1
|
Li J, Li Y, Fu L, Chen H, Du F, Wang Z, Zhang Y, Huang Y, Miao J, Xiao Y. Targeting ncRNAs to overcome metabolic reprogramming‑mediated drug resistance in cancer (Review). Int J Oncol 2025; 66:35. [PMID: 40116120 PMCID: PMC12002672 DOI: 10.3892/ijo.2025.5741] [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: 01/22/2025] [Accepted: 03/07/2025] [Indexed: 03/23/2025] Open
Abstract
The emergence of resistance to antitumor drugs in cancer cells presents a notable obstacle in cancer therapy. Metabolic reprogramming is characterized by enhanced glycolysis, disrupted lipid metabolism, glutamine dependence and mitochondrial dysfunction. In addition to promoting tumor growth and metastasis, metabolic reprogramming mediates drug resistance through diverse molecular mechanisms, offering novel opportunities for therapeutic intervention. Non‑coding RNAs (ncRNAs), a diverse class of RNA molecules that lack protein‑coding function, represent a notable fraction of the human genome. Due to their distinct expression profiles and multifaceted roles in various cancers, ncRNAs have relevance in cancer pathophysiology. ncRNAs orchestrate metabolic abnormalities associated with drug resistance in cancer cells. The present review provides a comprehensive analysis of the mechanisms by which metabolic reprogramming drives drug resistance, with an emphasis on the regulatory roles of ncRNAs in glycolysis, lipid metabolism, mitochondrial dysfunction and glutamine metabolism. Furthermore, the present review aimed to discuss the potential of ncRNAs as biomarkers for predicting chemotherapy responses, as well as emerging strategies to target ncRNAs that modulate metabolism, particularly in the context of combination therapy with anti‑cancer drugs.
Collapse
Affiliation(s)
- Junxin Li
- Department of Pharmacy, Zigong Fourth People's Hospital, Zigong, Sichuan 643000, P.R. China
| | - Yanyu Li
- Department of Pharmacy, Zigong Fourth People's Hospital, Zigong, Sichuan 643000, P.R. China
| | - Lin Fu
- Department of Pharmacy, Zigong Fourth People's Hospital, Zigong, Sichuan 643000, P.R. China
| | - Huiling Chen
- Department of Pharmacy, Zigong Fourth People's Hospital, Zigong, Sichuan 643000, P.R. China
| | - Fei Du
- Department of Pharmacy, The Fourth Affiliated Hospital of Southwest Medical University, Meishan, Sichuan 64200, P.R. China
| | - Zhongshu Wang
- Department of Pharmacy, Zigong Fourth People's Hospital, Zigong, Sichuan 643000, P.R. China
| | - Yan Zhang
- Department of Pharmacy, Zigong Fourth People's Hospital, Zigong, Sichuan 643000, P.R. China
| | - Yu Huang
- Department of Pharmacy, Zigong Fourth People's Hospital, Zigong, Sichuan 643000, P.R. China
| | - Jidong Miao
- Department of Oncology, Zigong Fourth People's Hospital, Zigong, Sichuan 643000, P.R. China
| | - Yi Xiao
- Department of Pharmacy, Zigong Fourth People's Hospital, Zigong, Sichuan 643000, P.R. China
| |
Collapse
|
2
|
Chen P, Ren L, Guo Y, Sun Y. Boosting antitumor immunity in breast cancers: Potential of adjuvants, drugs, and nanocarriers. Int Rev Immunol 2025; 44:141-164. [PMID: 39611269 DOI: 10.1080/08830185.2024.2432499] [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: 05/13/2024] [Revised: 10/05/2024] [Accepted: 11/17/2024] [Indexed: 11/30/2024]
Abstract
Despite advancements in breast cancer treatment, therapeutic resistance, and tumor recurrence continue to pose formidable challenges. Therefore, a deep knowledge of the intricate interplay between the tumor and the immune system is necessary. In the pursuit of combating breast cancer, the awakening of antitumor immunity has been proposed as a compelling avenue. Tumor stroma in breast cancers contains multiple stromal and immune cells that impact the resistance to therapy and also the expansion of malignant cells. Activating or repressing these stromal and immune cells, as well as their secretions can be proposed for exhausting resistance mechanisms and repressing tumor growth. NK cells and T lymphocytes are the prominent components of breast tumor immunity that can be triggered by adjuvants for eradicating malignant cells. However, stromal cells like endothelial and fibroblast cells, as well as some immune suppressive cells, consisting of premature myeloid cells, and some subsets of macrophages and CD4+ T lymphocytes, can dampen antitumor immunity in favor of breast tumor growth and therapy resistance. This review article aims to research the prospect of harnessing the power of drugs, adjuvants, and nanoparticles in awakening the immune reactions against breast malignant cells. By investigating the immunomodulatory properties of pharmacological agents and the synergistic effects of adjuvants, this review seeks to uncover the mechanisms through which antitumor immunity can be triggered. Moreover, the current review delineates the challenges and opportunities in the translational journey from bench to bedside.
Collapse
Affiliation(s)
- Ping Chen
- Pharmacy Department, Zibo Central Hospital, Zibo, Shandong, China
| | - Lei Ren
- Pharmacy Department, Zibo Central Hospital, Zibo, Shandong, China
| | - Youwei Guo
- Pharmacy Department, Zibo Central Hospital, Zibo, Shandong, China
| | - Yan Sun
- Pharmacy Department, Zibo Central Hospital, Zibo, Shandong, China
| |
Collapse
|
3
|
Aden D, Zaheer S, Sureka N, Trisal M, Chaurasia JK, Zaheer S. Exploring immune checkpoint inhibitors: Focus on PD-1/PD-L1 axis and beyond. Pathol Res Pract 2025; 269:155864. [PMID: 40068282 DOI: 10.1016/j.prp.2025.155864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 01/20/2025] [Accepted: 02/25/2025] [Indexed: 04/19/2025]
Abstract
Immunotherapy emerges as a promising approach, marked by recent substantial progress in elucidating how the host immune response impacts tumor development and its sensitivity to various treatments. Immune checkpoint inhibitors have revolutionized cancer therapy by unleashing the power of the immune system to recognize and eradicate tumor cells. Among these, inhibitors targeting the programmed cell death protein 1 (PD-1) and its ligand (PD-L1) have garnered significant attention due to their remarkable clinical efficacy across various malignancies. This review delves into the mechanisms of action, clinical applications, and emerging therapeutic strategies surrounding PD-1/PD-L1 blockade. We explore the intricate interactions between PD-1/PD-L1 and other immune checkpoints, shedding light on combinatorial approaches to enhance treatment outcomes and overcome resistance mechanisms. Furthermore, we discuss the expanding landscape of immune checkpoint inhibitors beyond PD-1/PD-L1, including novel targets such as CTLA-4, LAG-3, TIM-3, and TIGIT. Through a comprehensive analysis of preclinical and clinical studies, we highlight the promise and challenges of immune checkpoint blockade in cancer immunotherapy, paving the way for future advancements in the field.
Collapse
Affiliation(s)
- Durre Aden
- Department of Pathology, Hamdard Institute of Medical science and research, Jamia Hamdard, New Delhi, India.
| | - Samreen Zaheer
- Department of Radiotherapy, Jawaharlal Nehru Medical College, AMU, Aligarh, India.
| | - Niti Sureka
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India.
| | - Monal Trisal
- Department of Pathology, Hamdard Institute of Medical science and research, Jamia Hamdard, New Delhi, India.
| | | | - Sufian Zaheer
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India.
| |
Collapse
|
4
|
Zhao B, Shi G, Shi J, Li Z, Xiao Y, Qiu Y, He L, Xie F, Yu D, Cao H, Du H, Zhang J, Zhou Y, Jiang C, Li W, Li M, Wang Z. Research progress on the mechanism and treatment of cachexia based on tumor microenvironment. Nutrition 2025; 133:112697. [PMID: 39999652 DOI: 10.1016/j.nut.2025.112697] [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: 12/07/2024] [Revised: 01/26/2025] [Accepted: 01/28/2025] [Indexed: 02/27/2025]
Abstract
Cachexia is a prevalent multifactorial syndrome characterized by a substantial decrease in food intake, which results from processes such as proteolysis, lipolysis, inflammatory activation, and autophagy, ultimately leading to weight loss. In cancer patients, this condition is referred to as cancer-related cachexia (CRC) and affects over 50% of this population. A comprehensive understanding of the intricate interactions between tumors and the host organism is essential for the development of effective treatments for tumor cachexia. This review aims to elucidate the role of the tumor microenvironment (TME) in the pathogenesis of tumor-associated cachexia and to summarize the current evidence supporting treatment modalities that target the TME.
Collapse
Affiliation(s)
- Bochen Zhao
- School of Basic Medical Sciences, The Fourth Military Medical University, Xi'an, China
| | - Gege Shi
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi'an, China
| | - Jiaxin Shi
- School of Basic Medical Sciences, The Fourth Military Medical University, Xi'an, China
| | - Zhaozhao Li
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi'an, China
| | - Yang Xiao
- Department of Experiment Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Yueyuan Qiu
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi'an, China
| | - Lei He
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi'an, China
| | - Fei Xie
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi'an, China
| | - Duo Yu
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi'an, China
| | - Haiyan Cao
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi'an, China
| | - Haichen Du
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi'an, China
| | - Jieyu Zhang
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi'an, China
| | - Yang Zhou
- School of Basic Medical Sciences, The Fourth Military Medical University, Xi'an, China
| | - Caiyi Jiang
- School of Basic Medical Sciences, The Fourth Military Medical University, Xi'an, China
| | - Weina Li
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi'an, China
| | - Meng Li
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi'an, China
| | - Zhaowei Wang
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi'an, China.
| |
Collapse
|
5
|
Anwer MS, Abdel-Rasol MA, El-Sayed WM. Emerging therapeutic strategies in glioblastsoma: drug repurposing, mechanisms of resistance, precision medicine, and technological innovations. Clin Exp Med 2025; 25:117. [PMID: 40223032 PMCID: PMC11994545 DOI: 10.1007/s10238-025-01631-0] [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: 02/16/2025] [Accepted: 03/11/2025] [Indexed: 04/15/2025]
Abstract
Glioblastoma (GBM) is an aggressive Grade IV brain tumor with a poor prognosis. It results from genetic mutations, epigenetic changes, and factors within the tumor microenvironment (TME). Traditional treatments like surgery, radiotherapy, and chemotherapy provide limited survival benefits due to the tumor's heterogeneity and resistance mechanisms. This review examines novel approaches for treating GBM, focusing on repurposing existing medications such as antipsychotics, antidepressants, and statins for their potential anti-GBM effects. Advances in molecular profiling, including next-generation sequencing, artificial intelligence (AI), and nanotechnology-based drug delivery, are transforming GBM diagnosis and treatment. The TME, particularly GBM stem cells and immune evasion, plays a key role in therapeutic resistance. Integrating multi-omics data and applying precision medicine show promise, especially in combination therapies and immunotherapies, to enhance clinical outcomes. Addressing challenges such as drug resistance, targeting GBM stem cells, and crossing the blood-brain barrier is essential for improving treatment efficacy. While current treatments offer limited benefits, emerging strategies such as immunotherapies, precision medicine, and drug repurposing show significant potential. Technologies like liquid biopsies, AI-powered diagnostics, and nanotechnology could help overcome obstacles like the blood-brain barrier and GBM stem cells. Ongoing research into combination therapies, targeted drug delivery, and personalized treatments is crucial. Collaborative efforts and robust clinical trials are necessary to translate these innovations into effective therapies, offering hope for improved survival and quality of life for GBM patients.
Collapse
Affiliation(s)
- Mohamed S Anwer
- Department of Zoology, Faculty of Science, Ain Shams University, Abbassia, Cairo, 11566, Egypt
| | - Mohammed A Abdel-Rasol
- Department of Zoology, Faculty of Science, Ain Shams University, Abbassia, Cairo, 11566, Egypt.
| | - Wael M El-Sayed
- Department of Zoology, Faculty of Science, Ain Shams University, Abbassia, Cairo, 11566, Egypt.
| |
Collapse
|
6
|
Lv Y, Pu L, Ran B, Xiang B. Targeting tumor angiogenesis and metabolism with photodynamic nanomedicine. Front Cell Dev Biol 2025; 13:1558393. [PMID: 40235732 PMCID: PMC11996804 DOI: 10.3389/fcell.2025.1558393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Accepted: 03/24/2025] [Indexed: 04/17/2025] Open
Abstract
Photodynamic therapy (PDT) holds considerable promise as a tumor treatment modality, characterized by its targeted action, compatibility with other therapeutic approaches, and non - invasive features. PDT can achieve remarkable spatiotemporal precision in tumor ablation through the generation of reactive oxygen species (ROS). Nevertheless, despite its potential in tumor treatment, PDT encounters multiple challenges in practical applications. PDT is highly oxygen - dependent, and thus the effectiveness of PDT can be markedly influenced by tumor hypoxia. The co-existence of abnormal vasculature and metabolic deregulation gives rise to a hypoxic microenvironment, which not only sustains tumor survival but also undermines the therapeutic efficacy of PDT. Consequently, targeting tumor angiogenesis and metabolism is essential for revitalizing PDT. This review emphasizes the mechanisms and strategies for revitalizing PDT in tumor treatment, predominantly concentrating on interfering with tumor angiogenesis and reprogramming tumor cell metabolism. Lastly, the outlining future perspectives and current limitations of PDT are also summarized. This could provide new insights and methodologies for overcoming the challenges associated with PDT in tumor treatment, ultimately advancing the field of PDT.
Collapse
Affiliation(s)
- Yong Lv
- Department of Pediatric Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Lihui Pu
- Department of Critical Care, West China Hospital, Sichuan University, Chengdu, China
| | - Bei Ran
- School of Biomedical Engineering, Sichuan University, Chengdu, China
| | - Bo Xiang
- Department of Pediatric Surgery, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
7
|
Kunz LV, Schaefer R, Kacem H, Ollivier J, Togno M, Chappuis F, Weber D, Lomax A, Limoli CL, Psoroulas S, Vozenin MC. Plasmid DNA Strand Breaks Are Dose Rate Independent at Clinically Relevant Proton Doses and Under Biological Conditions. Radiat Res 2025; 203:214-222. [PMID: 40010373 DOI: 10.1667/rade-24-00118.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 11/13/2024] [Indexed: 02/28/2025]
Abstract
We investigated the effect of proton FLASH radiation on plasmid DNA. Purified supercoiled pBR322 plasmids were irradiated with clinical doses (≤10 Gy) of protons at ultra-high and conventional dose rates using the Paul Scherrer Institute (PSI) isochronous cyclotron. The proton beam in this clinical facility has been validated to produce the FLASH effect in preclinical models. Plasmid samples were irradiated under various oxygen tensions, scavenger levels, pH conditions and Fe (II) concentrations as these biochemical parameters vary across tissues and tumors. Over the range of doses used, plasmid DNA strand breaks were found to be dose rate independent at all conditions investigated. Irradiation within the Bragg peak and spread-out Bragg peak increased clustered strand breaks, except in the presence of scavengers. With this model system, we demonstrate conclusively that plasmid DNA strand breakage is dose rate independent at doses below 10 Gy and does not constitute a high throughput assay endpoint predictive of the biological effect of FLASH.
Collapse
Affiliation(s)
- Louis V Kunz
- Sector of Radiobiology Applied to Radiotherapy, Radiation Oncology Department, Geneva University Hospital, Geneva, Switzerland
- LiRR - Laboratory of Innovation in Radiobiology Applied to Radiotherapy, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Laboratory of Radiation Oncology, Radiation Oncology Service, Department of Oncology, Centre Hospitalier Universitaire Vaudois, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Robert Schaefer
- Center for Proton Therapy, Paul Scherrer Institute, 5323 Villigen PSI, Switzerland
| | - Houda Kacem
- Sector of Radiobiology Applied to Radiotherapy, Radiation Oncology Department, Geneva University Hospital, Geneva, Switzerland
- LiRR - Laboratory of Innovation in Radiobiology Applied to Radiotherapy, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Laboratory of Radiation Oncology, Radiation Oncology Service, Department of Oncology, Centre Hospitalier Universitaire Vaudois, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Jonathan Ollivier
- Sector of Radiobiology Applied to Radiotherapy, Radiation Oncology Department, Geneva University Hospital, Geneva, Switzerland
- LiRR - Laboratory of Innovation in Radiobiology Applied to Radiotherapy, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Laboratory of Radiation Oncology, Radiation Oncology Service, Department of Oncology, Centre Hospitalier Universitaire Vaudois, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Michele Togno
- Center for Proton Therapy, Paul Scherrer Institute, 5323 Villigen PSI, Switzerland
| | - Flore Chappuis
- Institute of Radiation Physics (IRA), Centre Hospitalier Universitaire Vaudois, Lausanne University Hospital, Lausanne, Switzerland
| | - Damien Weber
- Center for Proton Therapy, Paul Scherrer Institute, 5323 Villigen PSI, Switzerland
| | - Anthony Lomax
- Center for Proton Therapy, Paul Scherrer Institute, 5323 Villigen PSI, Switzerland
| | - Charles L Limoli
- Department of Radiation Oncology, University of California, Irvine, Irvine, California
| | - Serena Psoroulas
- Center for Proton Therapy, Paul Scherrer Institute, 5323 Villigen PSI, Switzerland
| | - Marie-Catherine Vozenin
- Sector of Radiobiology Applied to Radiotherapy, Radiation Oncology Department, Geneva University Hospital, Geneva, Switzerland
- LiRR - Laboratory of Innovation in Radiobiology Applied to Radiotherapy, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Laboratory of Radiation Oncology, Radiation Oncology Service, Department of Oncology, Centre Hospitalier Universitaire Vaudois, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
8
|
Richiardone E, Giolito MV, Al Roumi R, Ambroise J, Boidot R, Drotleff B, Ghesquière B, Lupo B, Trusolino L, Bardelli A, Arena S, Feron O, Corbet C. Acidosis overrides molecular heterogeneity to shape therapeutically targetable metabolic phenotypes in colon cancers. Cancer Lett 2025; 613:217512. [PMID: 39900217 DOI: 10.1016/j.canlet.2025.217512] [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: 11/04/2024] [Revised: 01/20/2025] [Accepted: 01/28/2025] [Indexed: 02/05/2025]
Abstract
Colorectal cancer (CRC) represents a prototypical example of a cancer type for which inter- and intra-tumor heterogeneities remain major challenges for the clinical management of patients. Besides genotype-mediated phenotypic alterations, tumor microenvironment (TME) conditions are increasingly recognized to promote intrinsic diversity and phenotypic plasticity and sustain disease progression. In particular, acidosis is a common hallmark of solid tumors, including CRC, and it is known to induce aggressive cancer cell phenotypes. In this study, we report that long-term adaptation to acidic pH conditions is associated with common metabolic alterations, including a glycolysis-to-respiration switch and a higher reliance on the activity of phosphoglycerate dehydrogenase (PHGDH), in CRC cells initially displaying molecularly heterogeneous backgrounds. Pharmacological inhibition of PHGDH activity or mitochondrial respiration induces greater growth-inhibitory effects in acidosis-exposed CRC cells in 2D and 3D culture conditions, and in patient-derived CRC organoids. These data pave the way for drugs targeting the acidic tumor compartment as a "one-size-fits-all" therapeutic approach to delay CRC progression.
Collapse
Affiliation(s)
- Elena Richiardone
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, Avenue Hippocrate 57, B1.57.04, B-1200, Brussels, Belgium
| | - Maria Virginia Giolito
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, Avenue Hippocrate 57, B1.57.04, B-1200, Brussels, Belgium
| | - Rim Al Roumi
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, Avenue Hippocrate 57, B1.57.04, B-1200, Brussels, Belgium
| | - Jérôme Ambroise
- Centre des Technologies Moléculaires Appliquées (CTMA), Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, Avenue Hippocrate 54, B-1200, Brussels, Belgium
| | - Romain Boidot
- Unit of Molecular Biology, Department of Biology and Pathology of Tumors, Georges-François Leclerc Cancer Center-UNICANCER, 21079, Dijon, France
| | | | - Bart Ghesquière
- Laboratory of Applied Mass Spectrometry, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium; Metabolomics Core Facility Leuven, Center for Cancer Biology, VIB, Leuven, Belgium
| | - Barbara Lupo
- Department of Oncology, University of Torino, Candiolo (TO), Italy; Candiolo Cancer Institute - FPO IRCCS, Candiolo (TO), Italy
| | - Livio Trusolino
- Department of Oncology, University of Torino, Candiolo (TO), Italy; Candiolo Cancer Institute - FPO IRCCS, Candiolo (TO), Italy
| | - Alberto Bardelli
- Department of Oncology, Molecular Biotechnology Center, University of Torino, Torino, Italy; IFOM ETS, The AIRC Institute of Molecular Oncology, 20139, Milan, Italy
| | - Sabrina Arena
- Department of Oncology, University of Torino, Candiolo (TO), Italy; Candiolo Cancer Institute - FPO IRCCS, Candiolo (TO), Italy
| | - Olivier Feron
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, Avenue Hippocrate 57, B1.57.04, B-1200, Brussels, Belgium; WEL Research Institute, Avenue Pasteur 6, B-1300, Wavre, Belgium
| | - Cyril Corbet
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, Avenue Hippocrate 57, B1.57.04, B-1200, Brussels, Belgium; WEL Research Institute, Avenue Pasteur 6, B-1300, Wavre, Belgium.
| |
Collapse
|
9
|
Malla R, Bhamidipati P, Samudrala AS, Nuthalapati Y, Padmaraju V, Malhotra A, Rolig AS, Malhotra SV. Exosome-Mediated Cellular Communication in the Tumor Microenvironment Imparts Drug Resistance in Breast Cancer. Cancers (Basel) 2025; 17:1167. [PMID: 40227747 PMCID: PMC11987792 DOI: 10.3390/cancers17071167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2025] [Revised: 03/22/2025] [Accepted: 03/29/2025] [Indexed: 04/15/2025] Open
Abstract
Globally, breast cancer (BC) is the leading cause of cancer-related death for women. BC is characterized by heterogeneity, aggressive behavior, and high metastatic potential. Chemotherapy, administered as monotherapy or adjuvant therapy, remains a cornerstone of treatment; however, acquired drug resistance is a significant clinical challenge. Deciphering mechanisms of drug resistance will be central to developing more efficient treatment options and improving patient outcomes. The current review examines the multifaceted nature of exosomes in conferring drug resistance in BC through complex communication networks within the tumor microenvironment. We further explore recent advances in understanding how exosomes contribute to resistance against established chemotherapeutic agents such as tamoxifen, paclitaxel, doxorubicin, platinum-based drugs, trastuzumab, and newer immunotherapies, such as immune checkpoint inhibitors. Moreover, we discuss existing systematic approaches to investigating the exosome-drug resistance relationship in BC. Finally, we explore promising therapeutic approaches to overcome exosome-dependent drug resistance in BC, highlighting potential avenues for improved treatment efficacy. Investigating the distinct functions and cargo of exosomes offers potential for developing innovative approaches to overcoming treatment resistance.
Collapse
Affiliation(s)
- RamaRao Malla
- Cancer Biology Group, Cancer Biology Laboratory, Department of Life Sciences, GITAM School of Science, GITAM (Deemed to be University), Visakhapatnam 530045, Andhra Pradesh, India
| | - Priyamvada Bhamidipati
- Cancer Biology Group, Cancer Biology Laboratory, Department of Life Sciences, GITAM School of Science, GITAM (Deemed to be University), Visakhapatnam 530045, Andhra Pradesh, India
| | - Anuveda Sree Samudrala
- Cancer Biology Group, Cancer Biology Laboratory, Department of Life Sciences, GITAM School of Science, GITAM (Deemed to be University), Visakhapatnam 530045, Andhra Pradesh, India
| | - Yerusha Nuthalapati
- Cancer Biology Group, Cancer Biology Laboratory, Department of Life Sciences, GITAM School of Science, GITAM (Deemed to be University), Visakhapatnam 530045, Andhra Pradesh, India
| | - Vasudevaraju Padmaraju
- Cancer Biology Group, Cancer Biology Laboratory, Department of Life Sciences, GITAM School of Science, GITAM (Deemed to be University), Visakhapatnam 530045, Andhra Pradesh, India
| | - Aditya Malhotra
- Center for Experimental Therapeutics, Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97201, USA
| | - Annah S. Rolig
- Center for Experimental Therapeutics, Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97201, USA
| | - Sanjay V. Malhotra
- Center for Experimental Therapeutics, Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97201, USA
| |
Collapse
|
10
|
Gu XY, Yang JL, Lai R, Zhou ZJ, Tang D, Hu L, Zhao LJ. Impact of lactate on immune cell function in the tumor microenvironment: mechanisms and therapeutic perspectives. Front Immunol 2025; 16:1563303. [PMID: 40207222 PMCID: PMC11979165 DOI: 10.3389/fimmu.2025.1563303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2025] [Accepted: 03/10/2025] [Indexed: 04/11/2025] Open
Abstract
Lactate has emerged as a key regulator in the tumor microenvironment (TME), influencing both tumor progression and immune dynamics. As a byproduct of aerobic glycolysis, lactate satisfies the metabolic needs of proliferating tumor cells while reshaping the TME to facilitate immune evasion. Elevated lactate levels inhibit effector immune cells such as CD8+ T and natural killer cells, while supporting immunosuppressive cells, such as regulatory T cells and myeloid-derived suppressor cells, thus fostering an immunosuppressive environment. Lactate promotes epigenetic reprogramming, stabilizes hypoxia-inducible factor-1α, and activates nuclear factor kappa B, leading to further immunological dysfunction. In this review, we examined the role of lactate in metabolic reprogramming, immune suppression, and treatment resistance. We also discuss promising therapeutic strategies targeting lactate metabolism, including lactate dehydrogenase inhibitors, monocarboxylate transporter inhibitors, and TME neutralization methods, all of which can restore immune function and enhance immunotherapy outcomes. By highlighting recent advances, this review provides a theoretical foundation for integrating lactate-targeted therapies into clinical practice. We also highlight the potential synergy between these therapies and current immunotherapeutic strategies, providing new avenues for addressing TME-related challenges and improving outcomes for patients with cancer.
Collapse
Affiliation(s)
- Xuan-Yu Gu
- Department of General Surgery, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Jia-Li Yang
- Department of General Surgery, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Rui Lai
- Department of Otolaryngology-Head and Neck Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Zheng-Jun Zhou
- Department of General Surgery, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Dan Tang
- Department of General Surgery, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- Hepatobiliary and Pancreatic Surgery, the Second Affiliated Hospital of Zunyi Medical University, Zunyi, China
- Department of Hepatobiliary and Pancreatic Surgery, Suzhou Medical College of Soochow University, Suzhou, China
| | - Long Hu
- Wisdom Lake Academy of Pharmacy, Xi’an Jiaotong-Liverpool University, Suzhou, China
| | - Li-Jin Zhao
- Department of General Surgery, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| |
Collapse
|
11
|
Jiya N, Sanap A, Srivastava S, Kheur S, Sharma A. Microbial Signatures in Oral Submucous Fibrosis and Oral Squamous Cell Carcinoma: Implications of Tobacco and Betel Quid Consumption. J Oral Biosci 2025:100656. [PMID: 40127779 DOI: 10.1016/j.job.2025.100656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 03/18/2025] [Accepted: 03/22/2025] [Indexed: 03/26/2025]
Abstract
OBJECTIVES We compared the oral bacteriome and mycobiome of patients with oral submucosal fibrosis (OSMF) and oral squamous cell carcinoma (OSCC) who consumed tobacco/betel quid with those of healthy individuals. METHODS Targeted amplicon sequencing coupled with bioinformatics analysis was performed to assess the bacterial and fungal communities in the oral cavity. RESULTS Distinct variations in the microbial communities were observed among the healthy, OSMF, and OSCC samples. Firmicutes was dominant at the phylum level: 21.71%, 21.73%, and 24.56% in the healthy control, OSMF, and OSCC samples, respectively. Lactobacillus (9.6%, 9.04%, and 8.9%) and Streptococcus (7.3%, 7.55%, and 6.1%) showed minimal variation among the healthy, OSMF, and OSCC samples, respectively, at the genus level. Rothia (2.73%) and Veillonella (1.52%) were prominent in those with OSMF, and Gemella (2.2%) and Fusobacterium (1.5%) were prominent in the OSCC samples. The oral mycobiome was dominated by Ascomycota and Basidiomycota in all samples, with Ascomycota accounting for 18.95%, 16.05%, and 15.87%, and Basidiomycota for 13.55%, 15.79%, and 15.96% in the OSCC, OSMF, and healthy samples, respectively. PICRUSt2 analysis revealed the key metabolic pathways linked to disease and xenobiotic degradation, related to the harmful compounds from tobacco, highlighting the clinical relevance of tobacco and betel quid associated microbial communities as well as the differences between OSCC and OSMF. CONCLUSIONS Microbial signatures associated with OSCC and OSMF could serve as biomarkers for early cancer detection. These findings highlight how tobacco and betel quid consumption promote cancer and alter the oral microbome.
Collapse
Affiliation(s)
- Namrata Jiya
- National Centre for Microbial Resource, BRIC-National Centre for Cell Science-411007, Pune, India
| | - Avinash Sanap
- Regenerative Medicine Laboratory, Dr. D. Y. Patil Vidyapeeth, Pimpri, Pune, India
| | - Shruti Srivastava
- National Centre for Microbial Resource, BRIC-National Centre for Cell Science-411007, Pune, India
| | - Supriya Kheur
- Department of Oral Pathology and Microbiology, Dr. D. Y. Patil Dental College and Hospital, Dr. D. Y. Patil Vidyapeeth, Pimpri, Pune, India.
| | - Avinash Sharma
- National Centre for Microbial Resource, BRIC-National Centre for Cell Science-411007, Pune, India.
| |
Collapse
|
12
|
Robinson KS, Sennhenn P, Yuan DS, Liu H, Taddei D, Qian Y, Luo W. TMBIM6/BI-1 is an intracellular environmental regulator that induces paraptosis in cancer via ROS and Calcium-activated ERAD II pathways. Oncogene 2025; 44:494-512. [PMID: 39609612 PMCID: PMC11832424 DOI: 10.1038/s41388-024-03222-x] [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: 06/15/2023] [Revised: 10/28/2024] [Accepted: 11/05/2024] [Indexed: 11/30/2024]
Abstract
Transmembrane B cell lymphoma 2-associated X protein inhibitor motif-containing (TMBIM) 6, also known as Bax Inhibitor-1 (BI-1), has been heavily researched for its cytoprotective functions. TMBIM6 functional diversity includes modulating cell survival, stress, metabolism, cytoskeletal dynamics, organelle function, regulating cytosolic acidification, calcium, and reactive oxygen species (ROS). Clinical research shows TMBIM6 plays a key role in many of the world's top diseases/injuries (i.e., Alzheimer's, Parkinson's, diabetes, obesity, brain injury, liver disease, heart disease, aging, etc.), including cancer, where TMBIM6 expression impacts patient survival, chemoresistance, cancer progression, and metastasis. We show TMBIM6 is activated by, and undergoes, different conformational changes that dictate its function following a significant change in the cell's IntraCellular Environment (ICE). TMBIM6 agonism, following ICE change, can help the cell overcome multiple stresses including toxin exposure, viral infection, wound healing, and excitotoxicity. However, in cancer cells TMBIM6 agonism results in rapid paraptotic induction irrespective of the cancer type, sub-type, genotype or phenotype. Furthermore, the level of TMBIM6 expression in cancer did not dictate the level of paraptotic induction; however, it did dictate the rate at which paraptosis occurred. TMBIM6 agonism did not induce paraptosis in cancer via canonical routes involving p38 MAPK, JNK, ERK, UPR, autophagy, proteasomes, or Caspase-9. Instead, TMBIM6 agonism in cancer upregulates cytosolic Ca2+ and ROS, activates lysosome biogenesis, and induces paraptosis via ERAD II mechanisms. In xenograft models, we show TMBIM6 agonism induces rapid cancer cell death with no toxicity, even at high doses of TMBIM6 agonist (>450 mg/kg). In summary, this study shows TMBIM6's functional diversity is only activated by severe ICE change in diseased/injured cells, highlighting its transformative potential as a therapeutic target across various diseases and injuries, including cancer.
Collapse
Affiliation(s)
| | | | | | - Hai Liu
- Viva Biotech, Shanghai, China
| | | | | | - Wei Luo
- MicroQuin, Cambridge, MA, USA
| |
Collapse
|
13
|
Gupta PK, Li LZ, Singh DK, Nova S, Arias-Mendoza F, Orlovskiy S, Chawla S, Nelson DS, Farwell MD, Nath K. MRS and Optical Imaging Studies of Therapeutic Response to Combination Therapy Targeting BRAF/MEK in Murine Melanomas. Acad Radiol 2025:S1076-6332(25)00090-X. [PMID: 39984334 DOI: 10.1016/j.acra.2025.01.035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 01/24/2025] [Accepted: 01/26/2025] [Indexed: 02/23/2025]
Abstract
RATIONALE AND OBJECTIVES Melanoma, an aggressive skin cancer, often harbors BRAFV600E mutations driving tumor progression via the mitogen-activated protein kinase (MAPK) pathway. While targeted therapies like BRAF (dabrafenib) and MEK (trametinib) inhibitors have improved outcomes, resistance linked to metabolic reprogramming remains a challenge. This study investigates metabolic changes induced by dual BRAF/MEK inhibition in a BRAFV600E-mutant murine melanoma model using magnetic resonance spectroscopy (MRS), optical redox imaging (ORI), and biochemical assays. We aim to identify metabolic biomarkers for predicting therapeutic response or resistance. MATERIALS AND METHODS YUMM1.7 murine melanoma cells and tumored mice were treated with dabrafenib and trametinib. ORI assessed mitochondrial redox status by measuring reduced nicotinamide adenine dinucleotide (NADH), oxidized flavoproteins (Fp), and the redox ratio (Fp/(NADH+Fp)) in vitro. Glucose consumption and lactate production were analyzed using a YSI Biochemical Analyzer. In vivo metabolic changes were monitored via ¹H and ³¹P MRS, evaluating lactate, alanine, pH, βNTP/Pi, and total NAD(P)(H), which represents combined oxidized nicotinamide adenine dinucleotide (NAD+), NADH, and reduced nicotinamide adenine dinucleotide phosphate (NADPH). RESULTS Under the combined therapeutic regimen of dabrafenib and trametinib, YUMM1.7 murine melanoma cells exhibited significant inhibition of lactate generation, non-significant reduction of glucose utilization, decreased intracellular levels of NADH and total NAD(P)(H), and more oxidized redox status in vitro, which can be interpreted as inhibition of the Warburg effect and improved OXPHOS efficiency by targeting BRAF/MEK signaling activities. Furthermore, YUMM1.7 mouse tumors demonstrated less tissue acidification and improved bioenergetics (βNTP/Pi), in agreement with the in vitro data. CONCLUSION MRS, ORI, and biochemical assays identified critical metabolic changes, highlighting potential biomarkers and supporting the integration of metabolic inhibitors with MAPK-targeted therapies to improve clinical outcomes.
Collapse
Affiliation(s)
- Pradeep Kumar Gupta
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (P.K.G., L.Z.L., D.K.S., S.N., F.A.M., S.O., S.C., D.S.N., M.D.F., K.N.)
| | - Lin Z Li
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (P.K.G., L.Z.L., D.K.S., S.N., F.A.M., S.O., S.C., D.S.N., M.D.F., K.N.); Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania (L.Z.L., K.N.); Institute of Translational Medicine and Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania (L.Z.L., K.N.)
| | - Dinesh Kumar Singh
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (P.K.G., L.Z.L., D.K.S., S.N., F.A.M., S.O., S.C., D.S.N., M.D.F., K.N.)
| | - Skyler Nova
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (P.K.G., L.Z.L., D.K.S., S.N., F.A.M., S.O., S.C., D.S.N., M.D.F., K.N.)
| | - Fernando Arias-Mendoza
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (P.K.G., L.Z.L., D.K.S., S.N., F.A.M., S.O., S.C., D.S.N., M.D.F., K.N.); Advanced Imaging Research, Inc. Cleveland, Ohio (F.A.M.)
| | - Stepan Orlovskiy
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (P.K.G., L.Z.L., D.K.S., S.N., F.A.M., S.O., S.C., D.S.N., M.D.F., K.N.)
| | - Sanjeev Chawla
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (P.K.G., L.Z.L., D.K.S., S.N., F.A.M., S.O., S.C., D.S.N., M.D.F., K.N.)
| | - David S Nelson
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (P.K.G., L.Z.L., D.K.S., S.N., F.A.M., S.O., S.C., D.S.N., M.D.F., K.N.)
| | - Michael D Farwell
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (P.K.G., L.Z.L., D.K.S., S.N., F.A.M., S.O., S.C., D.S.N., M.D.F., K.N.)
| | - Kavindra Nath
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (P.K.G., L.Z.L., D.K.S., S.N., F.A.M., S.O., S.C., D.S.N., M.D.F., K.N.); Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania (L.Z.L., K.N.); Institute of Translational Medicine and Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania (L.Z.L., K.N.).
| |
Collapse
|
14
|
Sun M, Wang K, Lu F, Yu D, Liu S. Regulatory role and therapeutic prospect of lactate modification in cancer. Front Pharmacol 2025; 16:1508552. [PMID: 40034817 PMCID: PMC11872897 DOI: 10.3389/fphar.2025.1508552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 01/20/2025] [Indexed: 03/05/2025] Open
Abstract
Post-translational modifications (PTMs) of proteins refer to the process of adding chemical groups, sugars, or other molecules to specific residues of target proteins following their biosynthesis by ribosomes. PTMs play a crucial role in processes such as signal transduction, epigenetics, and disease development. Lactylation is a newly discovered PTM that, due to its close association with lactate-the end product of glycolytic metabolism-provides a new perspective on the connection between cellular metabolic reprogramming and epigenetic regulation. Studies have demonstrated that lactylation plays a significant role in tumor progression and is associated with poor clinical prognosis. Abnormal histone lactylation can influence gene expression in both tumor cells and immune cells, thereby regulating tumor progression and immunosuppression. Lactylation of non-histone proteins can also modulate processes such as tumor proliferation and drug resistance. This review summarizes the latest research progress in the field of lactylation, highlighting its roles and mechanisms in tumorigenesis, tumor development, the tumor microenvironment, and immunosuppression. It also explores the potential application value of lactylation in tumor-targeted therapy and combined immunotherapy.
Collapse
Affiliation(s)
- Mengdi Sun
- Graduate School, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Kejing Wang
- Graduate School, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Fang Lu
- Institute of Traditional Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Donghua Yu
- Institute of Traditional Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Shumin Liu
- Institute of Traditional Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin, China
| |
Collapse
|
15
|
Liao F, Li Y, Zhang Z, Yu Q, Liu H. Pulsed electromagnetic fields modulate energy metabolism during wound healing process: an in vitro model study. BMC Complement Med Ther 2025; 25:50. [PMID: 39939866 PMCID: PMC11823022 DOI: 10.1186/s12906-025-04792-3] [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: 02/01/2024] [Accepted: 01/28/2025] [Indexed: 02/14/2025] Open
Abstract
BACKGROUND Pulsed electromagnetic fields (PEMFs) therapy was extensively investigated to treat wound healing, which is a highly metabolically demanding process. However, the effect of PEMFs on energy metabolism in wound healing remains largely unexplored. Therefore, our study aims to demonstrate the role of PEMFs on energy metabolism in wound healing. METHODS Scratch-wound healing assay and cell viability assay were performed for the in vitro study of the effect of PEMFs on cell migration and viability. Seahorse assay was conducted for energy metabolism analysis while holo-tomographic microscopy for fine changes of L929 cells. Mitochondrial membrane potential assay and intracellular reactive oxygen species (ROS) and pH assay were performed for analyzing the changes of mitochondrial function. RESULTS PEMFs with specific parameter (4mT, 80 Hz) promoted cell migration and viability. Glycolysis stress and mitochondria stress test revealed that PEMFs-exposed L929 cells was highly glycolytic for energy generation. Besides, PEMFs enhanced intracellular acidification and maintained low level of intracellular ROS in L929 cells. Compared to control group, much more vesicles were generated and then transported to regions close to the nuclear in L929 cells treated with PEMFs. CONCLUSIONS Our major findings revealed for the first time that PEMFs induce metabolic reprogramming of fibroblast shifting from mitochondrial respiration to glycolysis, accompanied with an increase of vesicular transport, which is closely related to wound healing in vitro.
Collapse
Affiliation(s)
- Feng Liao
- Department of Orthopedics, Sichuan Provincial People's Hospital,School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, PR China
| | - Yamei Li
- Department of Rehabilitation Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, PR China
| | - Zhou Zhang
- Department of Rehabilitation Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, PR China
| | - Qian Yu
- Department of Rehabilitation Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, PR China.
| | - Huifang Liu
- Department of Rehabilitation Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, PR China.
| |
Collapse
|
16
|
Li Y, He C, Shen AN, Wang Y, Xu ZP, Zhang L, Wang R. pH of Microenvironment Directly Modulates the Phenotype and Function of Cancer-Associated Fibroblasts. ACS OMEGA 2025; 10:3937-3943. [PMID: 39926491 PMCID: PMC11799978 DOI: 10.1021/acsomega.4c09716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 12/07/2024] [Accepted: 12/26/2024] [Indexed: 02/11/2025]
Abstract
Cancer-associated fibroblasts within the tumor microenvironment have been studied extensively, including their differential roles in promoting cancer growth and metastasis, promoting an immune suppressive microenvironment, and reshaping the stiffness of the extracellular matrix. Fibroblasts have diverse functions owing to their heterogeneous phenotypes shaped by the microenvironment. Increased acidity is a crucial feature of the tumor microenvironment, contributing to the generation of cancer-associated fibroblasts. Our data show that a low pH drives the formation of cancer-associated fibroblasts in vitro, while increasing pH activates the self-remodeling features of these cells by limiting their proliferation and downregulating the production of extracellular matrix-associated proteins. Our findings show that cancer-associated fibroblasts are a versatile population that can be reprogramed toward a quiescent phenotype with reduced acidity in the tumor microenvironment. pH regulation could be a potential strategy to target fibroblasts for cancer therapy.
Collapse
Affiliation(s)
- Ying Li
- Institute
of Systems and Physical Biology, Shenzhen
Bay Laboratory, Gaoke
Innovation Center, Guangming District, Shenzhen, Guangdong 518132, P. R. China
| | - Chao He
- Institute
of Systems and Physical Biology, Shenzhen
Bay Laboratory, Gaoke
Innovation Center, Guangming District, Shenzhen, Guangdong 518132, P. R. China
| | - Ai Ning Shen
- Institute
of Systems and Physical Biology, Shenzhen
Bay Laboratory, Gaoke
Innovation Center, Guangming District, Shenzhen, Guangdong 518132, P. R. China
| | - Yu Wang
- Institute
of Systems and Physical Biology, Shenzhen
Bay Laboratory, Gaoke
Innovation Center, Guangming District, Shenzhen, Guangdong 518132, P. R. China
| | - Zhi Ping Xu
- Institute
of Systems and Physical Biology, Shenzhen
Bay Laboratory, Gaoke
Innovation Center, Guangming District, Shenzhen, Guangdong 518132, P. R. China
| | - Lingxiao Zhang
- Interdisciplinary
Nanoscience Center (iNANO), Aarhus University, DK-8000 Aarhus
C, Denmark
| | - Ran Wang
- Institute
of Systems and Physical Biology, Shenzhen
Bay Laboratory, Gaoke
Innovation Center, Guangming District, Shenzhen, Guangdong 518132, P. R. China
- Mater
Research Institute, The University of Queensland, St Lucia, Brisbane, Queensland 4102, Australia
| |
Collapse
|
17
|
Peppicelli S, Calorini L, Bianchini F, Papucci L, Magnelli L, Andreucci E. Acidity and hypoxia of tumor microenvironment, a positive interplay in extracellular vesicle release by tumor cells. Cell Oncol (Dordr) 2025; 48:27-41. [PMID: 39023664 PMCID: PMC11850579 DOI: 10.1007/s13402-024-00969-z] [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] [Accepted: 06/28/2024] [Indexed: 07/20/2024] Open
Abstract
The complex and continuously evolving features of the tumor microenvironment, varying between tumor histotypes, are characterized by the presence of host cells and tumor cells embedded in a milieu shaped by hypoxia and low pH, resulting from the frequent imbalance between vascularity and tumor cell proliferation. These microenvironmental metabolic stressors play a crucial role in remodeling host cells and tumor cells, contributing to the stimulation of cancer cell heterogeneity, clonal evolution, and multidrug resistance, ultimately leading to progression and metastasis. The extracellular vesicles (EVs), membrane-enclosed structures released into the extracellular milieu by tumor/host cells, are now recognized as critical drivers in the complex intercellular communication between tumor cells and the local cellular components in a hypoxic/acidic microenvironment. Understanding the intricate molecular mechanisms governing the interactions between tumor and host cells within a hypoxic and acidic microenvironment, triggered by the release of EVs, could pave the way for innovative strategies to disrupt the complex interplay of cancer cells with their microenvironment. This approach may contribute to the development of an efficient and safe therapeutic strategy to combat cancer progression. Therefore, we review the major findings on the release of EVs in a hypoxic/acidic tumor microenvironment to appreciate their role in tumor progression toward metastatic disease.
Collapse
Affiliation(s)
- Silvia Peppicelli
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, 50134, Italy.
| | - Lido Calorini
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, 50134, Italy
| | - Francesca Bianchini
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, 50134, Italy
| | - Laura Papucci
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, 50134, Italy
| | - Lucia Magnelli
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, 50134, Italy
| | - Elena Andreucci
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, 50134, Italy
| |
Collapse
|
18
|
Greenshields-Watson A, Vavourakis O, Spoendlin FC, Cagiada M, Deane CM. Challenges and compromises: Predicting unbound antibody structures with deep learning. Curr Opin Struct Biol 2025; 90:102983. [PMID: 39862761 DOI: 10.1016/j.sbi.2025.102983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 12/31/2024] [Accepted: 01/02/2025] [Indexed: 01/27/2025]
Abstract
Therapeutic antibodies are manufactured, stored and administered in the free state; this makes understanding the unbound form key to designing and improving development pipelines. Prediction of unbound antibodies is challenging, specifically modelling of the CDRH3 loop, where inaccuracies are potentially worse due to a bias in structural data towards antibody-antigen complexes. This class imbalance provides a challenge for deep learning models trained on this data, potentially limiting generalisation to unbound forms. Here we discuss the importance of unbound structures in antibody development pipelines. We explore how the latest generation of structure predictors can provide new insights and assess how conformational heterogeneity may influence binding kinetics. We hypothesise that generative models may address some of these issues. While prediction of antibodies in complex is essential, we should not ignore the need for progress in modelling the unbound form.
Collapse
Affiliation(s)
- Alexander Greenshields-Watson
- Oxford Protein Informatics Group, Department of Statistics, University of Oxford, 24-29 St Giles', Oxford, OX1 3LB, United Kingdom.
| | - Odysseas Vavourakis
- Oxford Protein Informatics Group, Department of Statistics, University of Oxford, 24-29 St Giles', Oxford, OX1 3LB, United Kingdom
| | - Fabian C Spoendlin
- Oxford Protein Informatics Group, Department of Statistics, University of Oxford, 24-29 St Giles', Oxford, OX1 3LB, United Kingdom
| | - Matteo Cagiada
- Oxford Protein Informatics Group, Department of Statistics, University of Oxford, 24-29 St Giles', Oxford, OX1 3LB, United Kingdom; Linderstrøm-Lang Centre for Protein Science, Department of Biology, University of Copenhagen, DK-2200, Copenhagen, Denmark
| | - Charlotte M Deane
- Oxford Protein Informatics Group, Department of Statistics, University of Oxford, 24-29 St Giles', Oxford, OX1 3LB, United Kingdom
| |
Collapse
|
19
|
Wodtke P, Grashei M, Schilling F. Quo Vadis Hyperpolarized 13C MRI? Z Med Phys 2025; 35:8-32. [PMID: 38160135 PMCID: PMC11910262 DOI: 10.1016/j.zemedi.2023.10.004] [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: 08/29/2023] [Revised: 10/16/2023] [Accepted: 10/20/2023] [Indexed: 01/03/2024]
Abstract
Over the last two decades, hyperpolarized 13C MRI has gained significance in both preclinical and clinical studies, hereby relying on technologies like PHIP-SAH (ParaHydrogen-Induced Polarization-Side Arm Hydrogenation), SABRE (Signal Amplification by Reversible Exchange), and dDNP (dissolution Dynamic Nuclear Polarization), with dDNP being applied in humans. A clinical dDNP polarizer has enabled studies across 24 sites, despite challenges like high cost and slow polarization. Parahydrogen-based techniques like SABRE and PHIP offer faster, more cost-efficient alternatives but require molecule-specific optimization. The focus has been on imaging metabolism of hyperpolarized probes, which requires long T1, high polarization and rapid contrast generation. Efforts to establish novel probes, improve acquisition techniques and enhance data analysis methods including artificial intelligence are ongoing. Potential clinical value of hyperpolarized 13C MRI was demonstrated primarily for treatment response assessment in oncology, but also in cardiology, nephrology, hepatology and CNS characterization. In this review on biomedical hyperpolarized 13C MRI, we summarize important and recent advances in polarization techniques, probe development, acquisition and analysis methods as well as clinical trials. Starting from those we try to sketch a trajectory where the field of biomedical hyperpolarized 13C MRI might go.
Collapse
Affiliation(s)
- Pascal Wodtke
- Department of Nuclear Medicine, TUM School of Medicine and Health, Klinikum rechts der Isar of Technical University of Munich, 81675 Munich, Germany; Department of Radiology, University of Cambridge, Cambridge CB2 0QQ, United Kingdom; Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge UK
| | - Martin Grashei
- Department of Nuclear Medicine, TUM School of Medicine and Health, Klinikum rechts der Isar of Technical University of Munich, 81675 Munich, Germany
| | - Franz Schilling
- Department of Nuclear Medicine, TUM School of Medicine and Health, Klinikum rechts der Isar of Technical University of Munich, 81675 Munich, Germany; Munich Institute of Biomedical Engineering, Technical University of Munich, 85748 Garching, Germany; German Cancer Consortium (DKTK), Partner Site Munich and German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, Heidelberg 69120, Germany.
| |
Collapse
|
20
|
Rahmani D, Taheri RA, Moosazadeh Moghaddam M. Targeted delivery of curcumin and CM11 peptide against hepatocellular carcinoma cells based on binding affinity of PreS1-coated chitosan nanoparticles to SB3 protein. Amino Acids 2025; 57:12. [PMID: 39862295 PMCID: PMC11762422 DOI: 10.1007/s00726-024-03438-x] [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: 07/20/2024] [Accepted: 12/18/2024] [Indexed: 01/27/2025]
Abstract
In recent years, the use of cationic peptides as alternative drugs with anticancer activity has received attention. In this study, the targeted release of curcumin (Cur) and CM11 peptide alone and together against hepatocellular carcinoma (HCC) was evaluated using chitosan nanoparticles (CS NPs) coated with Pres1 that target the SB3 antigen of HCC cells (PreS1-Cur-CM11-CS NPs). SB3 protein is the specific antigen of HCC and the PreS1 peptide is a part of the hepatitis B antigen, which can specifically bind to the SB3 protein. Chitosan was used to prepare NPs. To Cur and CM11 loading, drugs were added to the CS solution in appropriate concentrations. Pres1 was coupled to the surface of the NPs using EDC catalyst to target NPs against HepG2 cells. SEM and DLS analysis confirmed that the PreS1-Cur-CM11-CS NPs had a size of about 132 nm, the ideal size for penetrating the cell membrane. The loading of Cur and CM11 was equal to 87% and 65%, respectively, which had a sustained and better release in the acidic environment than in the physiological environment. The MTT assay showed that PreS1-Cur-CM11-CS NPs act in a targeted and specific manner with the highest toxicity on the HepG2 cells compared to the control by a decrease in viability of about 26% after 48 h based on cell apoptosis. The results showed that PreS1-Cur-CM11-CS NPs are capable of targeted and specific drug release against HepG2 cancer cells and have significant potential to fight this cancer.
Collapse
Affiliation(s)
- Danial Rahmani
- Tissue Engineering and Regenerative Medicine Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Ramezan Ali Taheri
- Nanobiotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Mehrdad Moosazadeh Moghaddam
- Tissue Engineering and Regenerative Medicine Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
21
|
Horta M, Soares P, Leite Pereira C, Lima RT. Emerging Approaches in Glioblastoma Treatment: Modulating the Extracellular Matrix Through Nanotechnology. Pharmaceutics 2025; 17:142. [PMID: 40006509 PMCID: PMC11859630 DOI: 10.3390/pharmaceutics17020142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Revised: 01/10/2025] [Accepted: 01/16/2025] [Indexed: 02/27/2025] Open
Abstract
Glioblastoma's (GB) complex tumor microenvironment (TME) promotes its progression and resistance to therapy. A critical component of TME is the extracellular matrix (ECM), which plays a pivotal role in promoting the tumor's invasive behavior and aggressiveness. Nanotechnology holds significant promise for GB treatment, with the potential to address challenges posed by both the blood-brain barrier and the GB ECM. By enabling targeted delivery of therapeutic and diagnostic agents, nanotechnology offers the prospect of improving treatment efficacy and diagnostic accuracy at the tumor site. This review provides a comprehensive exploration of GB, including its epidemiology, classification, and current treatment strategies, alongside the intricacies of its TME. It highlights nanotechnology-based strategies, focusing on nanoparticle formulations such as liposomes, polymeric nanoparticles, and gold nanoparticles, which have shown promise in GB therapy. Furthermore, it explores how different emerging nanotechnology strategies modulate the ECM to overcome the challenges posed by its high density, which restricts drug distribution within GB tumors. By emphasizing the intersection of nanotechnology and GB ECM, this review underscores an innovative approach to advancing GB treatment. It addresses the limitations of current therapies, identifies new research avenues, and emphasizes the potential of nanotechnology to improve patient outcomes.
Collapse
Affiliation(s)
- Miguel Horta
- i3S—Instituto de Investigação e Inovação em Saúde, University of Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; (M.H.); (P.S.)
- IPATIMUP—Instituto de Patologia e Imunologia Molecular, University of Porto, Rua Júlio Amaral de Carvalho 45, 4200-135 Porto, Portugal
- FMUP—Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Paula Soares
- i3S—Instituto de Investigação e Inovação em Saúde, University of Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; (M.H.); (P.S.)
- IPATIMUP—Instituto de Patologia e Imunologia Molecular, University of Porto, Rua Júlio Amaral de Carvalho 45, 4200-135 Porto, Portugal
- FMUP—Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Catarina Leite Pereira
- i3S—Instituto de Investigação e Inovação em Saúde, University of Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; (M.H.); (P.S.)
- INEB—Instituto Nacional de Engenharia Biomédica, University of Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
| | - Raquel T. Lima
- i3S—Instituto de Investigação e Inovação em Saúde, University of Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; (M.H.); (P.S.)
- IPATIMUP—Instituto de Patologia e Imunologia Molecular, University of Porto, Rua Júlio Amaral de Carvalho 45, 4200-135 Porto, Portugal
- FMUP—Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| |
Collapse
|
22
|
Zhang S, Wang H. Targeting the lung tumour stroma: harnessing nanoparticles for effective therapeutic interventions. J Drug Target 2025; 33:60-86. [PMID: 39356091 DOI: 10.1080/1061186x.2024.2410462] [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: 07/29/2024] [Revised: 08/27/2024] [Accepted: 09/24/2024] [Indexed: 10/03/2024]
Abstract
Lung cancer remains an influential global health concern, necessitating the development of innovative therapeutic strategies. The tumour stroma, which is known as tumour microenvironment (TME) has a central impact on tumour expansion and treatment resistance. The stroma of lung tumours consists of numerous cells and molecules that shape an environment for tumour expansion. This environment not only protects tumoral cells against immune system attacks but also enables tumour stroma to attenuate the action of antitumor drugs. This stroma consists of stromal cells like cancer-associated fibroblasts (CAFs), suppressive immune cells, and cytotoxic immune cells. Additionally, the presence of stem cells, endothelial cells and pericytes can facilitate tumour volume expansion. Nanoparticles are hopeful tools for targeted drug delivery because of their extraordinary properties and their capacity to devastate biological obstacles. This review article provides a comprehensive overview of contemporary advancements in targeting the lung tumour stroma using nanoparticles. Various nanoparticle-based approaches, including passive and active targeting, and stimuli-responsive systems, highlighting their potential to improve drug delivery efficiency. Additionally, the role of nanotechnology in modulating the tumour stroma by targeting key components such as immune cells, extracellular matrix (ECM), hypoxia, and suppressive elements in the lung tumour stroma.
Collapse
Affiliation(s)
- Shushu Zhang
- Cancer Center (Oncology) Department, the Second Affiliated Hospital, Soochow University, Suzhou, Jiangsu, China
| | - Hui Wang
- Cancer Center (Oncology) Department, the Second Affiliated Hospital, Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
23
|
Muhammad FA, Altalbawy FMA, Mandaliya V, Saraswat SK, Rekha MM, Aulakh D, Chahar M, Mahdi MS, Jaber MA, Alhadrawi M. Targeting breast tumor extracellular matrix and stroma utilizing nanoparticles. Clin Transl Oncol 2024:10.1007/s12094-024-03793-x. [PMID: 39692807 DOI: 10.1007/s12094-024-03793-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 11/08/2024] [Indexed: 12/19/2024]
Abstract
Breast cancer is a complicated malignancy and is known as the most common cancer in women. Considerable experiments have been devoted to explore the basic impacts of the tumor stroma, particularly the extracellular matrix (ECM) and stromal components, on tumor growth and resistance to treatment. ECM is made up of an intricate system of proteins, glycosaminoglycans, and proteoglycans, and maintains structural support and controls key signaling pathways involved in breast tumors. ECM can block different drugs such as chemotherapy and immunotherapy drugs from entering the tumor stroma. Furthermore, the stromal elements, such as cancer-associated fibroblasts (CAFs), immune cells, and blood vessels, have crucial impacts on tumor development and therapeutic resistance. Recently, promising outcomes have been achieved in using nanotechnology for delivering drugs to tumor stroma and crossing ECM in breast malignancies. Nanoparticles have various benefits for targeting the breast tumor stroma, such as improved permeability and retention, extended circulation time, and the ability to actively target the area. This review covers the latest developments in nanoparticle therapies that focus on breast tumor ECM and stroma. We will explore different approaches using nanoparticles to target the delivery of anticancer drugs like chemotherapy, small molecule drugs, various antitumor products, and other specific synthetic therapeutic agents to the breast tumor stroma. Furthermore, we will investigate the utilization of nanoparticles in altering the stromal elements, such as reprogramming CAFs and immune cells, and also remodeling ECM.
Collapse
Affiliation(s)
| | - Farag M A Altalbawy
- Department of Chemistry, University College of Duba, University of Tabuk, Tabuk, Saudi Arabia.
- National Institute of Laser Enhanced Sciences (NILES), University of Cairo, Giza, 12613, Egypt.
| | - Viralkumar Mandaliya
- Department of Microbiology, Faculty of Science, Marwadi University Research Center, Marwadi University, Rajkot, Gujarat, 360003, India
| | | | - M M Rekha
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to Be University), Bangalore, Karnataka, India
| | - Damanjeet Aulakh
- Centre for Research Impact and Outcome, Chitkara University Institute of Engineering and Technology Chitkara University, Rajpura, Punjab, 140401, India
| | - Mamata Chahar
- Department of Chemistry, NIMS Institute of Engineering and Technology, NIMS University Rajasthan, Jaipur, India
| | | | | | - Merwa Alhadrawi
- Department of Refrigeration and air Conditioning Techniques, College of Technical Engineering, The Islamic University, Najaf, Iraq
- Department of Refrigeration and air Conditioning Techniques, College of Technical Engineering, The Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- Department of Refrigeration and air Conditioning Techniques, College of Technical Engineering, The Islamic University of Babylon, Babylon, Iraq
| |
Collapse
|
24
|
Martinelli S, Fortuna L, Coratti F, Passagnoli F, Amedei A, Cianchi F. Potential Probes for Targeted Intraoperative Fluorescence Imaging in Gastric Cancer. Cancers (Basel) 2024; 16:4141. [PMID: 39766041 PMCID: PMC11675003 DOI: 10.3390/cancers16244141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 11/29/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
Gastric cancer (GC) is a malignant tumor of the gastrointestinal tract associated with high mortality rates and accounting for approximately 1 million new cases diagnosed annually. Surgery, particularly radical gastrectomy, remains the primary treatment; however, there are currently no specific approaches to better distinguish malignant from healthy tissue or to differentiate between metastatic and non-metastatic lymph nodes. As a result, surgeons have to remove all lymph nodes indiscriminately, increasing intraoperative risks for patients and prolonging hospital stay. Near-infrared fluorescence imaging with indocyanine green (ICG) can provide real-time visualization of the surgical field using both conventional laparoscopy and robotic mini-invasive precision surgery platforms. However, its application shows some limits, as ICG is a non-targeted contrast agent. Several studies are now investigating the potential efficacy of fluorescent targeted agents that could selectively bind to the tumor tissue, offering a valuable tool for metastatic mapping during robotic gastrectomy. This review aims to summarize the key fluorescent agents that have been developed to recognize GC markers, as well as those targeting the tumor microenvironment (TME) and metabolic features. These agents hold great potential as valuable tools for enhancing precision surgery in robotic gastrectomy procedures improving the clinical recovery of GC patients.
Collapse
Affiliation(s)
- Serena Martinelli
- Department of Clinical and Experimental Medicine, University of Florence, 50139 Florence, Italy; (L.F.); (F.C.); (F.P.); (A.A.); (F.C.)
| | - Laura Fortuna
- Department of Clinical and Experimental Medicine, University of Florence, 50139 Florence, Italy; (L.F.); (F.C.); (F.P.); (A.A.); (F.C.)
| | - Francesco Coratti
- Department of Clinical and Experimental Medicine, University of Florence, 50139 Florence, Italy; (L.F.); (F.C.); (F.P.); (A.A.); (F.C.)
| | - Federico Passagnoli
- Department of Clinical and Experimental Medicine, University of Florence, 50139 Florence, Italy; (L.F.); (F.C.); (F.P.); (A.A.); (F.C.)
| | - Amedeo Amedei
- Department of Clinical and Experimental Medicine, University of Florence, 50139 Florence, Italy; (L.F.); (F.C.); (F.P.); (A.A.); (F.C.)
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), 50134 Florence, Italy
| | - Fabio Cianchi
- Department of Clinical and Experimental Medicine, University of Florence, 50139 Florence, Italy; (L.F.); (F.C.); (F.P.); (A.A.); (F.C.)
| |
Collapse
|
25
|
Makino Y, Rajapakshe KI, Chellakkan Selvanesan B, Okumura T, Date K, Dutta P, Abou-Elkacem L, Sagara A, Min J, Sans M, Yee N, Siemann MJ, Enriquez J, Smith P, Bhattacharya P, Kim M, Dede M, Hart T, Maitra A, Thege FI. Metabolic reprogramming by mutant GNAS creates an actionable dependency in intraductal papillary mucinous neoplasms of the pancreas. Gut 2024; 74:75-88. [PMID: 39277181 PMCID: PMC12014225 DOI: 10.1136/gutjnl-2024-332412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 09/04/2024] [Indexed: 09/17/2024]
Abstract
BACKGROUND Oncogenic 'hotspot' mutations of KRAS and GNAS are two major driver alterations in intraductal papillary mucinous neoplasms (IPMNs), which are bona fide precursors to pancreatic ductal adenocarcinoma. We previously reported that pancreas-specific Kras G12D and Gnas R201C co-expression in p48Cre; KrasLSL-G12D; Rosa26LSL-rtTA; Tg (TetO-GnasR201C) mice ('Kras;Gnas' mice) caused development of cystic lesions recapitulating IPMNs. OBJECTIVE We aim to unveil the consequences of mutant Gnas R201C expression on phenotype, transcriptomic profile and genomic dependencies. DESIGN We performed multimodal transcriptional profiling (bulk RNA sequencing, single-cell RNA sequencing and spatial transcriptomics) in the 'Kras;Gnas' autochthonous model and tumour-derived cell lines (Kras;Gnas cells), where Gnas R201C expression is inducible. A genome-wide CRISPR/Cas9 screen was conducted to identify potential vulnerabilities in KrasG12D;GnasR201C co-expressing cells. RESULTS Induction of Gnas R201C-and resulting G(s)alpha signalling-leads to the emergence of a gene signature of gastric (pyloric type) metaplasia in pancreatic neoplastic epithelial cells. CRISPR screening identified the synthetic essentiality of glycolysis-related genes Gpi1 and Slc2a1 in Kras G12D;Gnas R201C co-expressing cells. Real-time metabolic analyses in Kras;Gnas cells and autochthonous Kras;Gnas model confirmed enhanced glycolysis on Gnas R201C induction. Induction of Gnas R201C made Kras G12D expressing cells more dependent on glycolysis for their survival. Protein kinase A-dependent phosphorylation of the glycolytic intermediate enzyme 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3 (PFKFB3) was a driver of increased glycolysis on Gnas R201C induction. CONCLUSION Multiple orthogonal approaches demonstrate that Kras G12D and Gnas R201C co-expression results in a gene signature of gastric pyloric metaplasia and glycolytic dependency during IPMN pathogenesis. The observed metabolic reprogramming may provide a potential target for therapeutics and interception of IPMNs.
Collapse
Affiliation(s)
- Yuki Makino
- Translational Molecular Pathology, UTMDACC, Houston, Texas, USA
- Sheikh Ahmed Center for Pancreatic Cancer Research, UTMDACC, Houston, Texas, USA
| | - Kimal I Rajapakshe
- Translational Molecular Pathology, UTMDACC, Houston, Texas, USA
- Sheikh Ahmed Center for Pancreatic Cancer Research, UTMDACC, Houston, Texas, USA
| | - Benson Chellakkan Selvanesan
- Translational Molecular Pathology, UTMDACC, Houston, Texas, USA
- Sheikh Ahmed Center for Pancreatic Cancer Research, UTMDACC, Houston, Texas, USA
| | - Takashi Okumura
- Translational Molecular Pathology, UTMDACC, Houston, Texas, USA
- Sheikh Ahmed Center for Pancreatic Cancer Research, UTMDACC, Houston, Texas, USA
| | - Kenjiro Date
- Translational Molecular Pathology, UTMDACC, Houston, Texas, USA
- Sheikh Ahmed Center for Pancreatic Cancer Research, UTMDACC, Houston, Texas, USA
| | | | - Lotfi Abou-Elkacem
- Translational Molecular Pathology, UTMDACC, Houston, Texas, USA
- Sheikh Ahmed Center for Pancreatic Cancer Research, UTMDACC, Houston, Texas, USA
| | - Akiko Sagara
- Translational Molecular Pathology, UTMDACC, Houston, Texas, USA
- Sheikh Ahmed Center for Pancreatic Cancer Research, UTMDACC, Houston, Texas, USA
| | - Jimin Min
- Translational Molecular Pathology, UTMDACC, Houston, Texas, USA
- Sheikh Ahmed Center for Pancreatic Cancer Research, UTMDACC, Houston, Texas, USA
| | - Marta Sans
- Translational Molecular Pathology, UTMDACC, Houston, Texas, USA
- Sheikh Ahmed Center for Pancreatic Cancer Research, UTMDACC, Houston, Texas, USA
| | - Nathaniel Yee
- Translational Molecular Pathology, UTMDACC, Houston, Texas, USA
- Sheikh Ahmed Center for Pancreatic Cancer Research, UTMDACC, Houston, Texas, USA
| | - Megan J Siemann
- Translational Molecular Pathology, UTMDACC, Houston, Texas, USA
- Sheikh Ahmed Center for Pancreatic Cancer Research, UTMDACC, Houston, Texas, USA
| | - Jose Enriquez
- Cancer Systems Imaging, UTMDACC, Houston, Texas, USA
| | | | | | - Michael Kim
- Surgical Oncology, UTMDACC, Houston, Texas, USA
| | - Merve Dede
- Bioinformatics & Computational Biology, UTMDACC, Houston, Texas, USA
| | - Traver Hart
- Bioinformatics & Computational Biology, UTMDACC, Houston, Texas, USA
- Department of Cancer Biology, UTMDACC, Houston, Texas, USA
| | - Anirban Maitra
- Translational Molecular Pathology, UTMDACC, Houston, Texas, USA
- Sheikh Ahmed Center for Pancreatic Cancer Research, UTMDACC, Houston, Texas, USA
| | - Fredrik Ivar Thege
- Translational Molecular Pathology, UTMDACC, Houston, Texas, USA
- Sheikh Ahmed Center for Pancreatic Cancer Research, UTMDACC, Houston, Texas, USA
| |
Collapse
|
26
|
Fang G, Hao P, Qiao R, Liu BX, Shi X, Wang Z, Sun P. Stimuli-responsive chitosan based nanoparticles in cancer therapy and diagnosis: A review. Int J Biol Macromol 2024; 283:137709. [PMID: 39549789 DOI: 10.1016/j.ijbiomac.2024.137709] [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: 10/22/2024] [Revised: 11/02/2024] [Accepted: 11/13/2024] [Indexed: 11/18/2024]
Abstract
Chitosan, obtained through deacetylation of chitin, has been shown to a promising biopolymer for the development of nano- and micro-particles. In spite of inherent anti-cancer activity of chitosan, the employment of this carbohydrate polymer for the synthesis of nanoparticles opens a new gate in disease therapy. The properties of chitosan including biocompatibility, biodegradability, and modifiability are vital in enhancing these nanoparticles, allowing for improved solubility and interaction with cellular targets. Among the pathological events, cancer has demonstrated an increase in incidence rate and therefore, the chitosan nanoparticles have been significantly utilized in cancer therapy. The present review emphasizes on the role of stimuli-responsive chitosan nanoparticles in the field of cancer therapy. The stimuli-responsive nanoparticles can release the cargo in the tumor site that not only improves the anti-cancer activity of chemotherapy drugs, but also diminishes their systemic toxicity. The stimuli-responsive chitosan nanoparticles can respond to endogenous and exogenous stimuli including pH, redox and light to release cargo. This improves the specificity towards tumor cells and enhances accumulation of drugs and/or drugs. The light-responsive chitosan nanoparticles can cause photothermal and photodynamic therapy in tumor ablation and provide theranostic feature that is cancer diagnosis and therapy simultaneously.
Collapse
Affiliation(s)
- Guotao Fang
- Hebei Key Laboratory of Cancer Radiotherapy and Chemotherapy, Department of Medical Oncology, Affiliated Hospital of Hebei University, Baoding, Hebei, China
| | - Peng Hao
- Department of Joint Surgery, Southwest Hospital, Army Medical University, Third Military Medical University, Chongqing 400038, PR China
| | - Ruonan Qiao
- School of Basic Medicine, Hebei Medical University, Shijiazhuang, China
| | - Bi-Xia Liu
- Department of Gastroenterology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xiujuan Shi
- School of Medicine, Tongji university, Shanghai 200092, China.
| | - Zhenfei Wang
- The Laboratory for Tumor Molecular Diagnosis, Peking University Cancer Hospital, Inner Mongolia Campus, Afliated Cancer Hospital of Inner Mongolia Medical University, Hohhot 010020, China.
| | - Peng Sun
- Department of Ophthalmology, The First Affiliated Hospital of China Medical University, No. 155 Nanjing North Street, Shenyang, Liaoning, China.
| |
Collapse
|
27
|
Siciliano AC, Forciniti S, Onesto V, Iuele H, Cave DD, Carnevali F, Gigli G, Lonardo E, Del Mercato LL. A 3D Pancreatic Cancer Model with Integrated Optical Sensors for Noninvasive Metabolism Monitoring and Drug Screening. Adv Healthc Mater 2024; 13:e2401138. [PMID: 38978424 DOI: 10.1002/adhm.202401138] [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: 03/26/2024] [Revised: 06/06/2024] [Indexed: 07/10/2024]
Abstract
A distinct feature of pancreatic ductal adenocarcinoma (PDAC) is a prominent tumor microenvironment (TME) with remarkable cellular and spatial heterogeneity that meaningfully impacts disease biology and treatment resistance. The dynamic crosstalk between cancer cells and the dense stromal compartment leads to spatially and temporally heterogeneous metabolic alterations, such as acidic pH that contributes to drug resistance in PDAC. Thus, monitoring the extracellular pH metabolic fluctuations within the TME is crucial to predict and to quantify anticancer drug efficacy. Here, a simple and reliable alginate-based 3D PDAC model embedding ratiometric optical pH sensors and cocultures of tumor (AsPC-1) and stromal cells for simultaneously monitoring metabolic pH variations and quantify drug response is presented. By means of time-lapse confocal laser scanning microscopy (CLSM) coupled with a fully automated computational analysis, the extracellular pH metabolic variations are monitored and quantified over time during drug testing with gemcitabine, folfirinox, and paclitaxel, commonly used in PDAC therapy. In particular, the extracellular acidification is more pronounced after drugs treatment, resulting in increased antitumor effect correlated with apoptotic cell death. These findings highlight the importance of studying the influence of cellular metabolic mechanisms on tumor response to therapy in 3D tumor models, this being crucial for the development of personalized medicine approaches.
Collapse
Affiliation(s)
- Anna Chiara Siciliano
- Institute of Nanotechnology, National Research Council (Cnr-NANOTEC), c/o Campus Ecotekne, via Monteroni, Lecce, 73100, Italy
- Department of Mathematics and Physics "Ennio De Giorgi", University of Salento, c/o Campus Ecotekne, via Monteroni, Lecce, 73100, Italy
| | - Stefania Forciniti
- Institute of Nanotechnology, National Research Council (Cnr-NANOTEC), c/o Campus Ecotekne, via Monteroni, Lecce, 73100, Italy
| | - Valentina Onesto
- Institute of Nanotechnology, National Research Council (Cnr-NANOTEC), c/o Campus Ecotekne, via Monteroni, Lecce, 73100, Italy
| | - Helena Iuele
- Institute of Nanotechnology, National Research Council (Cnr-NANOTEC), c/o Campus Ecotekne, via Monteroni, Lecce, 73100, Italy
| | - Donatella Delle Cave
- Institute of Genetics and Biophysics Adriano Buzzati-Traverso, National Research Council (Cnr-IGB), Naples, 80131, Italy
| | - Federica Carnevali
- Institute of Nanotechnology, National Research Council (Cnr-NANOTEC), c/o Campus Ecotekne, via Monteroni, Lecce, 73100, Italy
- Department of Mathematics and Physics "Ennio De Giorgi", University of Salento, c/o Campus Ecotekne, via Monteroni, Lecce, 73100, Italy
| | - Giuseppe Gigli
- Institute of Nanotechnology, National Research Council (Cnr-NANOTEC), c/o Campus Ecotekne, via Monteroni, Lecce, 73100, Italy
- Department of Experimental Medicine, University of Salento, c/o Campus Ecotekne, via Monteroni, Lecce, 73100, Italy
| | - Enza Lonardo
- Institute of Genetics and Biophysics Adriano Buzzati-Traverso, National Research Council (Cnr-IGB), Naples, 80131, Italy
| | - Loretta L Del Mercato
- Institute of Nanotechnology, National Research Council (Cnr-NANOTEC), c/o Campus Ecotekne, via Monteroni, Lecce, 73100, Italy
| |
Collapse
|
28
|
Fox AC, Blazeck J. Applying metabolic control strategies to engineered T cell cancer therapies. Metab Eng 2024; 86:250-261. [PMID: 39490640 PMCID: PMC11611646 DOI: 10.1016/j.ymben.2024.10.009] [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: 07/25/2024] [Accepted: 10/22/2024] [Indexed: 11/05/2024]
Abstract
Chimeric antigen receptor (CAR) T cells are an engineered immunotherapy that express synthetic receptors to recognize and kill cancer cells. Despite their success in treating hematologic cancers, CAR T cells have limited efficacy against solid tumors, in part due to the altered immunometabolic profile within the tumor environment, which hinders T cell proliferation, infiltration, and anti-tumor activity. For instance, CAR T cells must compete for essential nutrients within tumors, while resisting the impacts of immunosuppressive metabolic byproducts. In this review, we will describe the altered metabolic features within solid tumors that contribute to immunosuppression of CAR T cells. We'll discuss how overexpression of key metabolic enzymes can enhance the ability of CAR T cells to resist corresponding tumoral metabolic changes or even revert the metabolic profile of a tumor to a less inhibitory state. In addition, metabolic remodeling is intrinsically linked to T cell activity, differentiation, and function, such that metabolic engineering strategies can also promote establishment of more or less efficacious CAR T cell phenotypes. Overall, we will show how applying metabolic engineering strategies holds significant promise in improving CAR T cells for the treatment of solid tumors.
Collapse
Affiliation(s)
- Andrea C Fox
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta GA 303332, USA
| | - John Blazeck
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta GA 303332, USA.
| |
Collapse
|
29
|
Bhattacharjya D, Sivalingam N. Mechanism of 5-fluorouracil induced resistance and role of piperine and curcumin as chemo-sensitizers in colon cancer. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:8445-8475. [PMID: 38878089 DOI: 10.1007/s00210-024-03189-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Accepted: 05/27/2024] [Indexed: 10/30/2024]
Abstract
Among cancer-related deaths worldwide, colorectal cancer ranks second, accounting for 1.2% of deaths in those under 50 years and 0.6% of deaths in those between 50 and 54 years. The anticancer drug 5-fluorouracil is widely used to treat colorectal cancer. Due to a better understanding of the drug's mechanism of action, its anticancer activity has been increased through a variety of therapeutic alternatives. Clinical use of 5-FU has been severely restricted due to drug resistance. The chemoresistance mechanism of 5-FU is challenging to overcome because of the existence of several drug efflux transporters, DNA repair enzymes, signaling cascades, classical cellular processes, cancer stem cells, metastasis, and angiogenesis. Curcumin, a potent phytocompound derived from Curcuma longa, functions as a nuclear factor (NF)-κB inhibitor and sensitizer to numerous chemotherapeutic drugs. Piperine, an alkaloid found in Piper longum, inhibits cancer cell growth, causing cell cycle arrest and apoptosis. This review explores the mechanism of 5-FU-induced chemoresistance in colon cancer cells and the role of curcumin and piperine in enhancing the sensitivity of 5-FU-based chemotherapy. CLINICAL TRIAL REGISTRATION: Not applicable.
Collapse
Affiliation(s)
- Dorothy Bhattacharjya
- Department of Biotechnology, School of Bioengineering, College of Engineering and Technology, Faculty of Engineering and Technology, SRM Institute of Science and Technology, SRM Nagar, Kattankulathur, 603 203, Chengalpattu District, Tamil Nadu, India
| | - Nageswaran Sivalingam
- Department of Biotechnology, School of Bioengineering, College of Engineering and Technology, Faculty of Engineering and Technology, SRM Institute of Science and Technology, SRM Nagar, Kattankulathur, 603 203, Chengalpattu District, Tamil Nadu, India.
| |
Collapse
|
30
|
Sabit H, Arneth B, Abdel-Ghany S, Madyan EF, Ghaleb AH, Selvaraj P, Shin DM, Bommireddy R, Elhashash A. Beyond Cancer Cells: How the Tumor Microenvironment Drives Cancer Progression. Cells 2024; 13:1666. [PMID: 39404428 PMCID: PMC11475877 DOI: 10.3390/cells13191666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 10/04/2024] [Accepted: 10/06/2024] [Indexed: 10/19/2024] Open
Abstract
Liver cancer represents a substantial global health challenge, contributing significantly to worldwide morbidity and mortality. It has long been understood that tumors are not composed solely of cancerous cells, but also include a variety of normal cells within their structure. These tumor-associated normal cells encompass vascular endothelial cells, fibroblasts, and various inflammatory cells, including neutrophils, monocytes, macrophages, mast cells, eosinophils, and lymphocytes. Additionally, tumor cells engage in complex interactions with stromal cells and elements of the extracellular matrix (ECM). Initially, the components of what is now known as the tumor microenvironment (TME) were thought to be passive bystanders in the processes of tumor proliferation and local invasion. However, recent research has significantly advanced our understanding of the TME's active role in tumor growth and metastasis. Tumor progression is now known to be driven by an intricate imbalance of positive and negative regulatory signals, primarily influenced by specific growth factors produced by both inflammatory and neoplastic cells. This review article explores the latest developments and future directions in understanding how the TME modulates liver cancer, with the aim of informing the design of novel therapies that target critical components of the TME.
Collapse
Affiliation(s)
- Hussein Sabit
- Department of Medical Biotechnology, College of Biotechnology, Misr University for Science and Technology, Giza P.O. Box 77, Egypt; (H.S.); (E.F.M.)
| | - Borros Arneth
- Institute of Laboratory Medicine and Pathobiochemistry, Molecular Diagnostics, Hospital of the Universities of Giessen and Marburg (UKGM), Philipps University Marburg, Baldinger Str., 35043 Marburg, Germany
- Institute of Laboratory Medicine and Pathobiochemistry, Molecular Diagnostics, Hospital of the Universities of Giessen and Marburg (UKGM), Justus Liebig University Giessen, Feulgenstr. 12, 35392 Giessen, Germany
| | - Shaimaa Abdel-Ghany
- Department of Environmental Biotechnology, College of Biotechnology, Misr University for Science and Technology, Giza P.O. Box 77, Egypt;
| | - Engy F. Madyan
- Department of Medical Biotechnology, College of Biotechnology, Misr University for Science and Technology, Giza P.O. Box 77, Egypt; (H.S.); (E.F.M.)
| | - Ashraf H. Ghaleb
- Department of Surgery, College of Medicine, Misr University for Science and Technology, Giza P.O. Box 77, Egypt;
- Department of Surgery, College of Medicine, Cairo University, Giza 12613, Egypt
| | - Periasamy Selvaraj
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA; (P.S.); (R.B.)
| | - Dong M. Shin
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA 30322, USA;
| | - Ramireddy Bommireddy
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA; (P.S.); (R.B.)
| | - Ahmed Elhashash
- Department of Biology, Texas A&M University, 3258 TAMU I, College Station, TX 77843-3258, USA
| |
Collapse
|
31
|
Fonte M, Rôla C, Santana S, Prudêncio M, Almeida J, Ferraz R, Prudêncio C, Teixeira C, Gomes P. Repurposing antiplasmodial leads for cancer: Exploring the antiproliferative effects of N-cinnamoyl-aminoacridines. Bioorg Med Chem Lett 2024; 111:129894. [PMID: 39043264 DOI: 10.1016/j.bmcl.2024.129894] [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: 04/14/2024] [Revised: 06/26/2024] [Accepted: 07/20/2024] [Indexed: 07/25/2024]
Abstract
Drug repurposing and rescuing have been widely explored as cost-effective approaches to expand the portfolio of chemotherapeutic agents. Based on the reported antitumor properties of both trans-cinnamic acids and quinacrine, an antimalarial aminoacridine, we explored the antiproliferative properties of two series of N-cinnamoyl-aminoacridines recently identified as multi-stage antiplasmodial leads. The compounds were evaluated in vitro against three cancer cell lines (MKN-28, Huh-7, and HepG2), and human primary dermal fibroblasts. One of the series displayed highly selective antiproliferative activity in the micromolar range against the three cancer cell lines tested, without any toxicity to non-carcinogenic cells.
Collapse
Affiliation(s)
- Mélanie Fonte
- LAQV-REQUIMTE, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, Portugal
| | - Catarina Rôla
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Portugal
| | - Sofia Santana
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Portugal
| | - Miguel Prudêncio
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Portugal
| | - Joana Almeida
- Centro de Investigação em Saúde Translacional e Biotecnologia Médica (TBIO)/Rede de Investigação em Saúde (RISE-Health), Escola Superior de Saúde, Instituto Politécnico do Porto, Portugal; Ciências Químicas e das Biomoléculas, Escola Superior de Saúde, Politécnico do Porto, Portugal
| | - Ricardo Ferraz
- LAQV-REQUIMTE, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, Portugal; Centro de Investigação em Saúde Translacional e Biotecnologia Médica (TBIO)/Rede de Investigação em Saúde (RISE-Health), Escola Superior de Saúde, Instituto Politécnico do Porto, Portugal; Ciências Químicas e das Biomoléculas, Escola Superior de Saúde, Politécnico do Porto, Portugal
| | - Cristina Prudêncio
- Centro de Investigação em Saúde Translacional e Biotecnologia Médica (TBIO)/Rede de Investigação em Saúde (RISE-Health), Escola Superior de Saúde, Instituto Politécnico do Porto, Portugal; Ciências Químicas e das Biomoléculas, Escola Superior de Saúde, Politécnico do Porto, Portugal
| | - Cátia Teixeira
- LAQV-REQUIMTE, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, Portugal; Gyros Protein Technologies Inc., Tucson, AZ, USA
| | - Paula Gomes
- LAQV-REQUIMTE, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, Portugal.
| |
Collapse
|
32
|
Osorio HM, Castillo-Solís F, Barragán SY, Rodríguez-Pólit C, Gonzalez-Pastor R. Graphene Quantum Dots from Natural Carbon Sources for Drug and Gene Delivery in Cancer Treatment. Int J Mol Sci 2024; 25:10539. [PMID: 39408866 PMCID: PMC11476599 DOI: 10.3390/ijms251910539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 09/10/2024] [Indexed: 10/20/2024] Open
Abstract
Cancer therapy is constantly evolving, with a growing emphasis on targeted and efficient treatment options. In this context, graphene quantum dots (GQDs) have emerged as promising agents for precise drug and gene delivery due to their unique attributes, such as high surface area, photoluminescence, up-conversion photoluminescence, and biocompatibility. GQDs can damage cancer cells and exhibit intrinsic photothermal conversion and singlet oxygen generation efficiency under specific light irradiation, enhancing their effectiveness. They serve as direct therapeutic agents and versatile drug delivery platforms capable of being easily functionalized with various targeting molecules and therapeutic agents. However, challenges such as achieving uniform size and morphology, precise bandgap engineering, and scalability, along with minimizing cytotoxicity and the environmental impact of their production, must be addressed. Additionally, there is a need for a more comprehensive understanding of cellular mechanisms and drug release processes, as well as improved purification methods. Integrating GQDs into existing drug delivery systems enhances the efficacy of traditional treatments, offering more efficient and less invasive options for cancer patients. This review highlights the transformative potential of GQDs in cancer therapy while acknowledging the challenges that researchers must overcome for broader application.
Collapse
Affiliation(s)
- Henrry M. Osorio
- Departamento de Física, Escuela Politécnica Nacional, Av. Ladrón de Guevara E11-253, Quito 170525, Ecuador; (H.M.O.); (S.Y.B.)
| | - Fabián Castillo-Solís
- Centro de Investigación Biomédica (CENBIO), Facultad de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Quito 170527, Ecuador; (F.C.-S.); (C.R.-P.)
| | - Selena Y. Barragán
- Departamento de Física, Escuela Politécnica Nacional, Av. Ladrón de Guevara E11-253, Quito 170525, Ecuador; (H.M.O.); (S.Y.B.)
| | - Cristina Rodríguez-Pólit
- Centro de Investigación Biomédica (CENBIO), Facultad de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Quito 170527, Ecuador; (F.C.-S.); (C.R.-P.)
- Escuela de Salud Pública, Universidad San Francisco de Quito USFQ, Quito 170527, Ecuador
- Centro de Referencia Nacional de Genómica, Secuenciación y Bioinformática, Instituto Nacional de Investigación en Salud Pública “Leopoldo Izquieta Pérez”, Quito 170403, Ecuador
| | - Rebeca Gonzalez-Pastor
- Centro de Investigación Biomédica (CENBIO), Facultad de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Quito 170527, Ecuador; (F.C.-S.); (C.R.-P.)
| |
Collapse
|
33
|
Omar H, Alkurdi YA, Fathima A, Alsharaeh EH. Investigation of the Application of Reduced Graphene Oxide-SPION Quantum Dots for Magnetic Hyperthermia. NANOMATERIALS (BASEL, SWITZERLAND) 2024; 14:1547. [PMID: 39404274 PMCID: PMC11477580 DOI: 10.3390/nano14191547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 09/05/2024] [Accepted: 09/22/2024] [Indexed: 10/19/2024]
Abstract
Integrating hyperthermia with conventional cancer therapies shows promise in improving treatment efficacy while mitigating their side effects. Nanotechnology-based hyperthermia, particularly using superparamagnetic iron oxide nanoparticles (SPIONs), offers a simplified solution for cancer treatment. In this study, we developed composites of SPION quantum dots (Fe3O4) with reduced graphene oxide (Fe3O4/RGO) using the coprecipitation method and investigated their potential application in magnetic hyperthermia. The size of Fe3O4 nanoparticles was controlled within the quantum dot range (≤10 nm) by varying the synthesis parameters, including reaction time as well as the concentration of ammonia and graphene oxide, where their biocompatibility was further improved with the inclusion of polyethylene glycol (PEG). These nanocomposites exhibited low cytotoxic effects on healthy cells (CHO-K1) over an incubation period of 24 h, though the inclusion of PEG enhanced their biocompatibility for longer incubation periods over 48 h. The Fe3O4/RGO composites dispersed in acidic pH buffer (pH 4.66) exhibited considerable heating effects, with the solution temperature increasing by ~10 °C within 5 min of exposure to pulsed magnetic fields, as compared to their dispersions in phosphate buffer and aqueous dimethylsulfoxide solutions. These results demonstrated the feasibility of using quantum dot Fe3O4/RGO composites for magnetic hyperthermia-based therapy to treat cancer, with further studies required to systematically optimize their magnetic properties and evaluate their efficacy for in vitro and in vivo applications.
Collapse
Affiliation(s)
| | | | | | - Edreese H. Alsharaeh
- College of Science and General Studies, Alfaisal University, P.O. Box 50927, Riyadh 11533, Saudi Arabia; (H.O.)
| |
Collapse
|
34
|
MK Nair P, Silwal K, Ramalakshmi R, Devibala M, Saranya M, Sivaranjani S, Ramasamy T, Palanisamy A, Mahalingam M. Beyond genetics: integrative oncology and the metabolic perspective on cancer treatment. Front Oncol 2024; 14:1455022. [PMID: 39376991 PMCID: PMC11456992 DOI: 10.3389/fonc.2024.1455022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 08/29/2024] [Indexed: 10/09/2024] Open
Abstract
Cancer is traditionally approached as a genetic disease, with standard treatments including chemotherapy, radiation, targeted therapy, immunotherapy, and surgery significantly improving survival rates and patient outcomes. However, there is a growing recognition of the need for integrative oncology, which expands cancer management by considering cancer as a metabolic disease. Integrative medicine physicians employ holistic therapies focused on patients' needs, aiming to correct the metabolic imbalances associated with cancer and alleviate cancer-related symptoms. Viewing cancer as a metabolic disease involves addressing factors such as an acidic microenvironment, vitamin C deficiency, mitochondrial dysfunction, reduced intracellular oxygen levels, elevated oxidative stress, dysfunctional autophagy, and psychological stress. This paper presents an overview of the evidence and comprehensive strategies supporting integrative medicine approaches in addressing cancer metabolism in integrative oncology settings. Furthermore, the paper underscores the necessity of integrating different cancer theories-genetic and metabolic-for improved patient outcomes and experiences. By combining these perspectives, integrative oncology offers a more holistic, patient-centered approach to cancer treatment.
Collapse
Affiliation(s)
- Pradeep MK Nair
- Department of Integrative Oncology, Mirakle Integrated Health Centre, Pollachi, India
| | - Karishma Silwal
- Department of Naturopathy, Sant Hirdaram Medical College of Naturopathy and Yogic Sciences, Bhopal, India
| | | | - Muniappan Devibala
- Department of Integrative Oncology, Mirakle Integrated Health Centre, Pollachi, India
| | | | - Sekar Sivaranjani
- Department of Integrative Oncology, Mirakle Integrated Health Centre, Pollachi, India
| | - Thangavelu Ramasamy
- Department of Integrative Oncology, Mirakle Integrated Health Centre, Pollachi, India
| | - Ayyappan Palanisamy
- Department of Integrative Oncology, Mirakle Integrated Health Centre, Pollachi, India
| | - Manickam Mahalingam
- Department of Integrative Oncology, Mirakle Integrated Health Centre, Pollachi, India
| |
Collapse
|
35
|
Bae JH, Kim HS. A pH-Responsive Protein Assembly through Clustering of a Charge-Tunable Single Amino Acid Repeat. ACS APPLIED MATERIALS & INTERFACES 2024; 16:47100-47109. [PMID: 39216082 DOI: 10.1021/acsami.4c07269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Specific targeting of tumor cells is a key to achieving high therapeutic efficacy while minimizing off-target side effects. As a general approach to targeting diverse tumor cells, considerable attention has been paid to the tumor microenvironment, particularly its slightly acidic pH (6.5-6.8). However, existing pH-sensitive nanomaterials, based on organic polymers and proteins, often lack sufficient pH sensitivity and specificity. Here, we demonstrate a strategy to construct a pH-responsive protein assembly through clustering of a single amino acid repeat as a charge-tunable moiety. As a proof of concept, a histidine peptide with varying lengths was displayed on the surface of a ferritin assembly composed of 24 subunits by genetic fusion to a subunit. The resulting self-assembled ferritin particles, termed "pHerricle (pH-responsive ferritin particle)", were shown to exhibit a specific binding to tumor cells in response to pH changes through cooperative effects of histidine peptides. Increasing the histidine peptide length from 0 to 12 residues increased the pHerricle's cell-binding capacity by 21-fold and allowed modulation of the targetable pH range. General applicability as a tumor cell-targeting platform was shown by specific delivery of a cytotoxic cargo by the pHerricle into tumor cells of various origins in a pH-dependent manner.
Collapse
Affiliation(s)
- Jin-Ho Bae
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 305-701, Korea
| | - Hak-Sung Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 305-701, Korea
| |
Collapse
|
36
|
Lu Q, Wang X, Fan X, Lin J, Hu J, Duan G, Yu H, Geng Z, Wang X, Dai H, Liu F, Wen L, Geng H. Wintersweet-like Nanohybrids of Titanium-doped Cerium Vanadate Loaded with Polypyrrole for Tumor Theranostic. Adv Healthc Mater 2024; 13:e2400830. [PMID: 38857527 DOI: 10.1002/adhm.202400830] [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: 03/04/2024] [Revised: 05/25/2024] [Indexed: 06/12/2024]
Abstract
Compromises between enhanced on-targeting reactivity and precise real-time monitoring in the tumor microenvironment (TME) are the main roadblocks for catalytic cancer therapy. The hallmark of a high level of hydrogen peroxide (H2O2) and acidic extracellular environment of the hypoxia solid tumor can underpin therapeutic and tracking performance. Herein, this work provides an activatable wintersweet-like nanohybrid consisting of titanium (Ti) doped cerium vanadate nanorods with the modification of polypyrrole (PPy) nanoparticles (CeVO4-Ti@PPy) for combinatorial therapies of breast carcinoma. The Ti dopants in the size-controllable CeVO4 nanorods lower the energy barrier (0.5 eV) of the rate-determining steps and elaborate peroxidase-like (POD-like) activities to improve the generation of toxic hydroxyl radical (·OH) according to the density functional theory (DFT) calculation. The multiple enzyme-like activities, including the intrinsic glutathione peroxidase (GPx) and catalase (CAT), achieve a record-high therapeutic efficiency. Coupling this oxidative stress with the photothermal effects of PPy enables enhanced catalytic tumor necrosis. The exterior PPy heterogeneous structure can be further doped with protons in the local acidic environment to intensify photoacoustic signals, allowing the non-invasive accurate tracking of tumors. The theranostic performance displayed negligible attenuated signals in near-infrared (NIR) windows. This organic-inorganic nanohybrid with a heterogeneous structure provides the potential to improve the overall outcomes of catalytic therapy.
Collapse
Affiliation(s)
- Qianyun Lu
- School of Environmental and Chemical Engineering, Jiangsu University of Science and Technology, Zhenjiang, Jiangsu, 212003, China
- Institute of Biomedical and Health Engineering, Shenzhen International Graduate School, Tsinghua University, Shenzhen, Guangdong, 518055, China
| | - Xiaotong Wang
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Suzhou, Jiangsu, 215123, China
| | - Xin Fan
- Institute of Biomedical and Health Engineering, Shenzhen International Graduate School, Tsinghua University, Shenzhen, Guangdong, 518055, China
| | - Jinguo Lin
- State Key Laboratory of Nonlinear Mechanics, Institute of Mechanics, Chinese Academy of Sciences, Beijing, 100190, China
| | - Jiayi Hu
- Institute of Biomedical and Health Engineering, Shenzhen International Graduate School, Tsinghua University, Shenzhen, Guangdong, 518055, China
| | - Guangxin Duan
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Suzhou, Jiangsu, 215123, China
| | - Huimin Yu
- Department of Chemical Engineering, Center for Synthetic and Systems Biology, Tsinghua University, Beijing, 100084, China
| | - Zihan Geng
- Tsinghua-Berkeley Shenzhen Institute, Shenzhen International Graduate School, Tsinghua University, Shenzhen, Guangdong, 518055, China
| | - Xingang Wang
- School of Environmental and Chemical Engineering, Jiangsu University of Science and Technology, Zhenjiang, Jiangsu, 212003, China
| | - Hongliang Dai
- School of Environmental and Chemical Engineering, Jiangsu University of Science and Technology, Zhenjiang, Jiangsu, 212003, China
| | - Feng Liu
- State Key Laboratory of Nonlinear Mechanics, Institute of Mechanics, Chinese Academy of Sciences, Beijing, 100190, China
| | - Ling Wen
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Suzhou, Jiangsu, 215123, China
| | - Hongya Geng
- Institute of Biomedical and Health Engineering, Shenzhen International Graduate School, Tsinghua University, Shenzhen, Guangdong, 518055, China
| |
Collapse
|
37
|
Kunachowicz D, Kłosowska K, Sobczak N, Kepinska M. Applicability of Quantum Dots in Breast Cancer Diagnostic and Therapeutic Modalities-A State-of-the-Art Review. NANOMATERIALS (BASEL, SWITZERLAND) 2024; 14:1424. [PMID: 39269086 PMCID: PMC11396817 DOI: 10.3390/nano14171424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 08/27/2024] [Accepted: 08/28/2024] [Indexed: 09/15/2024]
Abstract
The increasing incidence of breast cancers (BCs) in the world population and their complexity and high metastatic ability are serious concerns for healthcare systems. Despite the significant progress in medicine made in recent decades, the efficient treatment of invasive cancers still remains challenging. Chemotherapy, a fundamental systemic treatment method, is burdened with severe adverse effects, with efficacy limited by resistance development and risk of disease recurrence. Also, current diagnostic methods have certain drawbacks, attracting attention to the idea of developing novel, more sensitive detection and therapeutic modalities. It seems the solution for these issues can be provided by nanotechnology. Particularly, quantum dots (QDs) have been extensively evaluated as potential targeted drug delivery vehicles and, simultaneously, sensing and bioimaging probes. These fluorescent nanoparticles offer unlimited possibilities of surface modifications, allowing for the attachment of biomolecules, such as antibodies or proteins, and drug molecules, among others. In this work, we discuss the potential applicability of QDs in breast cancer diagnostics and treatment in light of the current knowledge. We begin with introducing the molecular and histopathological features of BCs, standard therapeutic regimens, and current diagnostic methods. Further, the features of QDs, along with their uptake, biodistribution patterns, and cytotoxicity, are described. Based on the reports published in recent years, we present the progress in research on possible QD use in improving BC diagnostics and treatment efficacy as chemotherapeutic delivery vehicles and photosensitizing agents, along with the stages of their development. We also address limitations and open questions regarding this topic.
Collapse
Affiliation(s)
- Dominika Kunachowicz
- Department of Pharmaceutical Biochemistry, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland
| | - Karolina Kłosowska
- Students' Scientific Association at the Department of Pharmaceutical Biochemistry (SKN No. 214), Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland
| | - Natalia Sobczak
- Students' Scientific Association of Biomedical and Environmental Analyses (SKN No. 85), Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland
| | - Marta Kepinska
- Department of Pharmaceutical Biochemistry, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland
| |
Collapse
|
38
|
Brooks A, Zhang Y, Chen J, Zhao CX. Cancer Metastasis-on-a-Chip for Modeling Metastatic Cascade and Drug Screening. Adv Healthc Mater 2024; 13:e2302436. [PMID: 38224141 DOI: 10.1002/adhm.202302436] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 01/06/2024] [Indexed: 01/16/2024]
Abstract
Microfluidic chips are valuable tools for studying intricate cellular and cell-microenvironment interactions. Traditional in vitro cancer models lack accuracy in mimicking the complexities of in vivo tumor microenvironment. However, cancer-metastasis-on-a-chip (CMoC) models combine the advantages of 3D cultures and microfluidic technology, serving as powerful platforms for exploring cancer mechanisms and facilitating drug screening. These chips are able to compartmentalize the metastatic cascade, deepening the understanding of its underlying mechanisms. This article provides an overview of current CMoC models, focusing on distinctive models that simulate invasion, intravasation, circulation, extravasation, and colonization, and their applications in drug screening. Furthermore, challenges faced by CMoC and microfluidic technologies are discussed, while exploring promising future directions in cancer research. The ongoing development and integration of these models into cancer studies are expected to drive transformative advancements in the field.
Collapse
Affiliation(s)
- Anastasia Brooks
- School of Chemical Engineering, University of Adelaide, Adelaide, 5005, Australia
| | - Yali Zhang
- School of Chemical Engineering, University of Adelaide, Adelaide, 5005, Australia
| | - Jiezhong Chen
- School of Chemical Engineering, University of Adelaide, Adelaide, 5005, Australia
| | - Chun-Xia Zhao
- School of Chemical Engineering, University of Adelaide, Adelaide, 5005, Australia
| |
Collapse
|
39
|
Liang X, Chen W, Wang C, Jiang K, Zhu J, Lu R, Lin Z, Cao Z, Zheng J. A mesoporous theranostic platform for ultrasound and photoacoustic dual imaging-guided photothermal and enhanced starvation therapy for cancer. Acta Biomater 2024; 183:264-277. [PMID: 38815685 DOI: 10.1016/j.actbio.2024.05.040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 05/14/2024] [Accepted: 05/23/2024] [Indexed: 06/01/2024]
Abstract
Tumor starvation therapy utilizing glucose oxidase (GOx), has gained traction due to its non-invasive and bio-safe attributes. However, its effectiveness is often hampered by severe hypoxia in the tumor microenvironment (TME), limiting GOx's catalytic activity. To address this issue, a multifunctional nanosystem based on mesoporous polydopamine nanoparticles (MPDA NPs) was developled to alleviate TME hypoxia. This nanosystem integrated GOx modification and oxygenated perfluoropentane (PFP) encapsulation to address hypoxia-related challenges in the TME. Under NIR laser irradiation, the MPDA NPs exhibit significant photothermal conversion efficacy, activating targeted tumor photothermal therapy (PTT), while also serving as proficient photoacoustic (PA) imaging agents. The ensuing temperature rise facilitates oxygen (O2) release and induces liquid-gas conversion of PFP, generating microbubbles for enhanced ultrasound (US) imaging signals. The supplied oxygen alleviates local hypoxia, thereby enhancing GOx-mediated endogenous glucose consumption for tumor starvation. Overall, the integration of ultrasound/photoacoustic dual imaging-guided PTT and starvation therapy within MPDA-GOx@PFP@O2 nanoparticles (MGPO NPs) presents a promising platform for enhancing the efficacay of tumor treatment by overcoming the complexities of the TME. STATEMENT OF SIGNIFICANCE: A multifunctional MPDA-based theranostic nanoagent was developed for US/PAI imaging-guided PTT and starvation therapy against tumor hypoxia by direct O2 delivery. The incorporation of oxygenated perfluoropentane (PFP) within the mesoporous structure of MGPO not only enables efficient US imaging but also helps in alleviating tumor hypoxia. Moreover, the strong near-infrared (NIR) absorption of MGPO NPs promote the generation of PFP microbubbles and release of oxygen, thereby enhancing US imaging and GOx-mediated starvation therapy. Such a multifunctional nanosystem leverages synergistic effects to enhance therapeutic efficacy while incorporating US/PA imaging for precise visualization of the tumor.
Collapse
Affiliation(s)
- Xiaotong Liang
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, No. 66. Gongchang Road, Guangming District, Shenzhen, Guangdong 518107, China
| | - Wenbo Chen
- Ultrasound Department of The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong, Shenzhen & Longgang District People's Hospital of Shenzhen, Shenzhen, 518172, China
| | - Chunan Wang
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, No. 66. Gongchang Road, Guangming District, Shenzhen, Guangdong 518107, China
| | - Kai Jiang
- Shenzhen International Institute for Biomedical Research, Longhua District, Shenzhen, 518116, Guangdong, China
| | - Jinjin Zhu
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, No. 66. Gongchang Road, Guangming District, Shenzhen, Guangdong 518107, China
| | - Ruitao Lu
- Shenzhen International Institute for Biomedical Research, Longhua District, Shenzhen, 518116, Guangdong, China
| | - Zhousheng Lin
- Department of Orthopaedics, Guangdong Second Provincial General Hospital, Guangzhou, Guangdong 510317, China
| | - Zhong Cao
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, No. 66. Gongchang Road, Guangming District, Shenzhen, Guangdong 518107, China; Shenzhen International Institute for Biomedical Research, Longhua District, Shenzhen, 518116, Guangdong, China.
| | - Jian Zheng
- Ultrasound Department of The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong, Shenzhen & Longgang District People's Hospital of Shenzhen, Shenzhen, 518172, China.
| |
Collapse
|
40
|
Garlisi B, Lauks S, Aitken C, Ogilvie LM, Lockington C, Petrik D, Eichhorn JS, Petrik J. The Complex Tumor Microenvironment in Ovarian Cancer: Therapeutic Challenges and Opportunities. Curr Oncol 2024; 31:3826-3844. [PMID: 39057155 PMCID: PMC11275383 DOI: 10.3390/curroncol31070283] [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: 05/15/2024] [Revised: 06/27/2024] [Accepted: 06/28/2024] [Indexed: 07/28/2024] Open
Abstract
The tumor microenvironment (TME) in ovarian cancer (OC) has much greater complexity than previously understood. In response to aggressive pro-angiogenic stimulus, blood vessels form rapidly and are dysfunctional, resulting in poor perfusion, tissue hypoxia, and leakiness, which leads to increased interstitial fluid pressure (IFP). Decreased perfusion and high IFP significantly inhibit the uptake of therapies into the tumor. Within the TME, there are numerous inhibitor cells, such as myeloid-derived suppressor cells (MDSCs), tumor association macrophages (TAMs), regulatory T cells (Tregs), and cancer-associated fibroblasts (CAFs) that secrete high numbers of immunosuppressive cytokines. This immunosuppressive environment is thought to contribute to the lack of success of immunotherapies such as immune checkpoint inhibitor (ICI) treatment. This review discusses the components of the TME in OC, how these characteristics impede therapeutic efficacy, and some strategies to alleviate this inhibition.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Jim Petrik
- Department of Biomedical Sciences, University of Guelph, Guelph, ON N1G 2W1, Canada; (B.G.); (S.L.); (C.A.); (L.M.O.); (C.L.); (D.P.); (J.S.E.)
| |
Collapse
|
41
|
Alshehri AM, Wilson OC. Biomimetic Hydrogel Strategies for Cancer Therapy. Gels 2024; 10:437. [PMID: 39057460 PMCID: PMC11275631 DOI: 10.3390/gels10070437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 06/18/2024] [Accepted: 06/28/2024] [Indexed: 07/28/2024] Open
Abstract
Recent developments in biomimetic hydrogel research have expanded the scope of biomedical technologies that can be used to model, diagnose, and treat a wide range of medical conditions. Cancer presents one of the most intractable challenges in this arena due to the surreptitious mechanisms that it employs to evade detection and treatment. In order to address these challenges, biomimetic design principles can be adapted to beat cancer at its own game. Biomimetic design strategies are inspired by natural biological systems and offer promising opportunities for developing life-changing methods to model, detect, diagnose, treat, and cure various types of static and metastatic cancers. In particular, focusing on the cellular and subcellular phenomena that serve as fundamental drivers for the peculiar behavioral traits of cancer can provide rich insights into eradicating cancer in all of its manifestations. This review highlights promising developments in biomimetic nanocomposite hydrogels that contribute to cancer therapies via enhanced drug delivery strategies and modeling cancer mechanobiology phenomena in relation to metastasis and synergistic sensing systems. Creative efforts to amplify biomimetic design research to advance the development of more effective cancer therapies will be discussed in alignment with international collaborative goals to cure cancer.
Collapse
Affiliation(s)
- Awatef M. Alshehri
- Department of Biomedical Engineering, The Catholic University of America, Washington, DC 20064, USA
- Department of Nanomedicine, King Abdullah International Medical Research Center (KAIMRC), King Saud bin Abdelaziz University for Health Sciences (KSAU-HS), Ministry of National Guard-Health Affairs (MNGHA), Riyadh 11426, Saudi Arabia;
| | - Otto C. Wilson
- Department of Biomedical Engineering, The Catholic University of America, Washington, DC 20064, USA
| |
Collapse
|
42
|
Sterin I, Tverdokhlebova A, Katz E, Smutok O. Time-Separated Pulse Release-Activation of an Enzyme from Alginate-Polyethylenimine Hydrogels Using Electrochemically Generated Local pH Changes. ACS APPLIED MATERIALS & INTERFACES 2024; 16:28222-28229. [PMID: 38779815 DOI: 10.1021/acsami.4c05273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2024]
Abstract
β-Glucosidase (EC 3.2.1.21) from sweet almond was encapsulated into pH-responsive alginate-polyethylenimine (alginate-PEI) hydrogel. Then, electrochemically controlled cyclic local pH changes resulting from ascorbate oxidation (acidification) and oxygen reduction (basification) were used for the pulsatile release of the enzyme from the composite hydrogel. Activation of the enzyme was controlled by the very same pH changes used for β-glucosidase release, separating these two processes in time. Importantly, the activity of the enzyme, which had not been released yet, was inhibited due to the buffering effect of PEI present in the gel. Thus, only a portion of the released enzyme was activated. Both enzymatic activity and release were monitored by confocal fluorescence microscopy and regular fluorescent spectroscopy. Namely, commercially available very little or nonfluorescent substrate 4-methylumbelliferyl-β-d-glucopyranoside was hydrolyzed by β-glucosidase to produce a highly fluorescent product 4-methylumbelliferone during the activation phase. At the same time, labeling of the enzyme with rhodamine B isothiocyanate was used for release observation. The proposed work represents an interesting smart release-activation system with potential applications in biomedical field.
Collapse
Affiliation(s)
- Ilya Sterin
- Department of Chemistry and Biomolecular Science, Clarkson University, Potsdam, New York 13699, United States
| | - Anna Tverdokhlebova
- Department of Chemistry and Biomolecular Science, Clarkson University, Potsdam, New York 13699, United States
| | - Evgeny Katz
- Department of Chemistry and Biomolecular Science, Clarkson University, Potsdam, New York 13699, United States
| | - Oleh Smutok
- Department of Chemistry and Biomolecular Science, Clarkson University, Potsdam, New York 13699, United States
| |
Collapse
|
43
|
Kuznetsova AB, Kolesova EP, Parodi A, Zamyatnin AA, Egorova VS. Reprogramming Tumor-Associated Macrophage Using Nanocarriers: New Perspectives to Halt Cancer Progression. Pharmaceutics 2024; 16:636. [PMID: 38794298 PMCID: PMC11124960 DOI: 10.3390/pharmaceutics16050636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 05/03/2024] [Accepted: 05/07/2024] [Indexed: 05/26/2024] Open
Abstract
Cancer remains a significant challenge for public healthcare systems worldwide. Within the realm of cancer treatment, considerable attention is focused on understanding the tumor microenvironment (TME)-the complex network of non-cancerous elements surrounding the tumor. Among the cells in TME, tumor-associated macrophages (TAMs) play a central role, traditionally categorized as pro-inflammatory M1 macrophages or anti-inflammatory M2 macrophages. Within the TME, M2-like TAMs can create a protective environment conducive to tumor growth and progression. These TAMs secrete a range of factors and molecules that facilitate tumor angiogenesis, increased vascular permeability, chemoresistance, and metastasis. In response to this challenge, efforts are underway to develop adjuvant therapy options aimed at reprogramming TAMs from the M2 to the anti-tumor M1 phenotype. Such reprogramming holds promise for suppressing tumor growth, alleviating chemoresistance, and impeding metastasis. Nanotechnology has enabled the development of nanoformulations that may soon offer healthcare providers the tools to achieve targeted drug delivery, controlled drug release within the TME for TAM reprogramming and reduce drug-related adverse events. In this review, we have synthesized the latest data on TAM polarization in response to TME factors, highlighted the pathological effects of TAMs, and provided insights into existing nanotechnologies aimed at TAM reprogramming and depletion.
Collapse
Affiliation(s)
- Alyona B. Kuznetsova
- Scientific Center for Translation Medicine, Sirius University of Science and Technology, 354340 Sochi, Russia; (A.B.K.); (E.P.K.); (A.P.)
| | - Ekaterina P. Kolesova
- Scientific Center for Translation Medicine, Sirius University of Science and Technology, 354340 Sochi, Russia; (A.B.K.); (E.P.K.); (A.P.)
| | - Alessandro Parodi
- Scientific Center for Translation Medicine, Sirius University of Science and Technology, 354340 Sochi, Russia; (A.B.K.); (E.P.K.); (A.P.)
| | - Andrey A. Zamyatnin
- Scientific Center for Translation Medicine, Sirius University of Science and Technology, 354340 Sochi, Russia; (A.B.K.); (E.P.K.); (A.P.)
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, 119234 Moscow, Russia
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992 Moscow, Russia
- Department of Biological Chemistry, Sechenov First Moscow State Medical University, 119991 Moscow, Russia
| | - Vera S. Egorova
- Scientific Center for Translation Medicine, Sirius University of Science and Technology, 354340 Sochi, Russia; (A.B.K.); (E.P.K.); (A.P.)
| |
Collapse
|
44
|
Carmignani A, Battaglini M, Marino A, Pignatelli F, Ciofani G. Drug-Loaded Polydopamine Nanoparticles for Chemo/Photothermal Therapy against Colorectal Cancer Cells. ACS APPLIED BIO MATERIALS 2024; 7:2205-2217. [PMID: 38489294 DOI: 10.1021/acsabm.3c01203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/17/2024]
Abstract
Colorectal cancer (CRC) is a common and deadly malignancy, ranking second in terms of mortality and third in terms of incidence on a global scale. The survival rates for CRC patients are unsatisfactory primarily because of the absence of highly effective clinical strategies. The efficacy of existing CRC treatments, such as chemotherapy (CT), is constrained by issues such as drug resistance and damage to healthy tissues. Alternative approaches such as photothermal therapy (PTT), while offering advantages over traditional therapies, suffer instead from a low efficiency in killing tumor cells when used alone. In this context, nanostructures can efficiently contribute to a selective and targeted treatment. Here, we combined CT and PTT by developing a nanoplatform based on polydopamine nanoparticles (PDNPs), selected for their biocompatibility, drug-carrying capabilities, and ability to produce heat upon exposure to near-infrared (NIR) irradiation. As a chemotherapy drug, sorafenib has been selected, a multikinase inhibitor already approved for clinical use. By encapsulating sorafenib in polydopamine nanoparticles (Sor-PDNPs), we were able to successfully improve the drug stability in physiological media and the consequent uptake by CRC cells, thereby increasing its therapeutic effects. Upon NIR stimulus, Sor-PDNPs can induce a temperature increment of about 10 °C, encompassing both PTT and triggering a localized and massive drug release. Sor-PDNPs were tested on healthy colon cells, showing minimal adverse outcomes; conversely, they demonstrated excellent efficacy against CRC cells, with a strong capability to hinder cancer cell proliferation and induce apoptosis. Obtained findings pave the way to new synergistic chemo-photothermal approaches, maximizing the therapeutic outcomes against CRC while minimizing side effects on healthy cells.
Collapse
Affiliation(s)
- Alessio Carmignani
- Smart Bio-Interfaces, Istituto Italiano di Tecnologia, Viale Rinaldo Piaggio 34, 56025 Pontedera, Italy
- Scuola Superiore Sant'Anna, The Biorobotics Institute, Viale Rinaldo Piaggio 34, 56025 Pontedera, Italy
| | - Matteo Battaglini
- Smart Bio-Interfaces, Istituto Italiano di Tecnologia, Viale Rinaldo Piaggio 34, 56025 Pontedera, Italy
| | - Attilio Marino
- Smart Bio-Interfaces, Istituto Italiano di Tecnologia, Viale Rinaldo Piaggio 34, 56025 Pontedera, Italy
| | - Francesca Pignatelli
- Smart Bio-Interfaces, Istituto Italiano di Tecnologia, Viale Rinaldo Piaggio 34, 56025 Pontedera, Italy
| | - Gianni Ciofani
- Smart Bio-Interfaces, Istituto Italiano di Tecnologia, Viale Rinaldo Piaggio 34, 56025 Pontedera, Italy
| |
Collapse
|
45
|
Guerrache A, Micheau O. TNF-Related Apoptosis-Inducing Ligand: Non-Apoptotic Signalling. Cells 2024; 13:521. [PMID: 38534365 PMCID: PMC10968836 DOI: 10.3390/cells13060521] [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: 02/07/2024] [Revised: 03/01/2024] [Accepted: 03/14/2024] [Indexed: 03/28/2024] Open
Abstract
TNF-related apoptosis-inducing ligand (TRAIL or Apo2 or TNFSF10) belongs to the TNF superfamily. When bound to its agonistic receptors, TRAIL can induce apoptosis in tumour cells, while sparing healthy cells. Over the last three decades, this tumour selectivity has prompted many studies aiming at evaluating the anti-tumoral potential of TRAIL or its derivatives. Although most of these attempts have failed, so far, novel formulations are still being evaluated. However, emerging evidence indicates that TRAIL can also trigger a non-canonical signal transduction pathway that is likely to be detrimental for its use in oncology. Likewise, an increasing number of studies suggest that in some circumstances TRAIL can induce, via Death receptor 5 (DR5), tumour cell motility, potentially leading to and contributing to tumour metastasis. While the pro-apoptotic signal transduction machinery of TRAIL is well known from a mechanistic point of view, that of the non-canonical pathway is less understood. In this study, we the current state of knowledge of TRAIL non-canonical signalling.
Collapse
Affiliation(s)
- Abderrahmane Guerrache
- Université de Bourgogne, 21000 Dijon, France
- INSERM Research Center U1231, «Equipe DesCarTes», 21000 Dijon, France
| | - Olivier Micheau
- Université de Bourgogne, 21000 Dijon, France
- INSERM Research Center U1231, «Equipe DesCarTes», 21000 Dijon, France
- Laboratoire d’Excellence LipSTIC, 21000 Dijon, France
| |
Collapse
|
46
|
Malik S, Biswas J, Sarkar P, Nag S, Gain C, Ghosh Roy S, Bhattacharya B, Ghosh D, Saha A. Differential carbonic anhydrase activities control EBV-induced B-cell transformation and lytic cycle reactivation. PLoS Pathog 2024; 20:e1011998. [PMID: 38530845 PMCID: PMC10997083 DOI: 10.1371/journal.ppat.1011998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 04/05/2024] [Accepted: 03/01/2024] [Indexed: 03/28/2024] Open
Abstract
Epstein-Barr virus (EBV) contributes to ~1% of all human cancers including several B-cell neoplasms. A characteristic feature of EBV life cycle is its ability to transform metabolically quiescent B-lymphocytes into hyperproliferating B-cell blasts with the establishment of viral latency, while intermittent lytic cycle induction is necessary for the production of progeny virus. Our RNA-Seq analyses of both latently infected naïve B-lymphocytes and transformed B-lymphocytes upon lytic cycle replication indicate a contrasting expression pattern of a membrane-associated carbonic anhydrase isoform CA9, an essential component for maintaining cell acid-base homeostasis. We show that while CA9 expression is transcriptionally activated during latent infection model, lytic cycle replication restrains its expression. Pharmacological inhibition of CA-activity using specific inhibitors retards EBV induced B-cell transformation, inhibits B-cells outgrowth and colony formation ability of transformed B-lymphocytes through lowering the intracellular pH, induction of cell apoptosis and facilitating degradation of CA9 transcripts. Reanalyses of ChIP-Seq data along with utilization of EBNA2 knockout virus, ectopic expression of EBNA2 and sh-RNA mediated knockdown of CA9 expression we further demonstrate that EBNA2 mediated CA9 transcriptional activation is essential for EBV latently infected B-cell survival. In contrast, during lytic cycle reactivation CA9 expression is transcriptionally suppressed by the key EBV lytic cycle transactivator, BZLF1 through its transactivation domain. Overall, our study highlights the dynamic alterations of CA9 expression and its activity in regulating pH homeostasis act as one of the major drivers for EBV induced B-cell transformation and subsequent B-cell lymphomagenesis.
Collapse
Affiliation(s)
- Samaresh Malik
- Institute of Health Sciences, Presidency University, Kolkata, West Bengal, India
| | - Joyanta Biswas
- Institute of Health Sciences, Presidency University, Kolkata, West Bengal, India
| | - Purandar Sarkar
- Institute of Health Sciences, Presidency University, Kolkata, West Bengal, India
| | - Subhadeep Nag
- Institute of Health Sciences, Presidency University, Kolkata, West Bengal, India
| | - Chandrima Gain
- Institute of Health Sciences, Presidency University, Kolkata, West Bengal, India
| | - Shatadru Ghosh Roy
- Institute of Health Sciences, Presidency University, Kolkata, West Bengal, India
| | - Bireswar Bhattacharya
- National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal, India
| | - Dipanjan Ghosh
- National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal, India
| | - Abhik Saha
- Institute of Health Sciences, Presidency University, Kolkata, West Bengal, India
| |
Collapse
|
47
|
Mancini C, Lori G, Pranzini E, Taddei ML. Metabolic challengers selecting tumor-persistent cells. Trends Endocrinol Metab 2024; 35:263-276. [PMID: 38071164 DOI: 10.1016/j.tem.2023.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 11/14/2023] [Accepted: 11/18/2023] [Indexed: 03/14/2024]
Abstract
Resistance to anticancer therapy still represents one of the main obstacles to cancer treatment. Numerous components of the tumor microenvironment (TME) contribute significantly to the acquisition of drug resistance. Microenvironmental pressures arising during cancer evolution foster tumor heterogeneity (TH) and facilitate the emergence of drug-resistant clones. In particular, metabolic pressures arising in the TME may favor epigenetic adaptations supporting the acquisition of persistence features in tumor cells. Tumor-persistent cells (TPCs) are characterized by high phenotypic and metabolic plasticity, representing a noticeable advantage in chemo- and radio-resistance. Understanding the crosslink between the evolution of metabolic pressures in the TME, epigenetics, and TPC evolution is significant for developing novel therapeutic strategies specifically targeting TPC vulnerabilities to overcome drug resistance.
Collapse
Affiliation(s)
- Caterina Mancini
- Department of Experimental and Clinical Medicine, University of Florence, Viale Morgagni 50, 50134 Florence, Italy
| | - Giulia Lori
- Department of Experimental and Clinical Medicine, University of Florence, Viale Morgagni 50, 50134 Florence, Italy
| | - Erica Pranzini
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale Morgagni 50, 50134 Florence, Italy.
| | - Maria Letizia Taddei
- Department of Experimental and Clinical Medicine, University of Florence, Viale Morgagni 50, 50134 Florence, Italy.
| |
Collapse
|
48
|
Gillard M, Troian-Gautier L, Decottignies A, Elias B. pH-Activatable Ruthenium(II) Fluorescein Salphen Schiff Base Photosensitizers for Theranostic Applications. J Med Chem 2024; 67:2549-2558. [PMID: 38345026 DOI: 10.1021/acs.jmedchem.3c01678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2024]
Abstract
Ruthenium(II) polypyridyl complexes exhibit a lack of selectivity toward cancer tissues despite extensive studies as photosensitizers for photodynamic therapy (PDT). Here, we report pH-activatable RuII photosensitizers for molecularly targeted PDT by exploiting the higher acidity of tumoral tissue. The fluorescein moiety, well known for its high pH sensitivity, was connected to a RuII center to yield novel photosensitizers for pH-sensitive 1O2 photogeneration. Their ability to photosensitize molecular dioxygen was studied at various pHs and revealed a drastic enhancement from 0.07 to 0.66 of the 1O2 quantum yield under acidic conditions (pH 7.5 to pH 5.5). Their photocytotoxicity against U2OS osteosarcoma cells was also investigated at pH 5.5 and 7.5 through IC50 determination. A strong enhancement of the photocytotoxicity reaching 930 nM was observed at pH 5.5, which showed the potential of such photosensitizers for pH-activatable PDT.
Collapse
Affiliation(s)
- Martin Gillard
- Institut de la Matière Condensée et des Nanosciences (IMCN), Molecular Chemistry, Materials and Catalysis (MOST), Université catholique de Louvain (UCLouvain), Place Louis Pasteur 1, bte L4.01.02, B-1348 Louvain-la-Neuve, Belgium
| | - Ludovic Troian-Gautier
- Institut de la Matière Condensée et des Nanosciences (IMCN), Molecular Chemistry, Materials and Catalysis (MOST), Université catholique de Louvain (UCLouvain), Place Louis Pasteur 1, bte L4.01.02, B-1348 Louvain-la-Neuve, Belgium
- Wel Research Institute, Avenue Pasteur 6, 1300 Wavre, Belgium
| | - Anabelle Decottignies
- Genetic and Epigenetic Alterations of Genomes, de Duve Institute, Université catholique de Louvain (UCLouvain), Avenue Hippocrate 75, 1200 Brussels, Belgium
| | - Benjamin Elias
- Institut de la Matière Condensée et des Nanosciences (IMCN), Molecular Chemistry, Materials and Catalysis (MOST), Université catholique de Louvain (UCLouvain), Place Louis Pasteur 1, bte L4.01.02, B-1348 Louvain-la-Neuve, Belgium
| |
Collapse
|
49
|
Jardim-Perassi BV, Irrera P, Oluwatola OE, Abrahams D, Estrella VC, Ordway B, Byrne SR, Ojeda AA, Whelan CJ, Kim J, Beatty MS, Damgaci-Erturk S, Longo DL, Gaspar KJ, Siegers GM, Centeno BA, Lau JYC, Pilon-Thomas SA, Ibrahim-Hashim A, Gillies RJ. L-DOS47 Elevates Pancreatic Cancer Tumor pH and Enhances Response to Immunotherapy. Biomedicines 2024; 12:461. [PMID: 38398062 PMCID: PMC10886509 DOI: 10.3390/biomedicines12020461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 01/17/2024] [Accepted: 01/25/2024] [Indexed: 02/25/2024] Open
Abstract
Acidosis is an important immunosuppressive mechanism that leads to tumor growth. Therefore, we investigated the neutralization of tumor acidity to improve immunotherapy response. L-DOS47, a new targeted urease immunoconjugate designed to neutralize tumor acidity, has been well tolerated in phase I/IIa trials. L-DOS47 binds to CEACAM6, a cell-surface protein that is highly expressed in gastrointestinal cancers, allowing urease to cleave endogenous urea into two NH4+ and one CO2, thereby raising local pH. To test the synergetic effect of neutralizing tumor acidity with immunotherapy, we developed a pancreatic orthotopic murine tumor model (KPC961) expressing human CEACAM6. Using chemical exchange saturation transfer-magnetic resonance imaging (CEST-MRI) to measure the tumor extracellular pH (pHe), we confirmed that L-DOS47 raises the tumor pHe from 4 h to 96 h post injection in acidic tumors (average increase of 0.13 units). Additional studies showed that combining L-DOS47 with anti-PD1 significantly increases the efficacy of the anti-PD1 monotherapy, reducing tumor growth for up to 4 weeks.
Collapse
Affiliation(s)
- Bruna Victorasso Jardim-Perassi
- Department of Metabolism and Physiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA (P.I.); (B.O.); (S.D.-E.)
| | - Pietro Irrera
- Department of Metabolism and Physiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA (P.I.); (B.O.); (S.D.-E.)
| | - Oluwaseyi E. Oluwatola
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA; (O.E.O.)
- Department of Molecular Medicine, University of South Florida, Tampa, FL 33620, USA
| | | | - Veronica C. Estrella
- Department of Metabolism and Physiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA (P.I.); (B.O.); (S.D.-E.)
| | - Bryce Ordway
- Department of Metabolism and Physiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA (P.I.); (B.O.); (S.D.-E.)
- Massachusetts General Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Samantha R. Byrne
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA; (O.E.O.)
| | - Andrew A. Ojeda
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA; (O.E.O.)
| | - Christopher J. Whelan
- Department of Metabolism and Physiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA (P.I.); (B.O.); (S.D.-E.)
- Department of Biological Sciences, University of Illinois, Chicago, IL 60607, USA
| | - Jongphil Kim
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Matthew S. Beatty
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA; (O.E.O.)
| | - Sultan Damgaci-Erturk
- Department of Metabolism and Physiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA (P.I.); (B.O.); (S.D.-E.)
| | - Dario Livio Longo
- Institute of Biostructures and Bioimaging (IBB), National Research Council of Italy (CNR), 10126 Turin, Italy
| | - Kim J. Gaspar
- Helix BioPharma Corp., Bay Adelaide Centre-North Tower, 40 Temperance Street, Suite 2700, Toronto, ON M5H 0B4, Canada
| | - Gabrielle M. Siegers
- Helix BioPharma Corp., Bay Adelaide Centre-North Tower, 40 Temperance Street, Suite 2700, Toronto, ON M5H 0B4, Canada
| | - Barbara A. Centeno
- Department of Pathology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Justin Y. C. Lau
- Small Animal Imaging Laboratory (SAIL), H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA;
| | - Shari A. Pilon-Thomas
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA; (O.E.O.)
| | - Arig Ibrahim-Hashim
- Department of Metabolism and Physiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA (P.I.); (B.O.); (S.D.-E.)
| | - Robert J. Gillies
- Department of Metabolism and Physiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA (P.I.); (B.O.); (S.D.-E.)
| |
Collapse
|
50
|
Bogdanov A, Verlov N, Bogdanov A, Burdakov V, Semiletov V, Egorenkov V, Volkov N, Moiseyenko V. Tumor alkalization therapy: misconception or good therapeutics perspective? - the case of malignant ascites. Front Oncol 2024; 14:1342802. [PMID: 38390269 PMCID: PMC10881708 DOI: 10.3389/fonc.2024.1342802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 01/24/2024] [Indexed: 02/24/2024] Open
Abstract
Tumor acidity has been identified as a key factor in promoting cancer progression, metastasis, and resistance. Tumor alkalization therapy has emerged as a potential strategy for cancer treatment. This article provides preclinical and clinical evidence for tumor alkalization therapy as a promising cancer treatment strategy. The potential of tumor alkalization therapy using sodium bicarbonate in the treatment of malignant ascites was studied. The concept of intraperitoneal perfusion with an alkalizing solution to increase the extracellular pH and its antitumor effect were explored. The significant extension in the overall survival of the Ehrlich ascites carcinoma mice treated with sodium bicarbonate solution compared to those treated with a sodium chloride solution was observed. In the sodium bicarbonate group, mice had a median survival of 30 days after tumor cell injection, which was significantly (p<0.05) different from the median survival of 18 days in the sodium chloride group and 14 days in the intact group. We also performed a case study of a patient with ovarian cancer malignant ascites resistant to previous lines of chemotherapy who underwent intraperitoneal perfusions with a sodium bicarbonate solution, resulting in a significant drop of CA-125 levels from 5600 U/mL to 2200 U/mL in and disappearance of ascites, indicating the potential effectiveness of the treatment. The preclinical and clinical results obtained using sodium bicarbonate perfusion in the treatment of malignant ascites represent a small yet significant contribution to the evolving field of tumor alkalization as a cancer therapy. They unequivocally affirm the good prospects of this concept.
Collapse
Affiliation(s)
- Alexey Bogdanov
- Napalkov Saint Petersburg Clinical Research and Practical Center of Specialized Types of Medical Care (Oncological), Saint Petersburg, Russia
| | | | | | | | | | | | | | | |
Collapse
|