalence of single mutations and a slightly higher proportion of tumors without detectable mutations compared to the public dataset, these findings present valuable insights into CRC’s mutational landscape. The discrepancies highlight important considerations, such as selective engraftment bias favoring more aggressive tumors, differences in sample size between the two cohorts, and potential bottleneck effects during PDX engraftment. Understanding these factors can help refine the use of PDX models in CRC research, enhancing their potential for more accurate and relevant applications in precision oncology. Full article
(This article belongs to the Section Molecular Oncology)
Show Figures

Figure 1

Back to TopTop