1
|
Sharma R, Modi U, Kumar R, Sharma C, Srivastav AK, Bhatia D, Solanki R. Bio-inspired, programmable biomacromolecules based nanostructures driven cancer therapy. BIOMATERIALS ADVANCES 2025; 171:214235. [PMID: 39978287 DOI: 10.1016/j.bioadv.2025.214235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 01/28/2025] [Accepted: 02/13/2025] [Indexed: 02/22/2025]
Abstract
Cancer remains a significant global health challenge, driving the development of advanced platforms for highly targeted and efficient drug delivery. Early-stage nanocarriers, such as synthetic polymeric and inorganic materials, face limitations in biocompatibility and biodegradability. In contrast, bioinspired nanocarriers derived from natural biomacromolecules mimic biological processes and present a promising alternative due to their biocompatibility, biodegradability and non-toxicity. The effectiveness of these drug delivery systems is influenced by factors such as size, shape, surface properties, morphology, functionalization, and preparation methods. Various biomacromolecule-inspired nanocarriers such as protein-based, lipid-based, carbohydrate-based and nucleic acid-based are now at the forefront of research. This review highlights the properties, advantages and limitations of different bioinspired materials. We also explore cutting-edge approaches for cancer therapy using these nanocarriers with recent in-vitro, in-vivo and patent evidence. Finally, we address the challenges and potential solutions associated with bioinspired nanocarriers, proposing future directions. Overall, this review explores nature-inspired drug delivery systems that have paved the way for advancements in cancer therapy.
Collapse
Affiliation(s)
- Rahul Sharma
- Department of Biological Sciences and Engineering, Indian Institute of Technology Gandhinagar, Palaj, Gujarat 382355, India
| | - Unnati Modi
- Department of Biological Sciences and Engineering, Indian Institute of Technology Gandhinagar, Palaj, Gujarat 382355, India
| | - Rahul Kumar
- Dr. B. R. A. Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, India
| | - Chirag Sharma
- Department of Biological Sciences and Engineering, Indian Institute of Technology Gandhinagar, Palaj, Gujarat 382355, India
| | | | - Dhiraj Bhatia
- Department of Biological Sciences and Engineering, Indian Institute of Technology Gandhinagar, Palaj, Gujarat 382355, India.
| | - Raghu Solanki
- Department of Biological Sciences and Engineering, Indian Institute of Technology Gandhinagar, Palaj, Gujarat 382355, India.
| |
Collapse
|
2
|
Kicken MP, Deenen MJ, van der Wekken AJ, van den Borne BEEM, van den Heuvel MM, Ter Heine R. Opportunities for Precision Dosing of Cytotoxic Drugs in Non-Small Cell Lung Cancer: Bridging the Gap in Precision Medicine. Clin Pharmacokinet 2025; 64:511-531. [PMID: 40045151 PMCID: PMC12041064 DOI: 10.1007/s40262-025-01492-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/09/2025] [Indexed: 04/30/2025]
Abstract
Precision dosing of classical cytotoxic drugs in oncology remains underdeveloped, especially in treating non-small cell lung cancer (NSCLC). Despite advancements in targeted therapy and immunotherapy, classical cytotoxic agents continue to play a critical role in NSCLC treatment. However, the current body surface area (BSA)-based dosing of these agents fails to adequately address interindividual variability in pharmacokinetics. By better considering patient characteristics, treatment outcomes can be improved, reducing risks of under-exposure and over-exposure. This narrative review explores opportunities for precision dosing for key cytotoxic agents used in NSCLC treatment: cisplatin, carboplatin, pemetrexed, docetaxel, (nab-)paclitaxel, gemcitabine, and vinorelbine. A comprehensive review of regulatory reports and an extensive literature search were conducted to evaluate current dosing practices, pharmacokinetics, pharmacodynamics, and exposure-response relationships. Our findings highlight promising developments in precision dosing, although the number of directly implementable strategies remains limited. The most compelling evidence supports using the biomarker cystatin C for more precise carboplatin dosing and adopting weekly dosing schedules for docetaxel, paclitaxel, and nab-paclitaxel. Additionally, we recommend direct implementation of therapeutic drug monitoring (TDM)-guided dosing for paclitaxel. This review stresses the urgent need to reassess conventional dosing paradigms for classical cytotoxic agents to better align with the principles of the precision dosing framework. Our recommendations show the potential of precision dosing to improve NSCLC treatment, addressing gaps in the current dosing of classical cytotoxic drugs. Given the large NSCLC patient population, optimising the dosing of these agents could significantly improve treatment outcomes and reduce toxicity for many patients.
Collapse
Affiliation(s)
- M P Kicken
- Department of Clinical Pharmacy, Catharina Hospital, Michelangelolaan 2, 5623 EJ, Eindhoven, The Netherlands.
- Department of Pharmacy, Radboudumc, Research Institute for Medical Innovation, Nijmegen, The Netherlands.
| | - M J Deenen
- Department of Clinical Pharmacy, Catharina Hospital, Michelangelolaan 2, 5623 EJ, Eindhoven, The Netherlands
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Centre, Leiden, The Netherlands
| | - A J van der Wekken
- Department of Pulmonology and Tuberculosis, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| | | | - M M van den Heuvel
- Department of Pulmonology, Radboudumc, Research Institute for Medical Innovation, Nijmegen, The Netherlands
- Department of Pulmonology, University Medical Center, Utrecht, The Netherlands
| | - R Ter Heine
- Department of Pharmacy, Radboudumc, Research Institute for Medical Innovation, Nijmegen, The Netherlands
| |
Collapse
|
3
|
Linderman SW, DeRidder L, Sanjurjo L, Foote MB, Alonso MJ, Kirtane AR, Langer R, Traverso G. Enhancing immunotherapy with tumour-responsive nanomaterials. Nat Rev Clin Oncol 2025; 22:262-282. [PMID: 40050505 DOI: 10.1038/s41571-025-01000-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/05/2025] [Indexed: 03/09/2025]
Abstract
The targeted delivery of immunotherapies to tumours using tumour-responsive nanomaterials is a promising area of cancer research with the potential to address the limitations of systemic administration such as on-target off-tumour toxicities and a lack of activity owing to the immunosuppressive tumour microenvironment (TME). Attempts to address these challenges include the design and functionalization of nanomaterials capable of releasing their cargoes in response to specific TME characteristics, thus facilitating the targeted delivery of immune-checkpoint inhibitors, cytokines, mRNAs, vaccines and, potentially, chimaeric antigen receptors as well as of agents that modulate the extracellular matrix and induce immunogenic cell death. In this Review, we describe these various research efforts in the context of the dynamic properties of the TME, such as pH, reductive conditions, reactive oxygen species, hypoxia, specific enzymes, high levels of ATP and locoregional aspects, which can be leveraged to enhance the specificity and efficacy of nanomaterial-based immunotherapies. Highlighting preclinical successes and ongoing clinical trials, we evaluate the current landscape and potential of these innovative approaches. We also consider future research directions as well as the most important barriers to successful clinical translation, emphasizing the transformative potential of tumour-responsive nanomaterials in overcoming the barriers that limit the activity of traditional immunotherapies, thus improving patient outcomes.
Collapse
Affiliation(s)
- Stephen W Linderman
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Division of Hospital Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Louis DeRidder
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Harvard-MIT Division of Health Science Technology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Lucía Sanjurjo
- Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
- Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Michael B Foote
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - María José Alonso
- Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela, Santiago de Compostela, Spain
- Department of Pharmacy and Pharmaceutical Technology, University of Santiago de Compostela, Santiago de Compostela, Spain
- IMDEA Nanosciences Institute, Madrid, Spain
| | - Ameya R Kirtane
- Department of Pharmaceutics, University of Minnesota, Minneapolis, MN, USA
| | - Robert Langer
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
| | - Giovanni Traverso
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Division of Gastroenterology, Hepatology and Endoscopy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
4
|
Yang Y, Ye T, Shang F, Chen D, Wang K, He S. Combined Albumin Polyester Nanocarriers with Docetaxel for Effective Against Lung Cancer in Mice Model. Int J Nanomedicine 2025; 20:2103-2118. [PMID: 39990292 PMCID: PMC11844320 DOI: 10.2147/ijn.s487344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 01/23/2025] [Indexed: 02/25/2025] Open
Abstract
Introduction Lung cancer, a deadly malignancy, often employs Docetaxel (DTX) as a chemotherapy option. However, DTX non-selective distribution limits its therapeutic effectiveness due to adverse side effects. This study aims to develop novel folate-targeted albumin polyester nanocarriers (FA-DTX-APs) encapsulating DTX for precise delivery, enhancing lung cancer treatment efficacy. Methods FA-DTX-APs were meticulously crafted utilizing the thin-film dispersion technique and subsequently evaluated for their physicochemical characteristics, encapsulation efficiency, and drug release profiles. To assess their biological properties, anti-tumor efficacy, and biosafety in the context of lung cancer, a comprehensive series of hemolysis assays, cellular studies, and animal experiments were conducted. Results FA-DTX-APs exhibit nanovesicle properties with a size of (223.65 ± 6.83) nm, a potential of (26.76 ± 3.15) mV, and encapsulate DTX with high efficiency (96.19 ± 3.27%) and loading capacity (9.75 ± 0.38%). FA-DTX-APs enable tumor-targeted drug delivery and slow release of the drug over a long period of time, with faster release in acidic environments. By efficiently targeting and entering lung cancer cells, FA-DTX-APs effectively hinder cancer growth (P < 0.05), demonstrating superior anti-tumor effects (P < 0.05), biocompatibility and enhanced biological safety (P < 0.05). Conclusion This study introduces FA-DTX-APs, an innovative nanocarrier characterized by exceptional biocompatibility and safety. It successfully targets lung cancer cells to deliver DTX in a sustained, slow-release manner, ensuring prolonged tumor-killing effects. As such, FA-DTX-APs hold immense promise as a novel nanoagent for lung cancer therapy.
Collapse
Affiliation(s)
- Yixiao Yang
- Research Center of Nanomedicine Technology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530000, People’s Republic of China
| | - Tao Ye
- Department of Oncology, Minhang Branch, Zhongshan Hospital, Fudan University, Shanghai, 201100, People’s Republic of China
| | - Fusheng Shang
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, People’s Republic of China
| | - Dagui Chen
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, People’s Republic of China
| | - Kai Wang
- Research Center of Nanomedicine Technology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530000, People’s Republic of China
| | - Shengli He
- Department of Hepatobiliary-Pancreatic and Integrative Oncology, Minhang Branch, Fudan University Shanghai Cancer Center, Shanghai, 200240, People’s Republic of China
| |
Collapse
|
5
|
Zhang Y, Lian Y, Zhou C, Cheng J, Zhao S, Liu H, Wang J, Lu X, Shi J, Du G. Self-assembled natural triterpenoids for the delivery of cyclin-dependent kinase 4/6 inhibitors to enhance cancer chemoimmunotherapy. J Control Release 2025; 378:791-802. [PMID: 39732370 DOI: 10.1016/j.jconrel.2024.12.067] [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: 09/01/2024] [Revised: 12/22/2024] [Accepted: 12/25/2024] [Indexed: 12/30/2024]
Abstract
Immunogenic cell death (ICD) has recently emerged as a promising strategy in reinforcing anti-PD-L1 blockade immunotherapy of triple-negative breast cancer (TNBC). The CDK4/6 inhibitor palbociclib (PAL), as a clinical star medicine targeting the cell cycle machinery, is an ideal candidate for fabricating a highly efficient ICD inducer for TNBC chemoimmunotherapy. However, the frequently observed chemoresistance and clinical adverse effects, as well as significant antagonistic effects when co-administered with certain chemotherapeutics, have seriously restricted the efficiency of PAL and the feasibility of combination strategies. Herein, we screened and identified six self-assembled natural pentacyclic triterpenoid (PT) molecules that can serve as competent co-administration nanoplatforms for the synergistic or combined delivery of PAL. Analysis of two representative PT-PAL nano-assemblies validated that PT-mediated co-assembly enhances the cytotoxicity and synergy of PAL by inhibiting the PI3K/AKT/mTOR signaling pathway, rather than directly targeting CDK4/6 proteins. Importantly, the PAL nanoassemblies exhibited multiple favorable therapeutic features and stronger accumulative ICD induction, ensuring highly efficient synergistic anti-PD-L1 chemoimmunotherapy by simultaneously facilitating T-cell immune response and reversing the immunosuppressive tumor microenvironment. This study offers possibilities for improving the anticancer efficacy of CDK4/6 inhibitors and potential avenues for clinical applications of chemoimmunotherapy in treating TNBC.
Collapse
Affiliation(s)
- Yongbo Zhang
- Key Laboratory of Natural Medicine Innovation and Transformation, Henan University, Kaifeng 475000, PR China
| | - Yajie Lian
- Key Laboratory of Natural Medicine Innovation and Transformation, Henan University, Kaifeng 475000, PR China
| | - Conglei Zhou
- Key Laboratory of Natural Medicine Innovation and Transformation, Henan University, Kaifeng 475000, PR China
| | - Jianjun Cheng
- Key Laboratory of Natural Medicine Innovation and Transformation, Henan University, Kaifeng 475000, PR China; State Key Laboratory of Antiviral Drugs, Henan University, Kaifeng 475000, PR China.
| | - Shuang Zhao
- Key Laboratory of Natural Medicine Innovation and Transformation, Henan University, Kaifeng 475000, PR China; State Key Laboratory of Antiviral Drugs, Henan University, Kaifeng 475000, PR China
| | - Hongjun Liu
- Key Laboratory of Natural Medicine Innovation and Transformation, Henan University, Kaifeng 475000, PR China; State Key Laboratory of Antiviral Drugs, Henan University, Kaifeng 475000, PR China
| | - Jiacheng Wang
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225001, PR China
| | - Xin Lu
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225001, PR China
| | - Jiahua Shi
- Key Laboratory of Natural Medicine Innovation and Transformation, Henan University, Kaifeng 475000, PR China; State Key Laboratory of Antiviral Drugs, Henan University, Kaifeng 475000, PR China.
| | - Guanhua Du
- Key Laboratory of Natural Medicine Innovation and Transformation, Henan University, Kaifeng 475000, PR China; State Key Laboratory of Antiviral Drugs, Henan University, Kaifeng 475000, PR China; Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, PR China.
| |
Collapse
|
6
|
Ye L, Meng X, Zhan Y, Li T, Huang X, Qiu H, Zhou J, Guo C. Research on genetic variant characteristics in ADME genes based on whole-exome sequencing in the Han Chinese population. Eur J Pharm Sci 2025; 205:106987. [PMID: 39672213 DOI: 10.1016/j.ejps.2024.106987] [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: 08/22/2024] [Revised: 12/08/2024] [Accepted: 12/09/2024] [Indexed: 12/15/2024]
Abstract
BACKGROUND Genetic variants in absorption, distribution, metabolism, and excretion (ADME) genes affect drug efficacy and side effects. Whole-exome sequencing (WES) effectively identifies these variants. Findings from Western populations may not apply to the Han Chinese population due to ethnic differences. OBJECTIVE This study aimed to investigate genetic variants, metabolic phenotypes, and drug impacts related to ADME genes in the Han Chinese population using WES data. METHODS ADME genes and drug profiles were sourced from multiple databases. WES data were collected from 357 samples at Third Xiangya Hospital and 5,000 samples from the "HuaBiao Project." After WES, fastp was used for quality control; BWA, samtools, and the Picard software were used for comparison, sequencing, and de-duplication; GATK was used for base quality score recalibration; and variant quality score recalibration was carried out after detecting the variants, and the variants were annotated using ANNOVAR. ADME genes and drug profiles were obtained through PharmGKB and other databases, and then all variants in the exonic regions of ADME genes were extracted. The distributional characteristics of these variants, ethnic differences, and metabolic phenotype distribution of important ADME genes, such as CYP2B6, were analyzed. Prediction of deleterious variants of ADME gene employed the ADME gene variant prediction framework, and western blotting and enzyme assays were used to validate the impact of harmful variants. RESULTS We integrated 604 ADME genes and 972 drugs. There were 33,925 single nucleotide polymorphisms (SNPs) and 484 insertion-deletions (InDels) in 5,357 Han Chinese WES samples; 88.9 % were rare. Of the SNPs, 60.9 % are new functional mutations in enzyme and transporter genes; InDels also affected these genes. We discovered Chinese-specific variants in key ADME genes and ethnic-specific metabolic phenotypes that could affect drug use. Finally, we screened 3,657 potentially harmful mutations; 45.6 % are novel. Western blotting and enzyme activity assays confirmed several harmful mutations significantly reduced CYP2C19 gene expression and function (P < 0.01).
Collapse
Affiliation(s)
- Ling Ye
- Department of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Xiangya Hospital, Central South University, China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, China; Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, China; National Clinical Research Center for Geriatric Disorders, China
| | - XiangGuang Meng
- Department of Pharmacy, Zhengzhou No. 7 People's Hospital, China
| | - Yan Zhan
- Department of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Xiangya Hospital, Central South University, China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, China; Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, China; National Clinical Research Center for Geriatric Disorders, China
| | - Tong Li
- Center of Clinical Pharmacology, the Third Xiangya Hospital, Central South University, China
| | - Xin Huang
- Center of Clinical Pharmacology, the Third Xiangya Hospital, Central South University, China
| | - Hui Qiu
- Center of Clinical Pharmacology, the Third Xiangya Hospital, Central South University, China
| | - Jianzhu Zhou
- Center of Clinical Pharmacology, the Third Xiangya Hospital, Central South University, China
| | - Chengxian Guo
- Center of Clinical Pharmacology, the Third Xiangya Hospital, Central South University, China.
| |
Collapse
|
7
|
Guarín-González YA, Cabello-Guzmán G, Reyes-Gasga J, Moreno-Navarro Y, Vergara-González L, Martin-Martín A, López-Muñoz R, Cárdenas-Triviño G, Barraza LF. Dual-Action Gemcitabine Delivery: Chitosan-Magnetite-Zeolite Capsules for Targeted Cancer Therapy and Antibacterial Defense. Gels 2024; 10:672. [PMID: 39451325 PMCID: PMC11507657 DOI: 10.3390/gels10100672] [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: 09/09/2024] [Revised: 10/02/2024] [Accepted: 10/08/2024] [Indexed: 10/26/2024] Open
Abstract
Cancer and infectious diseases are two of the world's major public health problems. Gemcitabine (GEM) is an effective chemotherapeutic agent against several types of cancer. In this study, we developed macrocapsules incorporating GEM into a chitosan matrix blended with magnetite and zeolite by ionic gelation. Physicochemical characterization was performed using HRTEM-ED, XRD, FESEM-EDS, FT-IR, TGA, encapsulation efficiency (%E.E.), and release profiles at pHs 7.4 and 5.0. Cell viability tests against A549 and H1299 cell lines, and microbiological properties against staphylococcal strains were performed. Our results revealed the successful production of hemispherical capsules with an average diameter of 1.22 mm, a rough surface, and characteristic FT-IR material interaction bands. The macrocapsules showed a high GEM encapsulation efficiency of over 86% and controlled release over 24 h. Cell viability assays revealed that similar cytotoxic effects to free GEM were achieved with a 45-fold lower GEM concentration, suggesting reduced dosing requirements and potentially fewer side effects. Additionally, the macrocapsules demonstrated potent antimicrobial activity, reducing Staphylococcus epidermidis growth by over 90%. These results highlight the macrocapsules dual role as a chemotherapeutic and antimicrobial agent, offering a promising strategy for treating lung cancer in patients at risk of infectious diseases or who are immunosuppressed.
Collapse
Affiliation(s)
- Yuly Andrea Guarín-González
- Laboratorio Térmico de Nano y Macromateriales, Edificio de Procesos Sustentables, Departamento de Ingeniería en Maderas, Universidad del Bío-Bío, Concepción 4081112, Chile;
| | - Gerardo Cabello-Guzmán
- Facultad de Ciencias, Departamento de Biología y Química, Universidad del Bío-Bío, Chillán 3780000, Chile;
| | - José Reyes-Gasga
- Instituto de Física, Departamento de Materia Condensada, Universidad Autónoma de México, Coyoacán 04510, Mexico;
| | - Yanko Moreno-Navarro
- Facultad de Ciencias, Departamento de Química, Universidad de la Serena, La Serena 1720170, Chile;
| | - Luis Vergara-González
- Departamento de Ciencias Biológicas y Químicas, Facultad de Medicina y Ciencia, Universidad San Sebastián, Campus Las Tres Pascualas, Lientur 1457, Concepción 4060000, Chile;
| | - Antonia Martin-Martín
- Instituto de Farmacología y Morfofisiología, Facultad de Ciencias Veterinarias, Universidad Austral de Chile, Valdivia 5090000, Chile; (A.M.-M.); (R.L.-M.)
| | - Rodrigo López-Muñoz
- Instituto de Farmacología y Morfofisiología, Facultad de Ciencias Veterinarias, Universidad Austral de Chile, Valdivia 5090000, Chile; (A.M.-M.); (R.L.-M.)
| | - Galo Cárdenas-Triviño
- Laboratorio Térmico de Nano y Macromateriales, Edificio de Procesos Sustentables, Departamento de Ingeniería en Maderas, Universidad del Bío-Bío, Concepción 4081112, Chile;
| | - Luis F. Barraza
- Departamento de Ciencias Biológicas y Químicas, Facultad de Medicina y Ciencia, Universidad San Sebastián, General Lagos 1163, Valdivia 5090000, Chile
| |
Collapse
|
8
|
DeRidder LB, Hare KA, Lopes A, Jenkins J, Fitzgerald N, MacPherson E, Fabian N, Morimoto J, Chu JN, Kirtane AR, Madani W, Ishida K, Kuosmanen JLP, Zecharias N, Colangelo CM, Huang HW, Chilekwa M, Lal NB, Srinivasan SS, Hayward AM, Wolpin BM, Trumper D, Quast T, Rubinson DA, Langer R, Traverso G. Closed-loop automated drug infusion regulator: A clinically translatable, closed-loop drug delivery system for personalized drug dosing. MED 2024; 5:780-796.e10. [PMID: 38663403 DOI: 10.1016/j.medj.2024.03.020] [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: 10/27/2023] [Revised: 01/26/2024] [Accepted: 03/21/2024] [Indexed: 07/15/2024]
Abstract
BACKGROUND Dosing of chemotherapies is often calculated according to the weight and/or height of the patient or equations derived from these, such as body surface area (BSA). Such calculations fail to capture intra- and interindividual pharmacokinetic variation, which can lead to order of magnitude variations in systemic chemotherapy levels and thus under- or overdosing of patients. METHODS We designed and developed a closed-loop drug delivery system that can dynamically adjust its infusion rate to the patient to reach and maintain the drug's target concentration, regardless of a patient's pharmacokinetics (PK). FINDINGS We demonstrate that closed-loop automated drug infusion regulator (CLAUDIA) can control the concentration of 5-fluorouracil (5-FU) in rabbits according to a range of concentration-time profiles (which could be useful in chronomodulated chemotherapy) and over a range of PK conditions that mimic the PK variability observed clinically. In one set of experiments, BSA-based dosing resulted in a concentration 7 times above the target range, while CLAUDIA keeps the concentration of 5-FU in or near the targeted range. Further, we demonstrate that CLAUDIA is cost effective compared to BSA-based dosing. CONCLUSIONS We anticipate that CLAUDIA could be rapidly translated to the clinic to enable physicians to control the plasma concentration of chemotherapy in their patients. FUNDING This work was supported by MIT's Karl van Tassel (1925) Career Development Professorship and Department of Mechanical Engineering and the Bridge Project, a partnership between the Koch Institute for Integrative Cancer Research at MIT and the Dana-Farber/Harvard Cancer Center.
Collapse
Affiliation(s)
- Louis B DeRidder
- Harvard-MIT Division of Health Science Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Division of Gastroenterology, Hepatology, and Endoscopy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Kyle A Hare
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Aaron Lopes
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Division of Gastroenterology, Hepatology, and Endoscopy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Josh Jenkins
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Nina Fitzgerald
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Division of Gastroenterology, Hepatology, and Endoscopy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Emmeline MacPherson
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Niora Fabian
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Josh Morimoto
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Division of Gastroenterology, Hepatology, and Endoscopy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Jacqueline N Chu
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Harvard Medical School, Boston, MA 02115, USA; Division of Gastroenterology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Ameya R Kirtane
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Division of Gastroenterology, Hepatology, and Endoscopy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Wiam Madani
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Keiko Ishida
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Division of Gastroenterology, Hepatology, and Endoscopy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Johannes L P Kuosmanen
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Naomi Zecharias
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | | | - Hen-Wei Huang
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Division of Gastroenterology, Hepatology, and Endoscopy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Makaya Chilekwa
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Nikhil B Lal
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Division of Gastroenterology, Hepatology, and Endoscopy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA; MIT Media Lab, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Shriya S Srinivasan
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Division of Gastroenterology, Hepatology, and Endoscopy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Alison M Hayward
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Division of Gastroenterology, Hepatology, and Endoscopy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA; Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Brian M Wolpin
- Harvard Medical School, Boston, MA 02115, USA; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - David Trumper
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Troy Quast
- College of Public Health, University of South Florida, Tampa, FL 33612, USA
| | - Douglas A Rubinson
- Harvard Medical School, Boston, MA 02115, USA; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Robert Langer
- Harvard-MIT Division of Health Science Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Surgery, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Giovanni Traverso
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Division of Gastroenterology, Hepatology, and Endoscopy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
9
|
Adams SC, Nambiar AK, Bressler EM, Raut CP, Colson YL, Wong WW, Grinstaff MW. Immunotherapies for locally aggressive cancers. Adv Drug Deliv Rev 2024; 210:115331. [PMID: 38729264 PMCID: PMC11228555 DOI: 10.1016/j.addr.2024.115331] [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: 02/06/2024] [Revised: 03/31/2024] [Accepted: 05/06/2024] [Indexed: 05/12/2024]
Abstract
Improving surgical resection outcomes for locally aggressive tumors is key to inducing durable locoregional disease control and preventing progression to metastatic disease. Macroscopically complete resection of the tumor is the standard of care for many cancers, including breast, ovarian, lung, sarcoma, and mesothelioma. Advancements in cancer diagnostics are increasing the number of surgically eligible cases through early detection. Thus, a unique opportunity arises to improve patient outcomes with decreased recurrence rates via intraoperative delivery treatments using local drug delivery strategies after the tumor has been resected. Of the current systemic treatments (e.g., chemotherapy, targeted therapies, and immunotherapies), immunotherapies are the latest approach to offer significant benefits. Intraoperative strategies benefit from direct access to the tumor microenvironment which improves drug uptake to the tumor and simultaneously minimizes the risk of drug entering healthy tissues thereby resulting in fewer or less toxic adverse events. We review the current state of immunotherapy development and discuss the opportunities that intraoperative treatment provides. We conclude by summarizing progress in current research, identifying areas for exploration, and discussing future prospects in sustained remission.
Collapse
Affiliation(s)
- Sarah C Adams
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - Arun K Nambiar
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - Eric M Bressler
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - Chandrajit P Raut
- Department of Surgery, Brigham and Women's Hospital, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Yolonda L Colson
- Massachusetts General Hospital, Department of Surgery, Boston, MA 02114, USA.
| | - Wilson W Wong
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA.
| | - Mark W Grinstaff
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA; Department of Chemistry, Boston University, Boston MA 02215, USA.
| |
Collapse
|
10
|
Chen H, Xing C, Lei H, Yan B, Zhang H, Tong T, Guan Y, Kang Y, Pang J. ROS-driven supramolecular nanoparticles exhibiting efficient drug delivery for chemo/Chemodynamic combination therapy for Cancer treatment. J Control Release 2024; 368:637-649. [PMID: 38484895 DOI: 10.1016/j.jconrel.2024.03.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Revised: 02/26/2024] [Accepted: 03/11/2024] [Indexed: 03/21/2024]
Abstract
Drug-based supramolecular self-assembling delivery systems have enhanced the bioavailability of chemotherapeutic drugs and reduced systemic side effects; however, improving the delivery efficiency and responsive release ability of these systems remains challenging. This study focuses primarily on the utilization of per-6-thio-β-cyclodextrin (CD) to link a significant quantity of paclitaxel (PTX) via ROS-sensitive thioketal (TK) linkages (designated as CDTP), thereby allowing efficiently drug release when exposed to high levels of reactive oxygen species (ROS) in the tumor microenvironment. To construct these supramolecular nanoparticles (NPs) with CDTP, we introduced PEGylated ferrocene (Fc) through host-guest interactions. The intracellular hydrogen peroxide (H2O2) is converted into hydroxyl radicals (•OH) through the Fc-catalyzed Fenton reaction. Additionally, the generated Fc+ consumes the antioxidant glutathione (GSH). In both in vivo and in vitro experiments, CDTP@Fc-PEG NPs were absorbed effectively by tumor cells, which increased levels of ROS and decreased levels of GSH, disrupting the redox balance of cancer cells and increasing their sensitivity to chemotherapy. Furthermore, CDTP@Fc-PEG NPs exhibited high tumor accumulation and cytotoxicity without causing significant toxicity to healthy organs. Collectively, our results suggest CDTP@Fc-PEG NPs as a promising supramolecular nano-delivery platform for high drug-loading of PTX and synergistic chemotherapy.
Collapse
Affiliation(s)
- Huikun Chen
- Department of Urology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China; Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China
| | - Chengyuan Xing
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China
| | - Hanqi Lei
- Department of Urology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China
| | - Binyuan Yan
- Department of Urology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China
| | - Hao Zhang
- Department of Urology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China; Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China
| | - Tongyu Tong
- Department of Urology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China; Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China
| | - Yupeng Guan
- Department of Urology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China; Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China
| | - Yang Kang
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China.
| | - Jun Pang
- Department of Urology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China.
| |
Collapse
|
11
|
Khan SU, Fatima K, Aisha S, Malik F. Unveiling the mechanisms and challenges of cancer drug resistance. Cell Commun Signal 2024; 22:109. [PMID: 38347575 PMCID: PMC10860306 DOI: 10.1186/s12964-023-01302-1] [Citation(s) in RCA: 81] [Impact Index Per Article: 81.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Accepted: 08/30/2023] [Indexed: 02/15/2024] Open
Abstract
Cancer treatment faces many hurdles and resistance is one among them. Anti-cancer treatment strategies are evolving due to innate and acquired resistance capacity, governed by genetic, epigenetic, proteomic, metabolic, or microenvironmental cues that ultimately enable selected cancer cells to survive and progress under unfavorable conditions. Although the mechanism of drug resistance is being widely studied to generate new target-based drugs with better potency than existing ones. However, due to the broader flexibility in acquired drug resistance, advanced therapeutic options with better efficacy need to be explored. Combination therapy is an alternative with a better success rate though the risk of amplified side effects is commonplace. Moreover, recent groundbreaking precision immune therapy is one of the ways to overcome drug resistance and has revolutionized anticancer therapy to a greater extent with the only limitation of being individual-specific and needs further attention. This review will focus on the challenges and strategies opted by cancer cells to withstand the current therapies at the molecular level and also highlights the emerging therapeutic options -like immunological, and stem cell-based options that may prove to have better potential to challenge the existing problem of therapy resistance. Video Abstract.
Collapse
Affiliation(s)
- Sameer Ullah Khan
- Division of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Holcombe Blvd, Houston, TX, 77030, USA.
- Division of Cancer Pharmacology, CSIR-Indian Institute of Integrative Medicine, Srinagar-190005, Jammu and Kashmir, India.
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad-201002, India.
| | - Kaneez Fatima
- Division of Cancer Pharmacology, CSIR-Indian Institute of Integrative Medicine, Srinagar-190005, Jammu and Kashmir, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad-201002, India
| | - Shariqa Aisha
- Division of Cancer Pharmacology, CSIR-Indian Institute of Integrative Medicine, Srinagar-190005, Jammu and Kashmir, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad-201002, India
| | - Fayaz Malik
- Division of Cancer Pharmacology, CSIR-Indian Institute of Integrative Medicine, Srinagar-190005, Jammu and Kashmir, India.
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad-201002, India.
| |
Collapse
|
12
|
Zou F, Du Q, Zhang Y, Zuo L, Sun Z. Pseudo-allergic reactions induced by Chinese medicine injections: a review. Chin Med 2023; 18:149. [PMID: 37953288 PMCID: PMC10642014 DOI: 10.1186/s13020-023-00855-0] [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: 09/15/2023] [Accepted: 10/23/2023] [Indexed: 11/14/2023] Open
Abstract
Traditional Chinese medicine injections (TCMIs) is a new dosage form of Chinese medicine, which plays a unique role in rescuing patients with critical illnesses that are difficult to replace. With the rapid development and widespread application of TCMIs in recent years, their adverse events have emerged and attracted much attention. Among them, pseudo-allergic reactions, i.e., the most significant adverse reactions occurring with the first dose without immunoglobulin E mediated conditions. Currently, studies on the types of TCMIs and antibiotic mechanisms that cause pseudo-allergic reactions are incomplete, and standard models and technical guidelines for assessing TCMIs have not been established. First, this review describes the causes of pseudo-allergic reactions, in which the components and structures responsible for pseudo-allergic reactions are summarized. Second, the mechanisms by which pseudo-allergic reactions are discussed, including direct stimulation of mast cells and complement activation. Then, research models of pseudo-allergic reaction diseases are reviewed, including animal models and cellular models. Finally, the outlook and future challenges for the development of pseudo-allergic reactions in traditional Chinese medicine (TCM) are outlined. This shed new light on the assessment and risk prevention of pseudo-allergic reactions in TCM and the prevention of clinical adverse reactions in TCM.
Collapse
Affiliation(s)
- Fanmei Zou
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan Zhengzhou, China
- Henan Engineering Research Center of Clinical Mass Spectrometry for Precision Medicine, Zhengzhou, 450052, Henan Zhengzhou, China
| | - Qiuzheng Du
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan Zhengzhou, China
- Henan Engineering Research Center of Clinical Mass Spectrometry for Precision Medicine, Zhengzhou, 450052, Henan Zhengzhou, China
| | - Yuanyuan Zhang
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan Zhengzhou, China
- Henan Engineering Research Center of Clinical Mass Spectrometry for Precision Medicine, Zhengzhou, 450052, Henan Zhengzhou, China
| | - Lihua Zuo
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan Zhengzhou, China
- Henan Engineering Research Center of Clinical Mass Spectrometry for Precision Medicine, Zhengzhou, 450052, Henan Zhengzhou, China
| | - Zhi Sun
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan Zhengzhou, China.
- Henan Engineering Research Center of Clinical Mass Spectrometry for Precision Medicine, Zhengzhou, 450052, Henan Zhengzhou, China.
| |
Collapse
|
13
|
Jin H, Liu X, Liu HX. Biological function, regulatory mechanism, and clinical application of mannose in cancer. Biochim Biophys Acta Rev Cancer 2023; 1878:188970. [PMID: 37657682 DOI: 10.1016/j.bbcan.2023.188970] [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: 04/19/2023] [Revised: 08/15/2023] [Accepted: 08/15/2023] [Indexed: 09/03/2023]
Abstract
Studies examining the regulatory roles and clinical applications of monosaccharides other than glucose in cancer have been neglected. Mannose, a common type of monosaccharide found in human body fluids and tissues, primarily functions in protein glycosylation rather than carbohydrate metabolism. Recent research has demonstrated direct anticancer effects of mannose in vitro and in vivo. Simply supplementing cell culture medium or drinking water with mannose achieved these effects. Moreover, mannose enhances the effectiveness of current cancer treatments including chemotherapy, radiotherapy, targeted therapy, and immune therapy. Besides the advancements in basic research on the anticancer effects of mannose, recent studies have reported its application as a biomarker for cancer or in the delivery of anticancer drugs using mannose-modified drug delivery systems. This review discusses the progress made in understanding the regulatory roles of mannose in cancer progression, the mechanisms underlying its anticancer effects, and its current application in cancer diagnosis and treatment.
Collapse
Affiliation(s)
- Haoyi Jin
- Department of Thoracic Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, 110042, Liaoning, China
| | - Xi Liu
- Department of Urology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, 110042, Liaoning, China
| | - Hong-Xu Liu
- Department of Thoracic Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, 110042, Liaoning, China; Department of Urology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, 110042, Liaoning, China.
| |
Collapse
|
14
|
Gou S, Wang G, Zou Y, Geng W, He T, Qin Z, Che L, Feng Q, Cai K. Non-Pore Dependent and MMP-9 Responsive Gelatin/Silk Fibroin Composite Microparticles as Universal Delivery Platform for Inhaled Treatment of Lung Cancer. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2303718. [PMID: 37625141 DOI: 10.1002/adma.202303718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 08/15/2023] [Indexed: 08/27/2023]
Abstract
Developing a drug delivery platform that possesses universal drug loading capacity to meet various requirements of cancer treatment is a challenging yet interesting task. Herein, a self-assembled gelatin/silk fibroin composite (GSC) particle based drug delivery system is developed via microphase separation followed by desolvation process. Thanks to its preassembled microphase stage, this GSC system is suitable for varying types of drugs. The desolvation process fix drugs inside GSC rapidly and densify the GSC structure, thereby achieving efficient drug loading and providing comprehensive protection for loaded drugs. Actually, the size of this brand-new non-pore dependent drug delivery system can be easily adjusted from 100 nm to 20 µm to fit different scenarios. This work selects GSC with 3 µm diameter as the universal inhaled drug delivery platform, which shows an excellent transmucosal penetration and lung retention ability. Additionally, the MMP-9 sensitive degradation property of GSC enhances the targeted efficiency of drugs and reduces side effects. Intestinally, GSC can self-amplify the regulation of innate immunity to reverse the cancerous microenvironment into an antitumor niche, significantly improving the therapeutic effect of drugs. This study of GSC universal drug platform provides a new direction to develop the next-generation of drug delivery system for lung cancer.
Collapse
Affiliation(s)
- Shuangquan Gou
- Key laboratory of Biorheological Science and Technology, Ministry of Educations, Collage of Bioengineering, Chongqing University, Chongqing, 40044, China
| | - Gangyang Wang
- Department of Orthopedic, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 200080, Shanghai, China
| | - Yanan Zou
- Key laboratory of Biorheological Science and Technology, Ministry of Educations, Collage of Bioengineering, Chongqing University, Chongqing, 40044, China
| | - Wenbo Geng
- Key laboratory of Biorheological Science and Technology, Ministry of Educations, Collage of Bioengineering, Chongqing University, Chongqing, 40044, China
| | - Tingting He
- Key laboratory of Biorheological Science and Technology, Ministry of Educations, Collage of Bioengineering, Chongqing University, Chongqing, 40044, China
| | - Zizhen Qin
- Key laboratory of Biorheological Science and Technology, Ministry of Educations, Collage of Bioengineering, Chongqing University, Chongqing, 40044, China
| | - Lingbin Che
- Department of Orthopedic, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 200080, Shanghai, China
| | - Qian Feng
- Key laboratory of Biorheological Science and Technology, Ministry of Educations, Collage of Bioengineering, Chongqing University, Chongqing, 40044, China
| | - Kaiyong Cai
- Key laboratory of Biorheological Science and Technology, Ministry of Educations, Collage of Bioengineering, Chongqing University, Chongqing, 40044, China
| |
Collapse
|
15
|
Sripetthong S, Eze FN, Sajomsang W, Ovatlarnporn C. Development of pH-Responsive N-benzyl- N- O-succinyl Chitosan Micelles Loaded with a Curcumin Analog (Cyqualone) for Treatment of Colon Cancer. Molecules 2023; 28:2693. [PMID: 36985665 PMCID: PMC10057334 DOI: 10.3390/molecules28062693] [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: 11/17/2022] [Revised: 02/19/2023] [Accepted: 02/25/2023] [Indexed: 03/19/2023] Open
Abstract
This work aimed at preparing nanomicelles from N-benzyl-N,O-succinyl chitosan (NBSCh) loaded with a curcumin analog, 2,6-bis((3-methoxy-4-hydroxyphenyl) methylene) cyclohexanone, a.k.a. cyqualone (CL), for antineoplastic colon cancer chemotherapy. The CL-loaded NBSCh micelles were spherical and less than 100 nm in size. The entrapment efficiency of CL in the micelles ranged from 13 to 39%. Drug release from pristine CL was less than 20% in PBS at pH 7.4, whereas the release from CL-NBSCh micelles was significantly higher. The release study of CL-NBSCh revealed that around 40% of CL content was released in simulated gastric fluid at pH 1.2; 79 and 85% in simulated intestinal fluids at pH 5.5 and 6.8, respectively; and 75% in simulated colonic fluid at pH 7.4. CL-NBSCh showed considerably high selective cytotoxicity towards mucosal epithelial human colon cancer (HT-29) cells and lower levels of toxicity towards mouse connective tissue fibroblasts (L929). CL-NBSCh was also more cytotoxic than the free CL. Furthermore, compared to free CL, CL-NBSCh micelles were found to be more efficient at arresting cell growth at the G2/M phase, and induced apoptosis earlier in HT-29 cells. Collectively, these results indicate the high prospective potential of CL-loaded NBSCh micelles as an oral therapeutic intervention for colon cancer.
Collapse
Affiliation(s)
- Sasikarn Sripetthong
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Prince of Songkla University, Hat Yai 90112, Songkhla, Thailand; (S.S.); (F.N.E.)
- Drug Delivery System Excellence Center, Faculty of Pharmaceutical Sciences, Prince of Songkla University, Hat Yai 90112, Songkhla, Thailand
| | - Fredrick Nwude Eze
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Prince of Songkla University, Hat Yai 90112, Songkhla, Thailand; (S.S.); (F.N.E.)
- Drug Delivery System Excellence Center, Faculty of Pharmaceutical Sciences, Prince of Songkla University, Hat Yai 90112, Songkhla, Thailand
| | - Warayuth Sajomsang
- Nanodelivery System Laboratory, National Nanotechnology Center, National Science and Technology Development Agency, Phathum Thani 12120, Thailand;
| | - Chitchamai Ovatlarnporn
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Prince of Songkla University, Hat Yai 90112, Songkhla, Thailand; (S.S.); (F.N.E.)
- Drug Delivery System Excellence Center, Faculty of Pharmaceutical Sciences, Prince of Songkla University, Hat Yai 90112, Songkhla, Thailand
| |
Collapse
|
16
|
The First-In-Class Anti-AXL×CD3ε Pronectin™-Based Bispecific T-Cell Engager Is Active in Preclinical Models of Human Soft Tissue and Bone Sarcomas. Cancers (Basel) 2023; 15:cancers15061647. [PMID: 36980534 PMCID: PMC10046451 DOI: 10.3390/cancers15061647] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 03/03/2023] [Accepted: 03/05/2023] [Indexed: 03/11/2023] Open
Abstract
Sarcomas are heterogeneous malignancies with limited therapeutic options and a poor prognosis. We developed an innovative immunotherapeutic agent, a first-in-class Pronectin™-based Bispecific T-Cell Engager (pAXL×CD3ε), for the targeting of AXL, a TAM family tyrosine kinase receptor highly expressed in sarcomas. AXL expression was first analyzed by flow cytometry, qRT-PCR, and Western blot on a panel of sarcoma cell lines. The T-cell-mediated pAXL×CD3ε cytotoxicity against sarcoma cells was investigated by flow cytometry, luminescence assay, and fluorescent microscopy imaging. The activation and degranulation of T cells induced by pAXL×CD3ε were evaluated by flow cytometry. The antitumor activity induced by pAXL×CD3ε in combination with trabectedin was also investigated. In vivo activity studies of pAXL×CD3ε were performed in immunocompromised mice (NSG), engrafted with human sarcoma cells and reconstituted with human peripheral blood mononuclear cells from healthy donors. Most sarcoma cells showed high expression of AXL. pAXL×CD3ε triggered T-lymphocyte activation and induced dose-dependent T-cell-mediated cytotoxicity. The combination of pAXL×CD3ε with trabectedin increased cytotoxicity. pAXL×CD3ε inhibited the in vivo growth of human sarcoma xenografts, increasing the survival of treated mice. Our data demonstrate the antitumor efficacy of pAXL×CD3ε against sarcoma cells, providing a translational framework for the clinical development of pAXL×CD3ε in the treatment of human sarcomas, aggressive and still-incurable malignancies.
Collapse
|