1
|
Xu L, Schaefer KG, King GM, Xie ZR, Bartlett MG. Insights into interactions between taxanes and P-glycoprotein using biophysical and in silico methods. J Pharm Sci 2025; 114:103708. [PMID: 40015511 DOI: 10.1016/j.xphs.2025.103708] [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: 11/11/2024] [Revised: 02/20/2025] [Accepted: 02/20/2025] [Indexed: 03/01/2025]
Abstract
Multidrug resistance mediated by P-glycoprotein (Pgp) is a significant obstacle to cancer chemotherapy. Taxane drugs, including paclitaxel, docetaxel, and cabazitaxel, are used to treat multiple types of cancer. All taxane drugs are Pgp substrates, but cabazitaxel is also a Pgp inhibitor, indicating potential differential interactions between Pgp and different taxanes. Here, we showed for the first time that cabazitaxel had a partial inhibitory effect on the ATPase activity at concentrations higher than 10 µM. We found the KD of paclitaxel, docetaxel, and cabazitaxel to Pgp are 0.85 µM, 40.59 µM, and 13.53 µM, respectively. Based on acrylamide quenching, paclitaxel induced Pgp into a wide inward-facing open conformation at a high concentration but a slightly occluded conformation at lower concentrations. Both docetaxel and cabazitaxel shifted Pgp towards occluded states, each drug resulting in a unique degree of occlusion. Furthermore, molecular docking and energy calculations revealed that cabazitaxel binds with the "access tunnel" and blocks the subsequent nucleotide-binding domain dimerization. Our results indicate that the preference of taxanes for different binding sites on Pgp leads to distinct transport mechanisms. These results provide valuable insight into the interaction between taxanes and Pgp, which will enhance future drug development.
Collapse
Affiliation(s)
- Longwen Xu
- Department of Pharmaceutical and Biomedical Sciences, University of Georgia, Athens, GA 30602, USA
| | - Katherine G Schaefer
- Department of Physics and Astronomy, University of Missouri, Columbia, MO 65211, USA
| | - Gavin M King
- Department of Physics and Astronomy, University of Missouri, Columbia, MO 65211, USA; Joint with Department of Biochemistry, University of Missouri, Columbia, MO 65211, USA
| | - Zhong-Ru Xie
- Computational Drug Discovery Laboratory, School of Electrical and Computer Engineering, College of Engineering, University of Georgia, Athens, GA 30602, USA
| | - Michael G Bartlett
- Department of Pharmaceutical and Biomedical Sciences, University of Georgia, Athens, GA 30602, USA.
| |
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
|
Silva LDQC, Raspantini GL, Abriata JP, Luiz MT, de Sousa ACC, Moreira TDS, Magalhães EP, de Menezes RRPPB, Petrilli R, Marchetti JM, Eloy JO. PLGA/TPGS nanoparticles for docetaxel delivery: The pegylation effect on nanoparticle physicochemical properties and uptake and cytotoxicity in prostate cancer cells. J Pharm Sci 2025; 114:103766. [PMID: 40120677 DOI: 10.1016/j.xphs.2025.103766] [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: 10/04/2024] [Revised: 03/13/2025] [Accepted: 03/13/2025] [Indexed: 03/25/2025]
Abstract
Prostate cancer is the most common malignancy in men worldwide and docetaxel (DTX) is the treatment of choice. However, both the drug and formulation excipients for drug solubilization can cause side effects. In this context, the development of polymeric nanoparticles offers advantages to improve drug delivery and reduce toxicity. In the present work, factorial design was used to evaluate the effect of the amount of poly(L-lactide-co-glycolide) (PLGA) or poly(L-lactide-co-glycolide acid-polyethylene glycol) (PLGA-PEG), D-Alpha-Tocopheryl Polyethylene Glycol Succinate (TPGS) and ratio between aqueous and oily phases on the nanoparticle characteristics. The nanocarriers were characterized regarding particle size, polydispersity, zeta potential, DTX encapsulation efficiency, morphology by transmission electron microscopy, DSC, TGA and FTIR. It was evaluated in vitro for cytotoxicity and cellular uptake in prostate cancer cells. Pegylated nanoparticles, which have a different composition (TPGS%, AP:OP ratio), reduced the nanoparticle size to 105.97 ± 5.16 nm, in PDI 0.13 ± 0.03, zeta potential of -34.73 ± 1.19 mV and increased the encapsulation efficiency to 96.78 ± 1.20%. Characterization by DSC, TGA and FTIR confirmed drug encapsulation and showed colloidal stability. Pegylated nanoparticles were more stable upon serum incubation and adsorbed less proteins. In conclusion, the pegylation of the nanoparticles affected the physicochemical parameters. Also, the pegylation of nanoparticles decreased uptake by macrophages. Finally, cellular uptake and cell cytotoxicity were higher in tumor cells when compared to non-tumor cells, although they were not affected by pegylation.
Collapse
Affiliation(s)
| | | | - Juliana Palma Abriata
- University of Sao Paulo, Faculty of Pharmaceutical Sciences of Ribeirão Preto, Ribeirão Preto, SP, Brazil
| | - Marcela Tavares Luiz
- Paulista State University, Faculty of Pharmaceutical Sciences, Araraquara, SP, Brazil
| | - Ana Carolina Cruz de Sousa
- Federal University of Ceará, Faculty of Pharmacy, Dentistry and Nursing, Department of Pharmacy, Fortaleza, CE, Brazil
| | - Thais da Silva Moreira
- Federal University of Ceará, Faculty of Pharmacy, Dentistry and Nursing, Department of Pharmacy, Fortaleza, CE, Brazil
| | - Emanuel Paula Magalhães
- Federal University of Ceará, Faculty of Pharmacy, Dentistry and Nursing, Department of Pharmacy, Fortaleza, CE, Brazil
| | | | - Raquel Petrilli
- Federal University of Ceará, Faculty of Pharmacy, Dentistry and Nursing, Department of Pharmacy, Fortaleza, CE, Brazil
| | | | - Josimar O Eloy
- Federal University of Ceará, Faculty of Pharmacy, Dentistry and Nursing, Department of Pharmacy, Fortaleza, CE, Brazil.
| |
Collapse
|
4
|
Beretta GL, Cassinelli G, Rossi G, Azzariti A, Corbeau I, Tosi D, Perego P. Novel insights into taxane pharmacology: An update on drug resistance mechanisms, immunomodulation and drug delivery strategies. Drug Resist Updat 2025; 81:101223. [PMID: 40086175 DOI: 10.1016/j.drup.2025.101223] [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: 01/29/2025] [Revised: 02/24/2025] [Accepted: 02/27/2025] [Indexed: 03/16/2025]
Abstract
Taxanes are effective in several solid tumors. Paclitaxel, the main clinically available taxane, was approved in the early nineties, for the treatment of ovarian cancer and later on, together with the analogs docetaxel and cabazitaxel, for other malignancies. By interfering with microtubule function and impairing the separation of sister cells at mitosis, taxanes act as antimitotic agents, thereby counteracting the high proliferation rate of cancer cells. The action of taxanes goes beyond their antimitotic function because their main cellular targets, the microtubules, participate in multiple processes such as intracellular transport and cell shape maintenance. The clinical efficacy of taxanes is limited by the development of multiple resistance mechanisms. Among these, extracellular vesicles have emerged as new players. In addition, taxane metronomic schedules shows an impact on the tumor microenvironment reflected by antiangiogenic and immunomodulatory effects, an aspect of growing interest considering their inclusion in treatment regimens with immunotherapeutics. Preclinical studies have paved the bases for synergistic combinations of taxanes both with conventional and targeted agents. A variety of drug delivery strategies have provided novel opportunities to increase the drug activity. The ability of taxanes to orchestrate different cellular effects amenable to modulation suggests novel options to improve cures in lethal malignancies.
Collapse
Affiliation(s)
- Giovanni Luca Beretta
- Molecular Pharmacology Unit, Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, via Amadeo 42, Milan 20133, Italy.
| | - Giuliana Cassinelli
- Molecular Pharmacology Unit, Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, via Amadeo 42, Milan 20133, Italy.
| | - Giacomina Rossi
- Unit of Neurology 8, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan 20133, Italy.
| | - Amalia Azzariti
- Laboratory of Experimental Pharmacology, IRCCS Istituto Tumori Giovanni Paolo II, V.le O. Flacco, 65, Bari 70124, Italy.
| | - Iléana Corbeau
- Early Clinical Trial Unit, Medical Oncology Department, Institut régional du Cancer de Montpellier, Inserm U1194, Montpellier University, 208, rue de Apothicaires, 34298 Montpellier, France; Fondazione Gianni Bonadonna, via Bertani, 14, Milan 20154, Italy.
| | - Diego Tosi
- Early Clinical Trial Unit, Medical Oncology Department, Institut régional du Cancer de Montpellier, Inserm U1194, Montpellier University, 208, rue de Apothicaires, 34298 Montpellier, France; Fondazione Gianni Bonadonna, via Bertani, 14, Milan 20154, Italy.
| | - Paola Perego
- Molecular Pharmacology Unit, Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, via Amadeo 42, Milan 20133, Italy.
| |
Collapse
|
5
|
Yashima H, Araki T, Ishikawa Y, Ohshima S, Nagano D, Obayashi K, Horiguchi J, Yamamoto K. Therapeutic drug monitoring of docetaxel administered for breast cancer in a patient receiving rifampicin and clarithromycin to treat nontuberculous mycobacteriosis: A case report. Mol Clin Oncol 2025; 22:12. [PMID: 39649026 PMCID: PMC11618036 DOI: 10.3892/mco.2024.2807] [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: 08/16/2024] [Accepted: 11/04/2024] [Indexed: 12/10/2024] Open
Abstract
Docetaxel is metabolized by cytochrome P450 3A4 (CYP3A4), and is transported by organic anion transporting peptides (OATPs) and ABCB1, and its blood concentration is known to affect the risk of some docetaxel-related adverse drug reactions (ADRs). Thus, the concomitant use of docetaxel with drugs that inhibit or induce these transporters or CYP3A4 requires careful attention. A 58-year-old woman was receiving clarithromycin (400 mg twice daily), rifampicin (450 mg once daily) and ethambutol (500 mg once daily) for nontuberculous mycobacteriosis. The patient was diagnosed as having stage IV HER2-positive breast cancer, which was treated with a regimen of trastuzumab (8 mg/kg), pertuzumab (first dose: 840 mg; second dose onward: 420 mg) and docetaxel (75 mg/m2) every 3 weeks. To predict the risk of serious drug interactions with rifampicin and clarithromycin, the blood concentration of docetaxel was analyzed after administration of the first course. The docetaxel levels at 22 and 159 h after administration were 36.1 and 6.5 ng/ml, respectively, which were higher than previously reported data. In addition, the calculated elimination half-life of 55.7 h was ~3 times longer than previously reported data. Although the docetaxel level was high, the same dosage was used in subsequent courses because no serious ADRs were observed during the first course of therapy. After 4 months of chemotherapy, the patient received complete remission. In conclusion, concomitant use of rifampicin and clarithromycin may increase the blood concentration of docetaxel.
Collapse
Affiliation(s)
- Hideaki Yashima
- Department of Clinical Pharmacology and Therapeutics, Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan
- Department of Pharmacy, Gunma University Hospital, Maebashi, Gunma 371-8511, Japan
| | - Takuya Araki
- Department of Clinical Pharmacology and Therapeutics, Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan
- Department of Pharmacy, Gunma University Hospital, Maebashi, Gunma 371-8511, Japan
| | - Yuya Ishikawa
- Department of Pharmacy, Gunma University Hospital, Maebashi, Gunma 371-8511, Japan
| | - Sohei Ohshima
- Department of Clinical Pharmacology and Therapeutics, Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan
| | - Daisuke Nagano
- Education and Research Center for Clinical Pharmacy, Faculty of Pharmaceutical Science, Niigata University of Pharmacy and Applied Life Science, Niigata 956-8603, Japan
| | - Kyoko Obayashi
- Education Center for Clinical Pharmacy, Faculty of Pharmacy, Takasaki University of Health and Welfare, Takasaki, Gunma 370-0033, Japan
| | - Jun Horiguchi
- Department of Breast Surgery, International University of Health and Welfare, Otawara, Tochigi 324-8501, Japan
- Breast and Endocrine Surgery, Gunma University Hospital, Maebashi, Gunma 371-8511, Japan
| | - Koujirou Yamamoto
- Department of Clinical Pharmacology and Therapeutics, Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan
- Department of Pharmacy, Gunma University Hospital, Maebashi, Gunma 371-8511, Japan
| |
Collapse
|
6
|
Tang Y, Shen Q, Lin P, Chen Z, Fan D, Zhuo M, Gan Y, Su Y, Qian Q, Lin L, Xue E, Chen Z. aPD-L1-facilitated theranostic and tumor microenvironment remodeling of pancreatic cancer via docetaxel-loaded phase-transformation nanoparticles triggered by low-intensity pulsed ultrasound. J Nanobiotechnology 2025; 23:48. [PMID: 39871305 PMCID: PMC11773723 DOI: 10.1186/s12951-025-03105-7] [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: 07/26/2024] [Accepted: 01/10/2025] [Indexed: 01/30/2025] Open
Abstract
Early diagnosis of pancreatic ductal adenocarcinoma (PDAC) is challenging because of its depth, which often leads to misdiagnosis during ultrasound examinations. The unique PDAC tumor microenvironment (TME) is characterized by significant fibrous tissue growth, and high interstitial pressure hinders drug penetration into tumors. Additionally, hypoxia and immune suppression within the tumor contribute to poor responses to radiotherapy and chemotherapy, ultimately leading to an unfavorable prognosis. In this study, aPD-L1-modified docetaxel and perfluoropentane-loaded liquid‒vapor phase-transformation lipid nanoparticles (aPDL1-DTX/PFP@Lipid) were synthesized and had an average diameter of 61.63 nm with 84.3% antibody modification. We demonstrated that the nanoparticles (NPs) exhibited excellent PDAC-targeting capabilities both in vitro and in vivo. Upon exposure to low-intensity pulsed ultrasound (LIPUS) stimulation, the NPs underwent a phase transformation to form microbubbles with substantial molecular ultrasound diagnostic effects, and combined treatment resulted in a tumor growth inhibition rate of 88.91%. This treatment strategy also led to the infiltration of CD8+ T cells, the downregulation of Treg cells, the promotion of M1 macrophage polarization, the inhibition of fibrosis to reduce tumor stromal pressure, and the facilitation of perfluoropentane (PFP) gasification to release O2 and improve tumor hypoxia. In conclusion, aPD-L1-modified liquid‒vapor phase-transformation nanoparticles loaded with docetaxel (DTX) and PFP were successfully combined with ultrasound for the molecular diagnosis and targeted treatment of PDAC. aPDL1-DTX/PFP@Lipid could reshape the PDAC TME, offering a new approach for ultrasound-mediated diagnosis and treatment with promising clinical applications.
Collapse
Affiliation(s)
- Yi Tang
- Department of Ultrasound, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, China
- Fujian Institute of Ultrasonic Medicine Laboratory, 29 Xinquan Road, Fuzhou, China
| | - Qingling Shen
- Department of Ultrasound, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, China
- Fujian Institute of Ultrasonic Medicine Laboratory, 29 Xinquan Road, Fuzhou, China
| | - Peng Lin
- Department of Ultrasound, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, China
| | - Zhixin Chen
- Fujian College Association Instrumental Analysis Center, Fuzhou University, 2 Xueyuan Road, Fuzhou, China
| | - Denghui Fan
- Department of General Surgery, Affiliated Zhongshan Hospital of Dalian University, No. 6 Jiefang Street, Zhongshan District, Dalian, China
| | - Minling Zhuo
- Department of Ultrasound, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, China
| | - Yajiao Gan
- Department of Ultrasound, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, China
| | - Yixi Su
- Department of Ultrasound, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, China
| | - Qingfu Qian
- Department of Ultrasound, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, China
| | - Liwu Lin
- Department of Ultrasound, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, China
| | - Ensheng Xue
- Department of Ultrasound, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, China
| | - Zhikui Chen
- Department of Ultrasound, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, China.
- Fujian Institute of Ultrasonic Medicine Laboratory, 29 Xinquan Road, Fuzhou, China.
| |
Collapse
|
7
|
Favatella N, Dalton D, Byon W, Merali SJ, Klem C. Clinical Implications of Co-administering Apixaban with Key Interacting Medications. Clin Pharmacol Drug Dev 2024; 13:961-973. [PMID: 39046333 DOI: 10.1002/cpdd.1446] [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: 03/23/2023] [Accepted: 06/17/2024] [Indexed: 07/25/2024]
Abstract
With many available data sources, clinicians need to consider the benefit-risk profile of individual anticoagulants when balancing the need for anticoagulation, including evaluating the risks in patients with comorbidities and potential drug-drug interactions. This narrative review presents clinical data across multiple phases of drug development for the use of apixaban, a selective factor Xa inhibitor, when taken concomitantly with other agents, and evaluates the benefit-risk profile of apixaban with these interacting medications. Key subgroup analyses from the phase 3 ARISTOTLE trial (NCT00412984) are presented using data from patients who received either concomitant inhibitors or inducers of cytochrome P450 3A4 and/or P‑glycoprotein. We also review the available evidence for the use of apixaban in patients with cancer-associated thromboembolism, as well as the use of apixaban in patients with COVID-19.
Collapse
|
8
|
Heerma van Voss MR, Notohardjo J, van Dodewaard-de Jong J, Bloemendal HJ, Ter Heine R. A sub-pharmacological test dose does not predict individual docetaxel exposure in prostate cancer patients. Cancer Chemother Pharmacol 2024; 94:437-441. [PMID: 38951305 PMCID: PMC11420247 DOI: 10.1007/s00280-024-04684-2] [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: 01/28/2024] [Accepted: 06/13/2024] [Indexed: 07/03/2024]
Abstract
PURPOSE Docetaxel is a cytotoxic drug used for first-line treatment of various malignancies. It has a narrow therapeutic index and shows wide interpatient variability in clearance and toxicity. Tools for individual dose optimization are needed to maximize efficacy and avoid toxicity. METHODS We performed a proof-of-concept study (EudraCT 2016-003785-77) to evaluate whether pharmacokinetics after a sub-pharmacological test dose of 1000 µg docetaxel (millidose) could be used to predict therapeutic dose exposure. Thirty prostate cancer patients eligible for treatment with docetaxel as part of routine clinical care were included. An intravenous docetaxel millidose was administered 1-7 days prior to therapeutic docetaxel. After both doses plasma docetaxel concentrations were measured by ultra- high performance liquid chromatography-tandem mass spectrometry. The docetaxel clearance was estimated with non-linear mixed effects modeling. RESULTS Geometric mean docetaxel clearance was 57.9 L/h (GCV 78.6%) after admission of a millidose and 40.3 L/h (GCV 60.7%) after admission of a therapeutic dose. The millidose and therapeutic dose in a single patient were not significantly correlated (Spearman's rho R = 0.02, P = 0.92). CONCLUSION Docetaxel pharmacokinetics at milli- and therapeutic dose level showed insufficient correlation for individual dose optimization. However, the clearance of a docetaxel millidose and full dose are within the same order of magnitude. Therefore, docetaxel millidose pharmacokinetics could potentially facilitate prediction of docetaxel pharmacokinetics at a population level in situations where therapeutic dose levels are impractical, such as pharmacokinetic drug-drug interaction studies or pediatric studies.
Collapse
Affiliation(s)
- Marise R Heerma van Voss
- Meander Medical Center, Department of Internal Medicine, Amersfoort, The Netherlands.
- Department of Internal Medicine, Amsterdam University Medical Center, Amsterdam, The Netherlands.
| | - Jessica Notohardjo
- Meander Medical Center, Department of Internal Medicine, Amersfoort, The Netherlands
- Department of Internal Medicine, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | | | - Haiko J Bloemendal
- Meander Medical Center, Department of Internal Medicine, Amersfoort, The Netherlands
- Department of Medical Oncology, Research Institute for Medical Innovation, Radboudumc, Nijmegen, The Netherlands
| | - Rob Ter Heine
- Department of Pharmacy, Research Institute for Medical Innovation, Radboudumc, Nijmegen, The Netherlands
| |
Collapse
|
9
|
Li T, Zhou S, Wang L, Zhao T, Wang J, Shao F. Docetaxel, cyclophosphamide, and epirubicin: application of PBPK modeling to gain new insights for drug-drug interactions. J Pharmacokinet Pharmacodyn 2024; 51:367-384. [PMID: 38554227 DOI: 10.1007/s10928-024-09912-z] [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: 12/25/2023] [Accepted: 02/20/2024] [Indexed: 04/01/2024]
Abstract
The new adjuvant chemotherapy of docetaxel, epirubicin, and cyclophosphamide has been recommended for treating breast cancer. It is necessary to investigate the potential drug-drug Interactions (DDIs) since they have a narrow therapeutic window in which slight differences in exposure might result in significant differences in treatment efficacy and tolerability. To guide clinical rational drug use, this study aimed to evaluate the DDI potentials of docetaxel, cyclophosphamide, and epirubicin in cancer patients using physiologically based pharmacokinetic (PBPK) models. The GastroPlus™ was used to develop the PBPK models, which were refined and validated with observed data. The established PBPK models accurately described the pharmacokinetics (PKs) of three drugs in cancer patients, and the predicted-to-observed ratios of all the PK parameters met the acceptance criterion. The PBPK model predicted no significant changes in plasma concentrations of these drugs during co-administration, which was consistent with the observed clinical phenomenon. Besides, the verified PBPK models were then used to predict the effect of other Cytochrome P450 3A4 (CYP3A4) inhibitors/inducers on these drug exposures. In the DDI simulation, strong CYP3A4 modulators changed the exposure of three drugs by 0.71-1.61 fold. Therefore, patients receiving these drugs in combination with strong CYP3A4 inhibitors should be monitored regularly to prevent adverse reactions. Furthermore, co-administration of docetaxel, cyclophosphamide, or epirubicin with strong CYP3A4 inducers should be avoided. In conclusion, the PBPK models can be used to further investigate the DDI potential of each drug and to develop dosage recommendations for concurrent usage by additional perpetrators or victims.
Collapse
Affiliation(s)
- Tongtong Li
- Phase I Clinical Trial Unit, The First Affiliated Hospital With Nanjing Medical University, Nanjing, 210029, China
- Department of Clinical Pharmacology, School of Pharmacy College, Nanjing Medical University, Nanjing, 211166, China
| | - Sufeng Zhou
- Phase I Clinical Trial Unit, The First Affiliated Hospital With Nanjing Medical University, Nanjing, 210029, China
| | - Lu Wang
- Phase I Clinical Trial Unit, The First Affiliated Hospital With Nanjing Medical University, Nanjing, 210029, China
| | - Tangping Zhao
- Phase I Clinical Trial Unit, The First Affiliated Hospital With Nanjing Medical University, Nanjing, 210029, China
- Department of Clinical Pharmacology, School of Pharmacy College, Nanjing Medical University, Nanjing, 211166, China
| | - Jue Wang
- Division of Breast Surgery, The First Affiliated Hospital With Nanjing Medical University, Guangzhou Road 300, Nanjing, 210029, Jiangsu Province, China
| | - Feng Shao
- Phase I Clinical Trial Unit, The First Affiliated Hospital With Nanjing Medical University, Nanjing, 210029, China.
- Department of Clinical Pharmacology, School of Pharmacy College, Nanjing Medical University, Nanjing, 211166, China.
| |
Collapse
|
10
|
Sun Y, Cheng Y, Hertz DL. Using maximum plasma concentration (C max) to personalize taxane treatment and reduce toxicity. Cancer Chemother Pharmacol 2024; 93:525-539. [PMID: 38734836 DOI: 10.1007/s00280-024-04677-1] [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: 02/01/2024] [Accepted: 05/06/2024] [Indexed: 05/13/2024]
Abstract
Taxanes are a widely used class of anticancer agents that play a vital role in the treatment of a variety of cancers. However, toxicity remains a major concern of using taxane drugs as some toxicities are highly prevalent, they can not only adversely affect patient prognosis but also compromise the overall treatment plan. Among all kinds of factors that associated with taxane toxicity, taxane exposure has been extensively studied, with different pharmacokinetic (PK) parameters being used as toxicity predictors. Compared to other widely used predictors such as the area under the drug plasma concentration curve versus time (AUC) and time above threshold plasma drug concentration, maximum plasma concentration (Cmax) is easier to collect and shows promise for use in clinical practice. In this article, we review the previous research on using Cmax to predict taxane treatment outcomes. While Cmax and toxicity have been extensively studied, research on the relationship between Cmax and efficacy is lacking. Most of the articles find a positive relationship between Cmax and toxicity but several articles have contradictory findings. Future clinical trials are needed to validate the relationship between Cmax and treatment outcome and determine whether Cmax can serve as a useful surrogate endpoint of taxane treatment efficacy.
Collapse
Affiliation(s)
- Yuchen Sun
- Department of Clinical Pharmacy, University of Michigan College of Pharmacy, Ann Arbor, MI, USA
| | - Yue Cheng
- Department of Clinical Pharmacy, University of Michigan College of Pharmacy, Ann Arbor, MI, USA
| | - Daniel L Hertz
- Department of Clinical Pharmacy, University of Michigan College of Pharmacy, Ann Arbor, MI, USA.
| |
Collapse
|
11
|
Beheshtizadeh N, Amiri Z, Tabatabaei SZ, Seraji AA, Gharibshahian M, Nadi A, Saeinasab M, Sefat F, Kolahi Azar H. Boosting antitumor efficacy using docetaxel-loaded nanoplatforms: from cancer therapy to regenerative medicine approaches. J Transl Med 2024; 22:520. [PMID: 38816723 PMCID: PMC11137998 DOI: 10.1186/s12967-024-05347-9] [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: 04/13/2024] [Accepted: 05/25/2024] [Indexed: 06/01/2024] Open
Abstract
The intersection of nanotechnology and pharmacology has revolutionized the delivery and efficacy of chemotherapeutic agents, notably docetaxel, a key drug in cancer treatment. Traditionally limited by poor solubility and significant side effects, docetaxel's therapeutic potential has been significantly enhanced through its incorporation into nanoplatforms, such as nanofibers and nanoparticles. This advancement offers targeted delivery, controlled release, and improved bioavailability, dramatically reducing systemic toxicity and enhancing patient outcomes. Nanofibers provide a versatile scaffold for the controlled release of docetaxel, utilizing techniques like electrospinning to tailor drug release profiles. Nanoparticles, on the other hand, enable precise drug delivery to tumor cells, minimizing damage to healthy tissues through sophisticated encapsulation methods such as nanoprecipitation and emulsion. These nanotechnologies not only improve the pharmacokinetic properties of docetaxel but also open new avenues in regenerative medicine by facilitating targeted therapy and cellular regeneration. This narrative review highlights the transformative impact of docetaxel-loaded nanoplatforms in oncology and beyond, showcasing the potential of nanotechnology to overcome the limitations of traditional chemotherapy and pave the way for future innovations in drug delivery and regenerative therapies. Through these advancements, nanotechnology promises a new era of precision medicine, enhancing the efficacy of cancer treatments while minimizing adverse effects.
Collapse
Affiliation(s)
- Nima Beheshtizadeh
- Department of Tissue Engineering, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
- Regenerative Medicine Group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
| | - Zahra Amiri
- Department of Materials Science and Engineering, Sharif University of Technology, 1458889694, Tehran, Iran
- Regenerative Medicine Group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Seyedeh Zoha Tabatabaei
- Cardiogenetic Research Center, Rajaie Cardiovascular Medical and Research Institute, Iran University of Medical Sciences, Tehran, Iran
| | - Amir Abbas Seraji
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, Canada
- Department of Polymer Engineering and Color Technology, Amirkabir University of Technology, Tehran, Iran
| | - Maliheh Gharibshahian
- Department of Tissue Engineering and Applied Cell Sciences, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
- Regenerative Medicine Group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Akram Nadi
- Stem Cell Biology Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Regenerative Medicine Group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Morvarid Saeinasab
- Department of Biomedical and Electronics Engineering, School of Engineering, University of Bradford, Bradford, UK
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Farshid Sefat
- Department of Biomedical and Electronics Engineering, School of Engineering, University of Bradford, Bradford, UK
- Interdisciplinary Research Centre in Polymer Science & Technology (Polymer IRC), University of Bradford, Bradford, UK
| | - Hanieh Kolahi Azar
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Pathology, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
12
|
Konoshenko M, Laktionov P, Bryzgunova O. Prostate cancer therapy outcome prediction: are miRNAs a suitable guide for therapeutic decisions? Andrology 2024; 12:705-718. [PMID: 37750354 DOI: 10.1111/andr.13535] [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: 07/11/2023] [Revised: 09/04/2023] [Accepted: 09/09/2023] [Indexed: 09/27/2023]
Abstract
BACKGROUND Radical prostatectomy, radiotherapy, chemotherapy, and androgen-deprivation therapy are among the most common treatment options for different forms of prostate cancer (PCa). However, making therapeutic decisions is difficult due to the lack of reliable prediction markers indicating therapy outcomes in clinical practice. The involvement of miRNAs in all mechanisms of the PCa development and their easy detection characterize them as attractive PCa biomarkers. Although there are extensive data on the role of miRNAs in PCa therapy resistance and sensitivity development, the issues of whether they could be used as a guide for therapy choice and, if so, how we can progress toward this goal, remain unclear. Thus, generalizable reviews and studies which summarize, compare, and analyze data on miRNA involvement in responses to different types of PCa therapies are required. OBJECTIVES Data on the involvement of miRNAs in therapy responses, on the role of cross-miRNA expression in different therapies, and on miRNA targets were analyzed in order to determine the miRNA-related factors which can lend perspective to the future development of personalized predictors of PCa sensitivity/resistance to therapies. MATERIALS AND METHODS The data available on the miRNAs associated with different PCa therapies (resistance and sensitivity therapies) are summarized and analyzed in this study, including analyses using bioinformatics resources. Special attention was dedicated to the mechanisms of the development of therapy resistance. RESULTS AND DISCUSSION A comprehensive combined analysis of the current data revealed a panel of miRNAs that were shown to be most closely associated with the PCa therapy response and were found to regulate the genes involved in PCa development via cell proliferation regulation, epithelial-mesenchymal transition (EMT), apoptosis, cell-cycle progression, angiogenesis, metastasis and invasion regulation, androgen-independent development, and colony formation. CONCLUSION The selected miRNA-based panel has the potential to be a guide for therapeutic decision making in the effective treatment of PCa.
Collapse
Affiliation(s)
- MariaYu Konoshenko
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch, Russian Academy of Sciences, Novosibirsk, Russia
| | - Pavel Laktionov
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch, Russian Academy of Sciences, Novosibirsk, Russia
| | - Olga Bryzgunova
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch, Russian Academy of Sciences, Novosibirsk, Russia
| |
Collapse
|
13
|
Hsu TI, Chen YP, Zhang RL, Chen ZA, Wu CH, Chang WC, Mou CY, Chan HWH, Wu SH. Overcoming the Blood-Brain Tumor Barrier with Docetaxel-Loaded Mesoporous Silica Nanoparticles for Treatment of Temozolomide-Resistant Glioblastoma. ACS APPLIED MATERIALS & INTERFACES 2024; 16:21722-21735. [PMID: 38629735 PMCID: PMC11071047 DOI: 10.1021/acsami.4c04289] [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: 03/14/2024] [Accepted: 04/01/2024] [Indexed: 05/03/2024]
Abstract
While temozolomide (TMZ) has been a cornerstone in the treatment of newly diagnosed glioblastoma (GBM), a significant challenge has been the emergence of resistance to TMZ, which compromises its clinical benefits. Additionally, the nonspecificity of TMZ can lead to detrimental side effects. Although TMZ is capable of penetrating the blood-brain barrier (BBB), our research addresses the need for targeted therapy to circumvent resistance mechanisms and reduce off-target effects. This study introduces the use of PEGylated mesoporous silica nanoparticles (MSN) with octyl group modifications (C8-MSN) as a nanocarrier system for the delivery of docetaxel (DTX), providing a novel approach for treating TMZ-resistant GBM. Our findings reveal that C8-MSN is biocompatible in vitro, and DTX@C8-MSN shows no hemolytic activity at therapeutic concentrations, maintaining efficacy against GBM cells. Crucially, in vivo imaging demonstrates preferential accumulation of C8-MSN within the tumor region, suggesting enhanced permeability across the blood-brain tumor barrier (BBTB). When administered to orthotopic glioma mouse models, DTX@C8-MSN notably prolongs survival by over 50%, significantly reduces tumor volume, and decreases side effects compared to free DTX, indicating a targeted and effective approach to treatment. The apoptotic pathways activated by DTX@C8-MSN, evidenced by the increased levels of cleaved caspase-3 and PARP, point to a potent therapeutic mechanism. Collectively, the results advocate DTX@C8-MSN as a promising candidate for targeted therapy in TMZ-resistant GBM, optimizing drug delivery and bioavailability to overcome current therapeutic limitations.
Collapse
Affiliation(s)
- Tsung-I Hsu
- Ph.D.
Program in Medical Neuroscience, College of Medical Science and Technology, Taipei Medical University and National Health Research
Institutes, Taipei 110, Taiwan
- International
Master Program in Medical Neuroscience, College of Medical Science
and Technology, Taipei Medical University, Taipei 110, Taiwan
| | - Yi-Ping Chen
- Graduate
Institute of Nanomedicine and Medical Engineering, Taipei Medical University, Taipei 110, Taiwan
- International
Ph.D. Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 110, Taiwan
| | - Rong-Lin Zhang
- Nano
Targeting & Therapy Biopharma Inc., Taipei 110, Taiwan
| | - Zih-An Chen
- Graduate
Institute of Nanomedicine and Medical Engineering, Taipei Medical University, Taipei 110, Taiwan
| | - Cheng-Hsun Wu
- Nano
Targeting & Therapy Biopharma Inc., Taipei 110, Taiwan
| | - Wen-Chang Chang
- Graduate
Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Chung-Yuan Mou
- Nano
Targeting & Therapy Biopharma Inc., Taipei 110, Taiwan
- Department
of Chemistry, National Taiwan University, Taipei 106, Taiwan
| | | | - Si-Han Wu
- Graduate
Institute of Nanomedicine and Medical Engineering, Taipei Medical University, Taipei 110, Taiwan
- International
Ph.D. Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 110, Taiwan
| |
Collapse
|
14
|
Wang G, Sun Q, Li X, Mei S, Li S, Li Z. A Cross-sectional Comparative Analysis of Eleven Population Pharmacokinetic Models for Docetaxel in Chinese Breast Cancer Patients. Curr Drug Metab 2024; 25:479-488. [PMID: 39161139 PMCID: PMC11826906 DOI: 10.2174/0113892002322494240816032948] [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: 05/09/2024] [Revised: 07/19/2024] [Accepted: 07/31/2024] [Indexed: 08/21/2024]
Abstract
OBJECTIVE Various population pharmacokinetic (PPK) models have been established to help determine the appropriate dosage of docetaxel, however, no clear consensus on optimal dosing has been achieved. The purpose of this study is to perform an external evaluation of published models in order to test their predictive performance, and to find an appropriate PPK model for Chinese breast cancer patients. METHODS A systematic literature search of docetaxel PPK models was performed using PubMed, Web of Science, China National Knowledge Infrastructure, and WanFang databases. The predictive performance of eleven identified models was evaluated using prediction-based and simulation-based diagnostics on an independent dataset (112 docetaxel concentrations from 56 breast cancer patients). The -2×log (likelihood) and Akaike information criterion were also calculated to evaluate model fit. RESULTS The median prediction error of eight of the eleven models was less than 10%. The model fitting results showed that the three-compartment model of Bruno et al. had the best prediction performance and that the three compartment model of Wang et al. had the best simulation effect. Furthermore, although the covariates that significantly affect PK parameters were different between them, seven models demonstrated that docetaxel PK parameters were influenced by liver function. CONCLUSIONS Three compartment PPK models may be predictive of optimal docetaxel dosage for Chinese breast cancer patients. However, for patients with impaired liver function, the choice of which model to use to predict the blood concentration of docetaxel still requires great care.
Collapse
Affiliation(s)
- Genzhu Wang
- Electric Power Teaching Hospital, Capital Medical University, Beijing, 100073, China
| | - Qiang Sun
- Electric Power Teaching Hospital, Capital Medical University, Beijing, 100073, China
| | - Xiaojing Li
- Electric Power Teaching Hospital, Capital Medical University, Beijing, 100073, China
| | - Shenghui Mei
- Beijing Tiantan Hospital,Capital Medical University, Beijing, 100070, China
| | - Shihui Li
- Electric Power Teaching Hospital, Capital Medical University, Beijing, 100073, China
| | - Zhongdong Li
- Electric Power Teaching Hospital, Capital Medical University, Beijing, 100073, China
| |
Collapse
|
15
|
Yang X, Grimstein M, Pressly M, Fletcher EP, Shord S, Leong R. Utility of Physiologically Based Pharmacokinetic Modeling to Investigate the Impact of Physiological Changes of Pregnancy and Cancer on Oncology Drug Pharmacokinetics. Pharmaceutics 2023; 15:2727. [PMID: 38140068 PMCID: PMC10748010 DOI: 10.3390/pharmaceutics15122727] [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: 09/18/2023] [Revised: 11/14/2023] [Accepted: 11/28/2023] [Indexed: 12/24/2023] Open
Abstract
BACKGROUND The treatment of cancer during pregnancy remains challenging with knowledge gaps in drug dosage, safety, and efficacy due to the under-representation of this population in clinical trials. Our aim was to investigate physiological changes reported in both pregnancy and cancer populations into a PBPK modeling framework that allows for a more accurate estimation of PK changes in pregnant patients with cancer. METHODS Paclitaxel and docetaxel were selected to validate a population model using clinical data from pregnant patients with cancer. The validated population model was subsequently used to predict the PK of acalabrutinib in pregnant patients with cancer. RESULTS The Simcyp pregnancy population model reasonably predicted the PK of docetaxel in pregnant patients with cancer, while a modified model that included a 2.5-fold increase in CYP2C8 abundance, consistent with the increased expression during pregnancy, was needed to reasonably predict the PK of paclitaxel in pregnant patients with cancer. Changes in protein binding levels of patients with cancer had a minimal impact on the predicted clearance of paclitaxel and docetaxel. PBPK modeling predicted approximately 60% lower AUC and Cmax for acalabrutinib in pregnant versus non-pregnant patients with cancer. CONCLUSIONS Our results suggest that PBPK modeling is a promising approach to investigate the effects of pregnancy and cancer on the PK of oncology drugs and potentially inform dosing for pregnant patients with cancer. Further evaluation and refinement of the population model are needed for pregnant patients with cancer with additional compounds and clinical PK data.
Collapse
Affiliation(s)
| | | | | | | | | | - Ruby Leong
- Office of Clinical Pharmacology, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, 10903 New Hampshire Avenue, Silver Spring, MD 20993, USA; (X.Y.); (M.G.); (S.S.)
| |
Collapse
|
16
|
Wimmer K, Sachet M, Ramos C, Frantal S, Birnleitner H, Brostjan C, Exner R, Filipits M, Bago-Horvath Z, Rudas M, Bartsch R, Gnant M, Singer CF, Balic M, Egle D, Oehler R, Fitzal F. Differential immunomodulatory effects of epirubicin/cyclophosphamide and docetaxel in breast cancer patients. J Exp Clin Cancer Res 2023; 42:300. [PMID: 37957750 PMCID: PMC10644559 DOI: 10.1186/s13046-023-02876-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: 05/17/2023] [Accepted: 10/29/2023] [Indexed: 11/15/2023] Open
Abstract
BACKGROUND Epirubicin/cyclophosphamide (EC) and docetaxel (D) are commonly used in a sequential regimen in the neoadjuvant treatment of early, high-risk or locally advanced breast cancer (BC). Novel approaches to increase the response rate combine this treatment with immunotherapies such as PD-1 inhibition. However, the expected stimulatory effect on lymphocytes may depend on the chemotherapy backbone. Therefore, we separately compared the immunomodulatory effects of EC and D in the setting of a randomized clinical trial. METHODS Tumor and blood samples of 154 patients from the ABCSG-34 trial were available (76 patients received four cycles of EC followed by four cycles of D; 78 patients get the reverse treatment sequence). Tumor-infiltrating lymphocytes, circulating lymphocytes and 14 soluble immune mediators were determined at baseline and at drug change. Furthermore, six BC cell lines were treated with E, C or D and co-cultured with immune cells. RESULTS Initial treatment with four cycles of EC reduced circulating B and T cells by 94% and 45%, respectively. In contrast, no comparable effects on lymphocytes were observed in patients treated with initial four cycles of D. Most immune mediators decreased under EC whereas D-treatment resulted in elevated levels of CXCL10, urokinase-type plasminogen activator (uPA) and its soluble receptor (suPAR). Accordingly, only the exposure of BC cell lines to D induced similar increases as compared to E. While treatment of BC cells with E was associated with cell shrinkage and apoptosis, D induced cell swelling and accumulation of cells in G2 phase. CONCLUSION The deleterious effect of EC on lymphocytes indicates strong immunosuppressive properties of this combination therapy. D, in contrast, has no effect on lymphocytes, but triggers the secretion of stimulatory proteins in vivo and in vitro, indicating a supportive effect on the immune system. Underlying differences in the induced cell death might be causal. These divergent immunomodulatory effects of epirubicin/cyclophosphamide and docetaxel should be considered when planning future combinations with immunotherapies in breast cancer.
Collapse
Affiliation(s)
- Kerstin Wimmer
- Department of General Surgery, Division of Visceral Surgery and Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, A-1090, Vienna, Austria
- Austrian Breast & Colorectal Cancer Study Group (ABCSG), Vienna, Austria
| | - Monika Sachet
- Department of General Surgery, Division of Visceral Surgery and Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, A-1090, Vienna, Austria
| | - Cristiano Ramos
- Department of General Surgery, Division of Visceral Surgery and Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, A-1090, Vienna, Austria
| | - Sophie Frantal
- Austrian Breast & Colorectal Cancer Study Group (ABCSG), Vienna, Austria
| | - Hanna Birnleitner
- Department of General Surgery, Division of Visceral Surgery and Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, A-1090, Vienna, Austria
| | - Christine Brostjan
- Department of General Surgery, Division of Vascular Surgery, Medical University of Vienna, 1090, Vienna, Austria
| | - Ruth Exner
- Department of General Surgery, Division of Visceral Surgery and Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, A-1090, Vienna, Austria
| | - Martin Filipits
- Austrian Breast & Colorectal Cancer Study Group (ABCSG), Vienna, Austria
- Center for Cancer Research, Medical University of Vienna, 1090, Vienna, Austria
| | - Zsuzsanna Bago-Horvath
- Austrian Breast & Colorectal Cancer Study Group (ABCSG), Vienna, Austria
- Department of Pathology, Medical University of Vienna, 1090, Vienna, Austria
- Comprehensive Cancer Center, Medical University of Vienna, Spitalgasse 23, 1090, Vienna, Austria
| | - Margaretha Rudas
- Department of Pathology, Medical University of Vienna, 1090, Vienna, Austria
- Comprehensive Cancer Center, Medical University of Vienna, Spitalgasse 23, 1090, Vienna, Austria
| | - Rupert Bartsch
- Austrian Breast & Colorectal Cancer Study Group (ABCSG), Vienna, Austria
- Department of Medicine 1, Division of Oncology, Medical University of Vienna, 1090, Vienna, Austria
| | - Michael Gnant
- Austrian Breast & Colorectal Cancer Study Group (ABCSG), Vienna, Austria
- Comprehensive Cancer Center, Medical University of Vienna, Spitalgasse 23, 1090, Vienna, Austria
| | - Christian F Singer
- Austrian Breast & Colorectal Cancer Study Group (ABCSG), Vienna, Austria
- Comprehensive Cancer Center, Medical University of Vienna, Spitalgasse 23, 1090, Vienna, Austria
- Department of Gynecology, Medical University of Vienna, 1090, Vienna, Austria
| | - Marija Balic
- Austrian Breast & Colorectal Cancer Study Group (ABCSG), Vienna, Austria
- Department of Oncology, Medical University of Graz, Graz, Austria
| | - Daniel Egle
- Austrian Breast & Colorectal Cancer Study Group (ABCSG), Vienna, Austria
- Department of Gynecology, Medical University Innsbruck, Innsbruck, Austria
| | - Rudolf Oehler
- Department of General Surgery, Division of Visceral Surgery and Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, A-1090, Vienna, Austria.
- Austrian Breast & Colorectal Cancer Study Group (ABCSG), Vienna, Austria.
| | - Florian Fitzal
- Department of General Surgery, Division of Visceral Surgery and Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, A-1090, Vienna, Austria
- Austrian Breast & Colorectal Cancer Study Group (ABCSG), Vienna, Austria
| |
Collapse
|
17
|
Cheng Y, Pang X, Wu J, Zhou L, Cao J, Wang L, Qian K, Yang P, Xu M, Sheng D, Meng R, Wang P, Guo Q, Xu S, Wei Y, Zhang Q. Medium-chain triglyceride-stabilized docetaxel-loaded HSA nanoparticles effectively inhibited metastatic non-small cell lung cancer. Drug Deliv Transl Res 2023; 13:2869-2884. [PMID: 37204680 DOI: 10.1007/s13346-023-01355-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/19/2023] [Indexed: 05/20/2023]
Abstract
Metastatic non-small cell lung cancer (NSCLC) is refractory with a very poor prognosis. Docetaxel (DTX) injection (Taxotere®) has been approved for the treatment of locally advanced or metastatic NSCLC. However, its clinical application is restricted by severe adverse effects and non-selective tissue distribution. In this study, we successfully developed DTX-loaded human serum albumin (HSA) nanoparticles (DNPs) with modified Nab technology, by introducing medium-chain triglyceride (MCT) as a stabilizer. The optimized formulation had a particle size of approximately 130 nm and a favorable stabilization time of more than 24 h. DNPs dissociated in circulation in a concentration-dependent manner and slowly released DTX. Compared with DTX injection, DNPs were more effectively taken up by NSCLC cells, thus exerting stronger inhibitory effects on their proliferation, adhesion, migration, and invasion. In addition, DNPs showed prolonged blood retention and increased tumor accumulation relative to DTX injection. Ultimately, DNPs produced more potent inhibitory effects on primary or metastatic tumor foci than DTX injections but caused markedly lower organ toxicity and hematotoxicity. Overall, these results support that DNPs hold great potential for the treatment of metastatic NSCLC in clinical.
Collapse
Affiliation(s)
- Yunlong Cheng
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Xiaoying Pang
- Department of Pharmacy, the Obstetrics and Gynecology Hospital, Fudan University, 200011, Shanghai, China
| | - Jing Wu
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Lingling Zhou
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Jinxu Cao
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Liuchang Wang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Kang Qian
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Peng Yang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Minjun Xu
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Dongyu Sheng
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Ran Meng
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Pengzhen Wang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Qian Guo
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Shuting Xu
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Yan Wei
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China.
| | - Qizhi Zhang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, 201203, China.
| |
Collapse
|
18
|
Gutiérrez-Saucedo RA, Gómez-López JC, Villanueva-Briseño AA, Topete A, Soltero-Martínez JFA, Mendizábal E, Jasso-Gastinel CF, Taboada P, Figueroa-Ochoa EB. Pluronic F127 and P104 Polymeric Micelles as Efficient Nanocarriers for Loading and Release of Single and Dual Antineoplastic Drugs. Polymers (Basel) 2023; 15:polym15102249. [PMID: 37242824 DOI: 10.3390/polym15102249] [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: 04/15/2023] [Revised: 05/03/2023] [Accepted: 05/05/2023] [Indexed: 05/28/2023] Open
Abstract
The potential application of biodegradable and biocompatible polymeric micelles formed by Pluronic F127 and P104 as nanocarriers of the antineoplastic drugs docetaxel (DOCE) and doxorubicin (DOXO) is presented in this work. The release profile was carried out under sink conditions at 37 °C and analyzed using the Higuchi, Korsmeyer-Peppas, and Peppas-Sahlin diffusion models. The cell viability of HeLa cells was evaluated using the proliferation cell counting kit CCK-8 assay. The formed polymeric micelles solubilized significant amounts of DOCE and DOXO, and released them in a sustained manner for 48 h, with a release profile composed of an initial rapid release within the first 12 h followed by a much slower phase the end of the experiments. In addition, the release was faster under acidic conditions. The model that best fit the experimental data was the Korsmeyer-Peppas one and denoted a drug release dominated by Fickian diffusion. When HeLa cells were exposed for 48 h to DOXO and DOCE drugs loaded inside P104 and F127 micelles, they showed lower IC50 values than those reported by other researchers using polymeric nanoparticles, dendrimers or liposomes as alternative carriers, indicating that a lower drug concentration is needed to decrease cell viability by 50%.
Collapse
Affiliation(s)
- Ramón A Gutiérrez-Saucedo
- Laboratorio de Proyectos Modulares, Departamento de Química, Centro Universitario de Ciencias Exactas e Ingeniería, Universidad de Guadalajara, Blvd. M. García Barragán 1421, Guadalajara 44430, Jalisco, Mexico
| | - Julio C Gómez-López
- Laboratorio de Proyectos Modulares, Departamento de Química, Centro Universitario de Ciencias Exactas e Ingeniería, Universidad de Guadalajara, Blvd. M. García Barragán 1421, Guadalajara 44430, Jalisco, Mexico
| | - Adrián A Villanueva-Briseño
- Laboratorio de Inmunología, Departamento de Fisiología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Sierra Mojada 950, Guadalajara 44340, Jalisco, Mexico
| | - Antonio Topete
- Laboratorio de Inmunología, Departamento de Fisiología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Sierra Mojada 950, Guadalajara 44340, Jalisco, Mexico
| | - J F Armando Soltero-Martínez
- Departamento de Ingeniería Química, Centro Universitario de Ciencias Exactas e Ingeniería, Universidad de Guadalajara, Blvd. M. García Barragán 1421, Guadalajara 44430, Jalisco, Mexico
| | - Eduardo Mendizábal
- Departamento de Ingeniería Química, Centro Universitario de Ciencias Exactas e Ingeniería, Universidad de Guadalajara, Blvd. M. García Barragán 1421, Guadalajara 44430, Jalisco, Mexico
| | - Carlos F Jasso-Gastinel
- Departamento de Ingeniería Química, Centro Universitario de Ciencias Exactas e Ingeniería, Universidad de Guadalajara, Blvd. M. García Barragán 1421, Guadalajara 44430, Jalisco, Mexico
| | - Pablo Taboada
- Grupo de Física de Coloides y Polímeros, Departamento de Física de Partículas e Instituto de Materiales (IMATUS), Universidad de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Edgar B Figueroa-Ochoa
- Laboratorio de Proyectos Modulares, Departamento de Química, Centro Universitario de Ciencias Exactas e Ingeniería, Universidad de Guadalajara, Blvd. M. García Barragán 1421, Guadalajara 44430, Jalisco, Mexico
| |
Collapse
|
19
|
Sousa-Pimenta M, Estevinho LM, Szopa A, Basit M, Khan K, Armaghan M, Ibrayeva M, Sönmez Gürer E, Calina D, Hano C, Sharifi-Rad J. Chemotherapeutic properties and side-effects associated with the clinical practice of terpene alkaloids: paclitaxel, docetaxel, and cabazitaxel. Front Pharmacol 2023; 14:1157306. [PMID: 37229270 PMCID: PMC10203197 DOI: 10.3389/fphar.2023.1157306] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 04/10/2023] [Indexed: 05/27/2023] Open
Abstract
Over the years, many biological and synthetic agents have been explored and tested in attempts to halt the spread of cancer and/or cure it. Currently, several natural compounds have and are being considered in this regard. For example, paclitaxel is a potent anticancer drug that originates from the tree Taxus brevifolia. Paclitaxel has several derivatives, namely, docetaxel and cabazitaxel. These agents work by disrupting microtubule assembling dynamics and inducing cell cycle arrest at the G2/M phase of the cell cycle, ultimately triggering apoptosis. Such features have helped to establish paclitaxel as an authoritative therapeutic compound against neoplastic disorders. After the completion of compound (hemi) synthesis, this drug received approval for the treatment of solid tumors either alone or in combination with other agents. In this review, we explore the mechanisms of action of paclitaxel and its derivatives, the different formulations available, as well as the molecular pathways of cancer resistance, potential risks, and other therapeutic applications. In addition, the role of paclitaxel in hematological malignancies is explored, and potential limitations in the therapeutic use of paclitaxel at the clinical level are examined. Furthermore, paclitaxel is known to cause increased antigen presentation. The immunomodulatory potential of taxanes, alone or in combination with other pharmacologic agents, is explored. Despite terpene-alkaloids derivatives' anti-mitotic potential, the impact of this class of drugs on other oncogenic pathways, such as epithelial-to-mesenchymal transition and the epigenetic modulation of the transcription profile of cancer cells, is also analyzed, shedding light on potential future chemotherapeutic approaches to cancer.
Collapse
Affiliation(s)
- Mário Sousa-Pimenta
- Department of Onco‐Hematology, Portuguese Institute of Oncology of Porto (IPO-Porto), Porto, Portugal
- i3S—Instituto de Investigação e Inovação em Saúde da Universidade do Porto, Porto, Portugal
| | - Letícia M. Estevinho
- Mountain Research Center (CIMO), Polytechnic Institute of Bragança, Campus Santa Apolónia, Bragança, Portugal
- Department of Biology and Biotechnology, Agricultural College of Bragança, Polytechnic Institute of Bragança, Campus Santa Apolónia, Bragança, Portugal
| | - Agnieszka Szopa
- Chair and Department of Pharmaceutical Botany, Medical College, Jagiellonian University, Kraków, Poland
| | - Mahnoor Basit
- Department of Healthcare Biotechnology, Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), Islamabad, Pakistan
| | - Khushbukhat Khan
- Department of Healthcare Biotechnology, Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), Islamabad, Pakistan
| | - Muhammad Armaghan
- Department of Healthcare Biotechnology, Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), Islamabad, Pakistan
| | - Manshuk Ibrayeva
- Department of Natural Sciences, Faculty of Science and Technology, Caspian University of Technology and Engineering named after Sh.Yessenov, Aktau, Kazakhstan
| | - Eda Sönmez Gürer
- Department of Pharmacognosy, Faculty of Pharmacy, Sivas Cumhuriyet University, Sivas, Türkiye
| | - Daniela Calina
- Department of Clinical Pharmacy, University of Medicine and Pharmacy of Craiova, Craiova, Romania
| | - Christophe Hano
- Department of Biological Chemistry, Université ď Orléans, Chartres, France
| | | |
Collapse
|
20
|
Wu Q, Sharma D. Autophagy and Breast Cancer: Connected in Growth, Progression, and Therapy. Cells 2023; 12:1156. [PMID: 37190065 PMCID: PMC10136604 DOI: 10.3390/cells12081156] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 03/29/2023] [Accepted: 04/05/2023] [Indexed: 05/17/2023] Open
Abstract
Despite an increase in the incidence of breast cancer worldwide, overall prognosis has been consistently improving owing to the development of multiple targeted therapies and novel combination regimens including endocrine therapies, aromatase inhibitors, Her2-targeted therapies, and cdk4/6 inhibitors. Immunotherapy is also being actively examined for some breast cancer subtypes. This overall positive outlook is marred by the development of resistance or reduced efficacy of the drug combinations, but the underlying mechanisms are somewhat unclear. It is interesting to note that cancer cells quickly adapt and evade most therapies by activating autophagy, a catabolic process designed to recycle damaged cellular components and provide energy. In this review, we discuss the role of autophagy and autophagy-associated proteins in breast cancer growth, drug sensitivity, tumor dormancy, stemness, and recurrence. We further explore how autophagy intersects and reduces the efficacy of endocrine therapies, targeted therapies, radiotherapy, chemotherapies as well as immunotherapy via modulating various intermediate proteins, miRs, and lncRNAs. Lastly, the potential application of autophagy inhibitors and bioactive molecules to improve the anticancer effects of drugs by circumventing the cytoprotective autophagy is discussed.
Collapse
Affiliation(s)
| | - Dipali Sharma
- Department of Oncology, Johns Hopkins University School of Medicine and the Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD 21287-0013, USA
| |
Collapse
|
21
|
Palange AL, Mascolo DD, Ferreira M, Gawne PJ, Spanò R, Felici A, Bono L, Moore TL, Salerno M, Armirotti A, Decuzzi P. Boosting the Potential of Chemotherapy in Advanced Breast Cancer Lung Metastasis via Micro-Combinatorial Hydrogel Particles. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2205223. [PMID: 36683230 PMCID: PMC10074128 DOI: 10.1002/advs.202205223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Revised: 12/01/2022] [Indexed: 06/17/2023]
Abstract
Breast cancer cell colonization of the lungs is associated with a dismal prognosis as the distributed nature of the disease and poor permeability of the metastatic foci challenge the therapeutic efficacy of small molecules, antibodies, and nanomedicines. Taking advantage of the unique physiology of the pulmonary circulation, here, micro-combinatorial hydrogel particles (µCGP) are realized via soft lithographic techniques to enhance the specific delivery of a cocktail of cytotoxic nanoparticles to metastatic foci. By cross-linking short poly(ethylene glycol) (PEG) chains with erodible linkers within a shape-defining template, a deformable and biodegradable polymeric skeleton is realized and loaded with a variety of therapeutic and imaging agents, including docetaxel-nanoparticles. In a model of advanced breast cancer lung metastasis, µCGP amplified the colocalization of docetaxel-nanoparticles with pulmonary metastatic foci, prolonged the retention of chemotoxic molecules at the diseased site, suppressed lesion growth, and boosted survival beyond 20 weeks post nodule engraftment. The flexible design and modular architecture of µCGP would allow the efficient deployment of complex combination therapies in other vascular districts too, possibly addressing metastatic diseases of different origins.
Collapse
Affiliation(s)
- Anna Lisa Palange
- Laboratory of Nanotechnology for Precision MedicineFondazione Istituto Italiano di TecnologiaVia Morego 30Genoa16163Italy
| | - Daniele Di Mascolo
- Laboratory of Nanotechnology for Precision MedicineFondazione Istituto Italiano di TecnologiaVia Morego 30Genoa16163Italy
| | - Miguel Ferreira
- Laboratory of Nanotechnology for Precision MedicineFondazione Istituto Italiano di TecnologiaVia Morego 30Genoa16163Italy
- Present address:
Harvard Medical School, Department of RadiologyMassachusetts General HospitalBostonMA02114USA
| | - Peter J. Gawne
- Laboratory of Nanotechnology for Precision MedicineFondazione Istituto Italiano di TecnologiaVia Morego 30Genoa16163Italy
| | - Raffaele Spanò
- Laboratory of Nanotechnology for Precision MedicineFondazione Istituto Italiano di TecnologiaVia Morego 30Genoa16163Italy
| | - Alessia Felici
- Laboratory of Nanotechnology for Precision MedicineFondazione Istituto Italiano di TecnologiaVia Morego 30Genoa16163Italy
- Present address:
Division of Oncology, Department of Medicine and Department of PathologyStanford University School of MedicineStanfordCA94305USA
| | - Luca Bono
- Analytical Chemistry FacilityFondazione Istituto Italiano di TecnologiaVia Morego 30Genoa16163Italy
| | - Thomas Lee Moore
- Laboratory of Nanotechnology for Precision MedicineFondazione Istituto Italiano di TecnologiaVia Morego 30Genoa16163Italy
| | - Marco Salerno
- Materials Characterization FacilityFondazione Istituto Italiano di TecnologiaVia Morego 30Genoa16163Italy
| | - Andrea Armirotti
- Analytical Chemistry FacilityFondazione Istituto Italiano di TecnologiaVia Morego 30Genoa16163Italy
| | - Paolo Decuzzi
- Laboratory of Nanotechnology for Precision MedicineFondazione Istituto Italiano di TecnologiaVia Morego 30Genoa16163Italy
| |
Collapse
|
22
|
Patil VM, Noronha V, Menon N, Singh A, Ghosh-Laskar S, Budrukkar A, Bhattacharjee A, Swain M, Mathrudev V, Nawale K, Balaji A, Peelay Z, Alone M, Pathak S, Mahajan A, Kumar S, Purandare N, Agarwal A, Puranik A, Pendse S, Reddy Yallala M, Sahu H, Kapu V, Dey S, Choudhary J, Krishna MR, Shetty A, Karuvandan N, Ravind R, Rai R, Jobanputra K, Chaturvedi P, Pai PS, Chaukar D, Nair S, Thiagarajan S, Prabhash K. Results of Phase III Randomized Trial for Use of Docetaxel as a Radiosensitizer in Patients With Head and Neck Cancer, Unsuitable for Cisplatin-Based Chemoradiation. J Clin Oncol 2023; 41:2350-2361. [PMID: 36706347 DOI: 10.1200/jco.22.00980] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
PURPOSE There is a lack of published literature on systemic therapeutic options in cisplatin-ineligible patients with locally advanced head and neck squamous cell carcinoma (LAHNSCC) undergoing chemoradiation. Docetaxel was assessed as a radiosensitizer in this situation. METHODS This was a randomized phase II/III study. Adult patients (age ≥ 18 years) with LAHNSCC planned for chemoradiation and an Eastern Cooperative Oncology Group performance status of 0-2 and who were cisplatin-ineligible were randomly assigned in 1:1 to either radiation alone or radiation with concurrent docetaxel 15 mg/m2 once weekly for a maximum of seven cycles. The primary end point was 2-year disease-free survival (DFS). RESULTS The study recruited 356 patients between July 2017 and May 2021. The 2-year DFS was 30.3% (95% CI, 23.6 to 37.4) versus 42% (95% CI, 34.6 to 49.2) in the RT and Docetaxel-RT arms, respectively (hazard ratio, 0.673; 95% CI, 0.521 to 0.868; P value = .002). The corresponding median overall survival (OS) was 15.3 months (95% CI, 13.1 to 22.0) and 25.5 months (95% CI, 17.6 to 32.5), respectively (log-rank P value = .035). The 2-year OS was 41.7% (95% CI, 34.1 to 49.1) versus 50.8% (95% CI, 43.1 to 58.1) in the RT and Docetaxel-RT arms, respectively (hazard ratio, 0.747; 95% CI, 0.569 to 0.980; P value = .035). There was a higher incidence of grade 3 or above mucositis (22.2% v 49.7%; P < .001), odynophagia (33.5% v 52.5%; P < .001), and dysphagia (33% v 49.7%; P = .002) with the addition of docetaxel. CONCLUSION The addition of docetaxel to radiation improved DFS and OS in cisplatin-ineligible patients with LAHNSCC.[Media: see text].
Collapse
Affiliation(s)
- Vijay Maruti Patil
- Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Vanita Noronha
- Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Nandini Menon
- Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Ajay Singh
- Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Sarbani Ghosh-Laskar
- Department of Radiation Oncology, Tata Memorial Hospital, Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Ashwini Budrukkar
- Department of Radiation Oncology, Tata Memorial Hospital, Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Atanu Bhattacharjee
- Leicester Real World Evidence Unit, Leicester University, Leicester, United Kingdom
| | - Monali Swain
- Department of Radiation Oncology, Tata Memorial Hospital, Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Vijayalakshmi Mathrudev
- Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Kavita Nawale
- Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Arun Balaji
- Department of Speech and Therapy, Tata Memorial Hospital, Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Zoya Peelay
- Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Mitali Alone
- Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Shruti Pathak
- Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Abhishek Mahajan
- Department of Radiology, Tata Memorial Hospital, Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Suman Kumar
- Department of Radiology, Tata Memorial Hospital, Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Nilendu Purandare
- Department of Nuclear Medicine, Tata Memorial Hospital, Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Archi Agarwal
- Department of Nuclear Medicine, Tata Memorial Hospital, Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Ameya Puranik
- Department of Nuclear Medicine, Tata Memorial Hospital, Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Shantanu Pendse
- Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Monica Reddy Yallala
- Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Harsh Sahu
- Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Venkatesh Kapu
- Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Sayak Dey
- Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Jatin Choudhary
- Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Madala Ravi Krishna
- Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Alok Shetty
- Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Naveen Karuvandan
- Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Rahul Ravind
- Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Rahul Rai
- Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Kunal Jobanputra
- Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Pankaj Chaturvedi
- Department of Head and Neck Surgery, Tata Memorial Hospital, Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Prathamesh S Pai
- Department of Head and Neck Surgery, Tata Memorial Hospital, Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Devendra Chaukar
- Department of Head and Neck Surgery, Tata Memorial Hospital, Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Sudhir Nair
- Department of Head and Neck Surgery, Tata Memorial Hospital, Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Shivakumar Thiagarajan
- Department of Head and Neck Surgery, Tata Memorial Hospital, Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Kumar Prabhash
- Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute (HBNI), Mumbai, India
| |
Collapse
|
23
|
Rijcken CJF, De Lorenzi F, Biancacci I, Hanssen RGJM, Thewissen M, Hu Q, Atrafi F, Liskamp RMJ, Mathijssen RHJ, Miedema IHC, Menke-van der Houven van Oordt CW, van Dongen GAMS, Vugts DJ, Timmers M, Hennink WE, Lammers T. Design, development and clinical translation of CriPec®-based core-crosslinked polymeric micelles. Adv Drug Deliv Rev 2022; 191:114613. [PMID: 36343757 DOI: 10.1016/j.addr.2022.114613] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 10/18/2022] [Accepted: 10/30/2022] [Indexed: 11/06/2022]
Abstract
Nanomedicines are used to improve the efficacy and safety of pharmacotherapeutic interventions. Unraveling the biological behavior of nanomedicines, including their biodistribution and target site accumulation, is essential to establish design criteria that contribute to superior performance. CriPec® technology is based on amphiphilic methoxy-poly(ethylene glycol)-b-poly[N-(2-hydroxypropyl) methacrylamide lactate] (mPEG-b-pHPMAmLacn) block copolymers, which are designed to upon self-assembly covalently entrap active pharmaceutical ingredients (API) in core-crosslinked polymeric micelles (CCPM). Key features of CCPM are a prolonged circulation time, high concentrations at pathological sites, and low levels of accumulation in the majority of healthy tissues. Proprietary hydrolysable linkers allow for tunable and sustained release of entrapped API, including hydrophobic and hydrophilic small molecules, as well as peptides and oligonucleotides. Preclinical imaging experiments provided valuable information on their tumor and tissue accumulation and distribution, as well as on uptake by cancer, healthy and immune cells. The frontrunner formulation CPC634, which refers to 65 nm-sized CCPM entrapping the chemotherapeutic drug docetaxel, showed excellent pharmacokinetic properties, safety, tumor accumulation and antitumor efficacy in multiple animal models. In the clinic, CPC634 also demonstrated favorable pharmacokinetics, good tolerability, signs of efficacy, and enhanced localization in tumor tissue as compared to conventional docetaxel. PET imaging of radiolabeled CPC634 showed quantifiable accumulation in ∼50 % of tumors and metastases in advanced-stage cancer patients, and demonstrated potential for use in a theranostic setting even when applied at a companion diagnostic dose. Altogether, the preclinical and clinical results obtained to date demonstrate that mPEG-b-pHPMAmLacn CCPM based on CriPec® technology are a potent, tunable, broadly applicable and well-tolerable platform for targeted drug delivery and improved anticancer therapy.
Collapse
Affiliation(s)
| | - Federica De Lorenzi
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, Aachen, Germany
| | - Ilaria Biancacci
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, Aachen, Germany
| | | | | | - Qizhi Hu
- Cristal Therapeutics, Maastricht, the Netherlands
| | - Florence Atrafi
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | | | - Ron H J Mathijssen
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Iris H C Miedema
- Amsterdam UMC Location Vrije Universiteit Amsterdam, Department of Medical Oncology, Amsterdam, the Netherlands; Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, the Netherlands
| | - C Willemien Menke-van der Houven van Oordt
- Amsterdam UMC Location Vrije Universiteit Amsterdam, Department of Medical Oncology, Amsterdam, the Netherlands; Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, the Netherlands
| | - Guus A M S van Dongen
- Amsterdam UMC Location Vrije Universiteit Amsterdam, Department of Radiology and Nuclear Medicine, Amsterdam, the Netherlands; Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, the Netherlands
| | - Danielle J Vugts
- Amsterdam UMC Location Vrije Universiteit Amsterdam, Department of Radiology and Nuclear Medicine, Amsterdam, the Netherlands; Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, the Netherlands
| | - Matt Timmers
- Cristal Therapeutics, Maastricht, the Netherlands
| | - Wim E Hennink
- Department of Pharmaceutics, Utrecht University, Utrecht, the Netherlands
| | - Twan Lammers
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, Aachen, Germany.
| |
Collapse
|
24
|
Lean Body Mass and Total Body Weight Versus Body Surface Area as a Determinant of Docetaxel Pharmacokinetics and Toxicity. Ther Drug Monit 2022; 44:755-761. [PMID: 36006609 DOI: 10.1097/ftd.0000000000001029] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 07/19/2022] [Indexed: 01/29/2023]
Abstract
AIM This study examined whether anthropometric and body composition parameters such as body surface area (BSA), lean body mass (LBM), and total body weight (TBW) are correlated with docetaxel clearance and exposure by analyzing area under the curve. In addition, LBM, TBW, and a fixed dose were compared with BSA as dosing parameters for dose individualization of docetaxel. METHODS Thirty-six patients receiving docetaxel chemotherapy for breast or metastatic castration-resistant prostate carcinoma were included. Before treatment, LBM was measured using a dual-energy X-ray absorptiometry scanner. Blood samples were collected up to 180 minutes after dosing to analyze docetaxel concentrations and determine individual pharmacokinetic parameters. RESULTS No significant correlations were found between docetaxel clearance and the anthropometric and body composition variables (BSA, LBM, and TBW). The area under the curve was significantly but poorly correlated with BSA [r = 0.452 ( P = 0.016)] and TBW [r = 0.476 ( P = 0.011)]. The mean absolute percentage error and mean error of simulated dosing based on LBM and fixed dosing were not significantly different from those of BSA. For TBW, only mean absolute percentage error was significantly higher compared with dosing based on BSA (24.1 versus 17.1, P = 0.001). CONCLUSIONS There was no clinically relevant correlation between docetaxel pharmacokinetics and the anthropometric and body composition variables BSA, LBM, and TBW. Therefore, dose individualization of docetaxel based on LBM, TBW, or fixed dosing cannot be recommended over BSA-based dosing.
Collapse
|
25
|
Lampropoulos I, Charoupa M, Kavousanakis M. Intra-tumor heterogeneity and its impact on cytotoxic therapy in a two-dimensional vascular tumor growth model. Chem Eng Sci 2022. [DOI: 10.1016/j.ces.2022.117792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
26
|
Ali Z, Sajid M, Manzoor S, Ahmad MM, Khan MI, Elboughdiri N, Kashif M, Shanableh A, Rajhi W, Mersni W, Bayraktar E, Salem SB. Biodegradable Magnetic Molecularly Imprinted Anticancer Drug Carrier for the Targeted Delivery of Docetaxel. ACS OMEGA 2022; 7:28516-28524. [PMID: 35990493 PMCID: PMC9386705 DOI: 10.1021/acsomega.2c03299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 07/22/2022] [Indexed: 05/07/2023]
Abstract
Molecularly imprinted biodegradable polymers are receiving considerable attention in drug delivery due to their ability of targeted recognition and biocompatibility. This study reports the synthesis of a novel fluorescence-active magnetic molecularly imprinted drug carrier (MIDC) using a glucose-based biodegradable cross-linking agent for the delivery of anticancer drug docetaxel. The magnetic molecularly imprinted polymer (MMIP) was characterized through scanning electron microscopy (SEM), Fourier transform infrared spectroscopy (FTIR), X-ray diffraction spectroscopy, and vibrating sample magnetometry (VSM). The MMIP presented a magnetization value of 0.0059 emu g-1 and binding capacity of 72 mg g-1 with docetaxel. In vitro and in vivo studies were performed to observe the effectiveness of the MIDC for drug delivery. The cell viability assay suggested that the MMIP did not present toxic effects on healthy cells. The magnetic property of the MMIP allowed quick identification of the drug carrier at the target site by applying the external magnetic field to mice (after 20 min of loading) and taking X-ray images. The novel MMIP-based drug carrier could thus deliver the drug at the target site without affecting the healthy cells.
Collapse
Affiliation(s)
- Zeeshan Ali
- Institute
of Chemical Sciences, Bahauddin Zakariya
University, Multan 60000, Pakistan
| | - Muhammad Sajid
- Institute
of Chemical Sciences, Bahauddin Zakariya
University, Multan 60000, Pakistan
- . Tel.: 00923040801998
| | - Suryyia Manzoor
- Institute
of Chemical Sciences, Bahauddin Zakariya
University, Multan 60000, Pakistan
| | | | - Muhammad Imran Khan
- Research
Institute of Sciences and Engineering (RISE), University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Noureddine Elboughdiri
- Chemical
Engineering Process Department, National
School of Engineers Gabes, University of Gabes, Gabes 6011, Tunisia
- . Tel.: 00966549571015
| | - Muhammad Kashif
- Department
of Chemistry, Emerson University, Multan 60000, Pakistan
| | - Abdallah Shanableh
- Research
Institute of Sciences and Engineering (RISE), University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Wajdi Rajhi
- Mechanical
Engineering Department, College of Engineering,
University of Ha’il, P.O. Box 2440, Ha’il 81441,Saudi Arabia
| | - Wael Mersni
- National
School of Engineers of Tunis, University
of Tunis El Manar, Tunis 1068, Tunisia
| | - Emin Bayraktar
- School
of Mechanical and Manufacturing Engineering, ISAE-SUPMECA Institute
of Mechanics of Paris, Saint-Ouen 93400, France
| | - Sahbi Ben Salem
- National
School of Engineers of Tunis, University
of Tunis El Manar, Tunis 1068, Tunisia
| |
Collapse
|
27
|
Patil VM, Kolkur M, Kumar Chinthala S, Waratkar G, Menon N, Noronha V, Ghosh Laskar S, Simha V, Talreja V, Dhumal S, Chandrasekharan A, Prabhash K. Long term toxicity and tolerance of concurrent docetaxel with radiotherapy in cisplatin-ineligible head and neck cancer patients. Oral Oncol 2022; 130:105908. [DOI: 10.1016/j.oraloncology.2022.105908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 05/07/2022] [Accepted: 05/08/2022] [Indexed: 12/01/2022]
|
28
|
Shrivastava SP, Elhence A, Jinwala P, Bansal S, Chitalkar P, Bhatnagar S, Patidar R, Asati V. A Rare Case of Life-Threatening Extensive Mucocutaneous Adverse Reaction Induced by Docetaxel in a Breast Cancer Patient: Toxic Epidermal Necrolysis, a Case Report with Review of Literature. Indian J Med Paediatr Oncol 2022. [DOI: 10.1055/s-0042-1743125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022] Open
Abstract
AbstractFever and extensive necrosis with 30% or more epidermal involvement along with mucous membrane is known as toxic epidermal necrolysis (TEN). It is a life-threatening mucocutaneous disease and is usually drug induced. We report a rare case of docetaxel-induced TEN. A patient with metastatic breast carcinoma received single agent docetaxel and developed severe skin and mucous membrane reaction involving more than 30% of the skin, and managed conservatively in intensive care unit but she succumbed to her illness. Although common toxicities reported with docetaxel include alopecia, nail damage, myelosuppression, and erythema multiforme major, TEN after docetaxel is very rare and can be a life-threatening complication as in our case.
Collapse
Affiliation(s)
- Shiv Prasad Shrivastava
- Department of Medical Oncology, Sri Aurobindo Institute of Medical Sciences, Indore, Madhya Pradesh, India
| | - Aditya Elhence
- Department of Medical Oncology, Sri Aurobindo Institute of Medical Sciences, Indore, Madhya Pradesh, India
| | - Prutha Jinwala
- Department of Medical Oncology, Sri Aurobindo Institute of Medical Sciences, Indore, Madhya Pradesh, India
| | - Shashank Bansal
- Department of Medical Oncology, Sri Aurobindo Institute of Medical Sciences, Indore, Madhya Pradesh, India
| | - Prakash Chitalkar
- Department of Medical Oncology, Sri Aurobindo Institute of Medical Sciences, Indore, Madhya Pradesh, India
| | - Shweta Bhatnagar
- Department of Radiology, Sri Aurobindo Institute of Medical Sciences, Indore, Madhya Pradesh, India
| | - Rajesh Patidar
- Department of Medical Oncology, Sri Aurobindo Institute of Medical Sciences, Indore, Madhya Pradesh, India
| | - Vikas Asati
- Department of Medical Oncology, Sri Aurobindo Institute of Medical Sciences, Indore, Madhya Pradesh, India
| |
Collapse
|
29
|
Aranda E, Teruel JA, Ortiz A, Pérez-Cárceles MD, Aranda FJ. Interaction of Docetaxel with Phosphatidylcholine Membranes: A Combined Experimental and Computational Study. J Membr Biol 2022; 255:277-291. [PMID: 35175383 PMCID: PMC9167220 DOI: 10.1007/s00232-022-00219-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 02/07/2022] [Indexed: 11/06/2022]
Abstract
The antineoplastic drug Docetaxel is a second generation taxane which is used against a great variety of cancers. The drug is highly lipophilic and produces a great array of severe toxic effects that limit its therapeutic effectiveness. The study of the interaction between Docetaxel and membranes is very scarce, however, it is required in order to get clues in relation with its function, mechanism of toxicity and possibilities of new formulations. Using phosphatidylcholine biomimetic membranes, we examine the interaction of Docetaxel with the phospholipid bilayer combining an experimental study, employing a series of biophysical techniques like Differential Scanning Calorimetry, X-Ray Diffraction and Infrared Spectroscopy, and a Molecular Dynamics simulation. Our experimental results indicated that Docetaxel incorporated into DPPC bilayer perturbing the gel to liquid crystalline phase transition and giving rise to immiscibility when the amount of the drug is increased. The drug promotes the gel ripple phase, increasing the bilayer thickness in the fluid phase, and is also able to alter the hydrogen-bonding interactions in the interfacial region of the bilayer producing a dehydration effect. The results from computational simulation agree with the experimental ones and located the Docetaxel molecule forming small clusters in the region of the carbon 8 of the acyl chain palisade overlapping with the carbonyl region of the phospholipid. Our results support the idea that the anticancer drug is embedded into the phospholipid bilayer to a limited amount and produces structural perturbations which might affect the function of the membrane.
Collapse
Affiliation(s)
- Elisa Aranda
- Departamento de Bioquímica y Biología Molecular-A, Facultad de Veterinaria, Universidad de Murcia, 30100, Murcia, Spain
- Hospital Universitario Virgen de la Arrixaca, Área de Salud 1, Murcia, Spain
| | - José A Teruel
- Departamento de Bioquímica y Biología Molecular-A, Facultad de Veterinaria, Universidad de Murcia, 30100, Murcia, Spain
| | - Antonio Ortiz
- Departamento de Bioquímica y Biología Molecular-A, Facultad de Veterinaria, Universidad de Murcia, 30100, Murcia, Spain
| | - María Dolores Pérez-Cárceles
- Departamento de Medicina Legal y Forense, Facultad de Medicina, Instituto de Investigación Biomédica (IMIB-Arrixaca), Universidad de Murcia, 30120, Murcia, Spain
| | - Francisco J Aranda
- Departamento de Bioquímica y Biología Molecular-A, Facultad de Veterinaria, Universidad de Murcia, 30100, Murcia, Spain.
| |
Collapse
|
30
|
Miedema IHC, Zwezerijnen GJC, Huisman MC, Doeleman E, Mathijssen RHJ, Lammers T, Hu Q, van Dongen GAMS, Rijcken CJF, Vugts DJ, Menke-van der Houven van Oordt CW. PET-CT Imaging of Polymeric Nanoparticle Tumor Accumulation in Patients. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2201043. [PMID: 35427430 DOI: 10.1002/adma.202201043] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 04/08/2022] [Indexed: 06/14/2023]
Abstract
Several FDA/EMA-approved nanomedicines have demonstrated improved pharmacokinetics and toxicity profiles compared to their conventional chemotherapeutic counterparts. The next step to increase therapeutic efficacy depends on tumor accumulation, which can be highly heterogeneous. A clinical tool for patient stratification is urgently awaited. Therefore, a docetaxel-entrapping polymeric nanoparticle (89 Zr-CPC634) is radiolabeled, and positron emission tomography/computed tomography (PET/CT) imaging is performed in seven patients with solid tumors with two different doses of CPC634: an on-treatment (containing 60 mg m-2 docetaxel) and a diagnostic (1-2 mg docetaxel) dose (NCT03712423). Pharmacokinetic half-life for 89 Zr-CPC634 is mean 97.0 ± 14.4 h on-treatment, and 62.4 ± 12.9 h for the diagnostic dose (p = 0.003). At these doses accumulation is observed in 46% and 41% of tumor lesions with a median accumulation in positive lesions 96 h post-injection of 4.94 and 4.45%IA kg-1 (p = 0.91), respectively. In conclusion, PET/CT imaging with a diagnostic dose of 89 Zr-CPC634 accurately reflects on-treatment tumor accumulation and thus opens the possibility for patient stratification in cancer nanomedicine with polymeric nanoparticles.
Collapse
Affiliation(s)
- Iris H C Miedema
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Medical Oncology, De Boelelaan 1117, Amsterdam, 1081 HV, The Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, De Boelelaan 1117, Amsterdam, 1081 HV, The Netherlands
| | - Gerben J C Zwezerijnen
- Cancer Center Amsterdam, Imaging and Biomarkers, De Boelelaan 1117, Amsterdam, 1081 HV, The Netherlands
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Radiology and Nuclear Medicine, De Boelelaan 1117, Amsterdam, 1081 HV, The Netherlands
| | - Marc C Huisman
- Cancer Center Amsterdam, Imaging and Biomarkers, De Boelelaan 1117, Amsterdam, 1081 HV, The Netherlands
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Radiology and Nuclear Medicine, De Boelelaan 1117, Amsterdam, 1081 HV, The Netherlands
| | - Ellen Doeleman
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Medical Oncology, De Boelelaan 1117, Amsterdam, 1081 HV, The Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, De Boelelaan 1117, Amsterdam, 1081 HV, The Netherlands
| | - Ron H J Mathijssen
- Erasmus University Medical Center, Erasmus University, Erasmus MC Cancer Institute, Department of Medical Oncology, Doctor Molewaterplein 40, Rotterdam, 3015 GD, The Netherlands
| | - Twan Lammers
- Institute for Experimental Molecular Imaging, Helmholtz Institute for Biomedical Engineering, RWTH - Aachen University, Templergraben 55, 52062, Aachen, Germany
| | - Qizhi Hu
- Cristal Therapeutics, Oxfordlaan 55, Maastricht, 6229 EV, The Netherlands
| | - Guus A M S van Dongen
- Cancer Center Amsterdam, Imaging and Biomarkers, De Boelelaan 1117, Amsterdam, 1081 HV, The Netherlands
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Radiology and Nuclear Medicine, De Boelelaan 1117, Amsterdam, 1081 HV, The Netherlands
| | | | - Danielle J Vugts
- Cancer Center Amsterdam, Imaging and Biomarkers, De Boelelaan 1117, Amsterdam, 1081 HV, The Netherlands
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Radiology and Nuclear Medicine, De Boelelaan 1117, Amsterdam, 1081 HV, The Netherlands
| | - C Willemien Menke-van der Houven van Oordt
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Medical Oncology, De Boelelaan 1117, Amsterdam, 1081 HV, The Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, De Boelelaan 1117, Amsterdam, 1081 HV, The Netherlands
| |
Collapse
|
31
|
Fu YK, Wang BJ, Tseng JC, Huang SH, Lin CY, Kuo YY, Hour TC, Chuu CP. Combination treatment of docetaxel with caffeic acid phenethyl ester suppresses the survival and the proliferation of docetaxel-resistant prostate cancer cells via induction of apoptosis and metabolism interference. J Biomed Sci 2022; 29:16. [PMID: 35197069 PMCID: PMC8864857 DOI: 10.1186/s12929-022-00797-z] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Accepted: 02/12/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Docetaxel has been approved by USFDA as a first-line treatment for castration-resistant prostate cancer (CRPC) patients. Patients receiving androgen deprivation therapy along with docetaxel result in superior survival, lower serum prostate specific antigen (PSA) level, and better quality of life. However, a significant proportion of these patients ultimately develop resistance to docetaxel within months. Caffeic acid phenethyl ester (CAPE), one of the main bioactive components extracted from the propolis, has been reported to be effective for repressing the tumor growth, the migration and invasion of prostate cancer (PCa) cells, as well as the downstream signaling and stability of androgen receptor (AR). We hence determined if combination treatment of docetaxel with CAPE can suppress the proliferation and the survival of docetaxel-resistant PCa cells. METHODS We established docetaxel-resistant PC/DX25 and DU/DX50 CRPC cell lines from PC-3 and DU-145 human PCa cells, respectively. Proliferation assay, MTT assay, flow cytometry with Annexin V staining, Comet Assay, and nude mice xenograft model were applied to determine the effects of combination treatment on cell proliferation and survival of the docetaxel-resistant PCa cells. Micro-Western Array (MWA) and qRT-PCR were used to investigate the molecular mechanism lying underneath. RESULTS Combination treatment effectively suppressed the proliferation, survival and tumor growth of docetaxel-resistant PCa cells both in vitro and in nude mice. Comet assay and flow cytometry indicated that combination treatment induced apoptosis in docetaxel-resistant PCa cells. MWA and Western blotting assay revealed that combination treatment suppressed protein expression of Bcl-2, AKT2, c-Myc, apoptosis and caspase activation inhibitor (AVEN), pyruvate kinase M2 (PKM2) but increased protein expression of Bax, caspase 3, cytochrome c, glucose-6-phosphate dehydrogenase (G6PD) and acylglycerol kinase (AGK). Overexpression of Bcl-2 in the docetaxel-resistant PCa cells enhanced cell proliferation of docetaxel-resistant PCa cells under combination treatment. Analysis with qRT-PCR suggested that combination treatment decreased cholesterol biosynthesis genes DHCR24 (24-dehydrocholesterol reductase) and LSS (lanosterol synthase) but increased genes involved in glycolysis and TCA cycle. CONCLUSIONS Combination treatment of docetaxel with CAPE effectively suppressed the proliferation and survival of docetaxel-resistant PCa cells via inhibition of Bcl-2 and c-Myc as well as induction of metabolism interference. Combination treatment can be beneficial for patients with docetaxel-resistant PCa.
Collapse
Affiliation(s)
- Yu-Ke Fu
- Institute of Cellular and System Medicine, National Health Research Institutes, No. 35, Keyan Road, Zhunan Town, 35053, Miaoli County, Taiwan
| | - Bi-Juan Wang
- Institute of Cellular and System Medicine, National Health Research Institutes, No. 35, Keyan Road, Zhunan Town, 35053, Miaoli County, Taiwan
| | - Jen-Chih Tseng
- Institute of Cellular and System Medicine, National Health Research Institutes, No. 35, Keyan Road, Zhunan Town, 35053, Miaoli County, Taiwan
| | - Shih-Han Huang
- Institute of Cellular and System Medicine, National Health Research Institutes, No. 35, Keyan Road, Zhunan Town, 35053, Miaoli County, Taiwan
| | - Ching-Yu Lin
- Institute of Cellular and System Medicine, National Health Research Institutes, No. 35, Keyan Road, Zhunan Town, 35053, Miaoli County, Taiwan
| | - Ying-Yu Kuo
- Institute of Cellular and System Medicine, National Health Research Institutes, No. 35, Keyan Road, Zhunan Town, 35053, Miaoli County, Taiwan
| | - Tzyh-Chyuan Hour
- Department of Biochemistry, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chih-Pin Chuu
- Institute of Cellular and System Medicine, National Health Research Institutes, No. 35, Keyan Road, Zhunan Town, 35053, Miaoli County, Taiwan. .,Graduate Program for Aging and Graduate Institute of Basic Research Sciences, China Medical University, Taichung, Taiwan. .,Biotechnology Center, National Chung Hsing University, Taichung City, Taiwan. .,Department of Life Sciences, National Central University, Taoyuan City, Taiwan.
| |
Collapse
|
32
|
Rizvi SSB, Akhtar N, Minhas MU, Mahmood A, Khan KU. Synthesis and Characterization of Carboxymethyl Chitosan Nanosponges with Cyclodextrin Blends for Drug Solubility Improvement. Gels 2022; 8:55. [PMID: 35049590 PMCID: PMC8775084 DOI: 10.3390/gels8010055] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 01/06/2022] [Accepted: 01/10/2022] [Indexed: 02/04/2023] Open
Abstract
This study aimed to enhance the solubility and release characteristics of docetaxel by synthesizing highly porous and stimuli responsive nanosponges, a nano-version of hydrogels with the additional qualities of both hydrogels and nano-systems. Nanosponges were prepared by the free radical polymerization technique and characterized by their solubilization efficiency, swelling studies, sol-gel studies, percentage entrapment efficiency, drug loading, FTIR, PXRD, TGA, DSC, SEM, zeta sizer and in vitro dissolution studies. In vivo toxicity study was conducted to assess the safety of the oral administration of prepared nanosponges. FTIR, TGA and DSC studies confirmed the successful grafting of components into the stable nano-polymeric network. A porous and sponge-like structure was visualized through SEM images. The particle size of the optimized formulation was observed in the range of 195 ± 3 nm. The fabricated nanosponges noticeably enhanced the drug loading and solubilization efficiency of docetaxel in aqueous media. The drug release of fabricated nanosponges was significantly higher at pH 6.8 as compared to pH 1.2 and 4.5. An acute oral toxicity study endorsed the safety of the system. Due to an efficient preparation technique, as well as its enhanced solubility, excellent physicochemical properties, improved dissolution and non-toxic nature, nanosponges could be an efficient and a promising approach for the oral delivery of poorly soluble drugs.
Collapse
Affiliation(s)
- Syeda Sadia Batool Rizvi
- Department of Pharmaceutics, Faculty of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur 63100, Punjab, Pakistan; (S.S.B.R.); (N.A.)
| | - Naveed Akhtar
- Department of Pharmaceutics, Faculty of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur 63100, Punjab, Pakistan; (S.S.B.R.); (N.A.)
| | - Muhammad Usman Minhas
- College of Pharmacy, University of Sargodha, University Road, Sargodha 40100, Punjab, Pakistan
| | - Arshad Mahmood
- College of Pharmacy, Al Ain University, Abu Dhabi Campus, Abu Dhabi 112612, United Arab Emirates;
| | | |
Collapse
|
33
|
de Keijzer MJ, de Klerk DJ, de Haan LR, van Kooten RT, Franchi LP, Dias LM, Kleijn TG, van Doorn DJ, Heger M. Inhibition of the HIF-1 Survival Pathway as a Strategy to Augment Photodynamic Therapy Efficacy. Methods Mol Biol 2022; 2451:285-403. [PMID: 35505024 DOI: 10.1007/978-1-0716-2099-1_19] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Photodynamic therapy (PDT) is a non-to-minimally invasive treatment modality that utilizes photoactivatable drugs called photosensitizers to disrupt tumors with locally photoproduced reactive oxygen species (ROS). Photosensitizer activation by light results in hyperoxidative stress and subsequent tumor cell death, vascular shutdown and hypoxia, and an antitumor immune response. However, sublethally afflicted tumor cells initiate several survival mechanisms that account for decreased PDT efficacy. The hypoxia inducible factor 1 (HIF-1) pathway is one of the most effective cell survival pathways that contributes to cell recovery from PDT-induced damage. Several hundred target genes of the HIF-1 heterodimeric complex collectively mediate processes that are involved in tumor cell survival directly and indirectly (e.g., vascularization, glucose metabolism, proliferation, and metastasis). The broad spectrum of biological ramifications culminating from the activation of HIF-1 target genes reflects the importance of HIF-1 in the context of therapeutic recalcitrance. This chapter elaborates on the involvement of HIF-1 in cancer biology, the hypoxic response mechanisms, and the role of HIF-1 in PDT. An overview of inhibitors that either directly or indirectly impede HIF-1-mediated survival signaling is provided. The inhibitors may be used as pharmacological adjuvants in combination with PDT to augment therapeutic efficacy.
Collapse
Affiliation(s)
- Mark J de Keijzer
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, People's Republic of China
- Department of Pharmaceutics, Utrecht Institute of Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Daniel J de Klerk
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, People's Republic of China
- Laboratory of Experimental Oncology, Department of Pathology, Erasmus MC, Rotterdam, The Netherlands
| | - Lianne R de Haan
- Laboratory of Experimental Oncology, Department of Pathology, Erasmus MC, Rotterdam, The Netherlands
| | - Robert T van Kooten
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - Leonardo P Franchi
- Departamento de Bioquímica e Biologia Molecular, Instituto de Ciências Biológicas (ICB) 2, Universidade Federal de Goiás (UFG), Goiânia, GO, Brazil
- Faculty of Philosophy, Sciences, and Letters of Ribeirão Preto, epartment of Chemistry, Center of Nanotechnology and Tissue Engineering-Photobiology and Photomedicine Research Group,University of São Paulo, São Paulo, Brazil
| | - Lionel M Dias
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, People's Republic of China
- Laboratory of Experimental Oncology, Department of Pathology, Erasmus MC, Rotterdam, The Netherlands
| | - Tony G Kleijn
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, People's Republic of China
- Laboratory of Experimental Oncology, Department of Pathology, Erasmus MC, Rotterdam, The Netherlands
| | - Diederick J van Doorn
- Department of Gastroenterology and Hepatology, Amsterdam UMC, Location AMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Michal Heger
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, People's Republic of China.
- Department of Pharmaceutics, Utrecht Institute of Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands.
- Laboratory of Experimental Oncology, Department of Pathology, Erasmus MC, Rotterdam, The Netherlands.
| |
Collapse
|
34
|
Elz AS, Trevaskis NL, Porter CJH, Bowen JM, Prestidge CA. Smart design approaches for orally administered lipophilic prodrugs to promote lymphatic transport. J Control Release 2021; 341:676-701. [PMID: 34896450 DOI: 10.1016/j.jconrel.2021.12.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 12/03/2021] [Accepted: 12/04/2021] [Indexed: 12/22/2022]
Abstract
Challenges to effective delivery of drugs following oral administration has attracted growing interest over recent decades. Small molecule drugs (<1000 Da) are generally absorbed across the gastrointestinal tract into the portal blood and further transported to the systemic circulation via the liver. This can result in a significant reduction to the oral bioavailability of drugs that are metabolically labile and ultimately lead to ineffective exposure and treatment. Targeting drug delivery to the intestinal lymphatics is attracting increased attention as an alternative route of drug transportation providing multiple benefits. These include bypassing hepatic first-pass metabolism and selectively targeting disease reservoirs residing within the lymphatic system. The particular physicochemical requirements for drugs to be able to access the lymphatics after oral delivery include high lipophilicity (logP>5) and high long-chain triglyceride solubility (> 50 mg/g), properties required to enable drug association with the lipoprotein transport pathway. The majority of small molecule drugs, however, are not this lipophilic and therefore not substantially transported via the intestinal lymph. This has contributed to a growing body of investigation into prodrug approaches to deliver drugs to the lymphatic system by chemical manipulation. Optimised lipophilic prodrugs have the potential to increase lymphatic transport thereby improving oral pharmacokinetics via a reduction in first pass metabolism and may also target of disease-specific reservoirs within the lymphatics. This may provide advantages for current pharmacotherapy approaches for a wide array of pathological conditions, e.g. immune disease, cancer and metabolic disease, and also presents a promising approach for advanced vaccination strategies. In this review, specific emphasis is placed on medicinal chemistry strategies that have been successfully employed to design lipophilic prodrugs to deliberately enable lymphatic transport. Recent progress and opportunities in medicinal chemistry and drug delivery that enable new platforms for efficacious and safe delivery of drugs are critically evaluated.
Collapse
Affiliation(s)
- Aurelia S Elz
- Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia.
| | - Natalie L Trevaskis
- Department of Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC 3052, Australia.
| | - Christopher J H Porter
- Department of Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC 3052, Australia.
| | - Joanne M Bowen
- School of Biomedicine, The University of Adelaide, Adelaide, SA 5005, Australia.
| | - Clive A Prestidge
- Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia.
| |
Collapse
|
35
|
Ai X, Qiu B, Zhou Y, Li S, Li Q, Huan J, Li J, Hu N, Chen N, Liu F, Wang D, Chu C, Wang B, Chen L, Jiang H, Huang S, Huang X, Bi N, Liu H. Comparison and quantification of different concurrent chemotherapy regimens with radiotherapy in locally advanced non-small cell lung cancer: Clinical outcomes and theoretical results from an extended LQ and TCP model. Radiother Oncol 2021; 167:34-41. [PMID: 34890734 DOI: 10.1016/j.radonc.2021.11.033] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Revised: 11/13/2021] [Accepted: 11/29/2021] [Indexed: 12/25/2022]
Abstract
PURPOSE To develop a new radiobiological model and compare the efficacy of four concurrent chemotherapy regimens administered with radiotherapy in locally advanced non-small-cell lung cancer (LANSCLC) by in-field locoregional progression-free survival (LPFS). MATERIALS AND METHODS 151 LANSCLC patients were reviewed and divided into 5 groups according to their concurrent chemotherapy regimens, including 24 patients treated with radiotherapy alone, 30 treated with concurrent 4-week etoposide-cisplatin (EP), 26 with 3-week pemetrexed-cisplatin (AP), 37 with weekly paclitaxel-cisplatin (TP) and 34 with weekly docetaxel-cisplatin (DP). In-field LPFS and toxicities were compared among groups. A novel tumor control probability (TCP) model, LQRGC, incorporating four "R"s of radiobiology, Gompertzian tumor growth and chemotherapeutic effect, was related to in-field LPFS. Chemo-induced biologically effective doses (BEDs) in LQRGC/TCP model were used to quantify the concurrent chemotherapeutic efficacy. RESULTS The median follow-up time was 54.5 months. The weekly DP and 4-week EP groups had favorable median in-field LPFS (EP:46.2 months, AP:30.3 months, TP:12.2 months, DP: not reached, radiotherapy alone: 12.2 months, p = 0.001). The 4-week EP group had a higher incidence of ≥grade 3 leukopenia (EP:76.7%, AP:15.4%, TP:24.3%, DP:14.7%, radiotherapy alone: 12.5%, p < 0.001) than the other four. The LQRGC/TCP model fitted well with the in-field LPFS with the average absolute and relative fitting errors of 6.36% and 12.12%. The chemo-induced BEDs of EP, AP, TP and DP were 5.17, 0.63, 1.89 and 2.52 Gy, respectively. CONCLUSION The LQRGC/TCP model achieved promising fitting accuracy for in-field LPFS. As quantified by the model, the 4-week EP and weekly DP showed higher chemo-induced BEDs when concurrently administered with radiotherapy in LANSCLC.
Collapse
Affiliation(s)
- XinLei Ai
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, PR China; State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, PR China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, PR China
| | - Bo Qiu
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, PR China; State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, PR China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, PR China; Lung Cancer Institute of Sun Yat-sen University, Guangzhou, PR China; Guangdong Association Study of Thoracic Oncology, Guangzhou, PR China
| | - Yin Zhou
- Evidance Medical Technologies Inc., Suzhou, PR China
| | - Su Li
- Clinical Research, Sun Yat-sen University Cancer Center, Guangzhou, PR China; State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, PR China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, PR China
| | - QiWen Li
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, PR China; State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, PR China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, PR China; Lung Cancer Institute of Sun Yat-sen University, Guangzhou, PR China; Guangdong Association Study of Thoracic Oncology, Guangzhou, PR China
| | - Jian Huan
- Department of Radiation Oncology, The Affiliated Suzhou Science & Technology Town Hospital of Nanjing Medical University, Suzhou, PR China
| | - JiBin Li
- Clinical Research, Sun Yat-sen University Cancer Center, Guangzhou, PR China; State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, PR China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, PR China
| | - Nan Hu
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, PR China; State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, PR China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, PR China
| | - NaiBin Chen
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, PR China; State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, PR China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, PR China
| | - FangJie Liu
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, PR China; State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, PR China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, PR China
| | - DaQuan Wang
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, PR China; State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, PR China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, PR China
| | - Chu Chu
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, PR China; State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, PR China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, PR China
| | - Bin Wang
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, PR China; State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, PR China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, PR China
| | - Li Chen
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, PR China; State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, PR China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, PR China
| | - HaiHang Jiang
- Homology Medical Technologies Inc., Ningbo, PR China
| | - ShiYu Huang
- Department of Radiation Oncology, Chengdu Western Hospital, Chengdu, PR China
| | - XiaoYan Huang
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, PR China; State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, PR China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, PR China
| | - Nan Bi
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, PR China.
| | - Hui Liu
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, PR China; State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, PR China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, PR China; Lung Cancer Institute of Sun Yat-sen University, Guangzhou, PR China; Guangdong Association Study of Thoracic Oncology, Guangzhou, PR China.
| |
Collapse
|
36
|
Single-Shot Local Injection of Microfragmented Fat Tissue Loaded with Paclitaxel Induces Potent Growth Inhibition of Hepatocellular Carcinoma in Nude Mice. Cancers (Basel) 2021; 13:cancers13215505. [PMID: 34771667 PMCID: PMC8583409 DOI: 10.3390/cancers13215505] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Revised: 10/29/2021] [Accepted: 10/30/2021] [Indexed: 11/17/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is poorly beneficiated by intravenous chemotherapy due to inadequate availability of drugs at the tumor site. We previously demonstrated that human micro-fragmented adipose tissue (MFAT) and its devitalized counterpart (DMFAT) could be effective natural scaffolds to deliver Paclitaxel (PTX) to tumors in both in vitro and in vivo tests, affecting cancer growth relapse. Here we tested the efficacy of DMFAT-PTX in a well-established HCC in nude mice. MFAT-PTX and DMFAT-PTX preparations were tested for anti-cancer activity in 2D and 3D assays using Hep-3B tumor cells. The efficacy of DMFAT-PTX was evaluated after a single-shot subcutaneous injection near a Hep-3B growing tumor by assessing tumor volumes, apoptosis rate, and drug pharmacokinetics in an in vivo model. Potent antiproliferative activity was seen in both in vitro 2D and 3D tests. Mice treated with DMFAT-PTX (10 mg/kg) produced potent Hep-3B growth inhibition with 33% complete tumor regressions. All treated animals experienced tumor ulceration at the site of DMFAT-PTX injection, which healed spontaneously. Lowering the drug concentration (5 mg/kg) prevented the formation of ulcers, maintaining statistically significant efficacy. Histology revealed a higher number of apoptotic cancer cells intratumorally, suggesting prolonged presence of PTX that was confirmed by the pharmacokinetic analysis. DMFAT may be a potent and valid new tool for local chemotherapy of HCC in an advanced stage of progression, also suggesting potential effectiveness in other human primary inoperable cancers.
Collapse
|
37
|
Hou Y, Zhao C, Xu B, Huang Y, Liu C. Effect of docetaxel on mechanical properties of ovarian cancer cells. Exp Cell Res 2021; 408:112853. [PMID: 34597679 DOI: 10.1016/j.yexcr.2021.112853] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Revised: 09/24/2021] [Accepted: 09/26/2021] [Indexed: 10/20/2022]
Abstract
Docetaxel could inhibit the proliferation of tumor cells by targeting microtubules. The extension of cellular microtubules plays an important role in the invasion and metastasis of tumor cells. This paper aims to study the distribution and mechanical properties of cytoskeletal proteins with low concentration of docetaxel. MTT assay was used to detect the minimum drug activity concentration of docetaxel on SKOV-3 cells, fluorescence staining was used to analyze the distribution of cytoskeleton proteins, scanning electron microscopy(SEM) was used to observe the morphology of single cells, and atomic force microscopy(AFM) was used to determine the microstructure and mechanical properties of cells. The results showed that the IC10 of docetaxel was 1 ng/ml. Docetaxel can effectively inhibit the formation of cell pseudopodia, hinder the indirectness between cells, reduce the cell extension area, and make the cells malformed. In addition, when AFM analyzes the effects of drugs on cell microstructure and mechanical properties, the average cell surface roughness and cell height are positively correlated with the concentration of docetaxel. Especially when the concentration was 100 ng/ml, the adhesion decreased by 37.04% and Young's modulus increased by 1.57 times compared with the control group. This may be because docetaxel leads to microtubule remodeling and membrane protein aggregation, which affects cell microstructure and increases cell strength, leading to significant changes in the mechanical properties of ovarian cells. This is of great significance to the study of the formation mechanism of tumor cell invasion and migration activities mediated by actin.
Collapse
Affiliation(s)
- Yue Hou
- School of Life Science and Technology, Changchun University of Science and Technology, Changchun, 130022, China
| | - Chunru Zhao
- School of Life Science and Technology, Changchun University of Science and Technology, Changchun, 130022, China
| | - Binglin Xu
- School of Life Science and Technology, Changchun University of Science and Technology, Changchun, 130022, China
| | - Yuxi Huang
- International Research Centre for Nano Handling and Manufacturing of China, Changchun University of Science and Technology, Changchun, 130022, China
| | - Chuanzhi Liu
- School of Life Science and Technology, Changchun University of Science and Technology, Changchun, 130022, China; International Research Centre for Nano Handling and Manufacturing of China, Changchun University of Science and Technology, Changchun, 130022, China.
| |
Collapse
|
38
|
Machulkin AE, Uspenskaya AA, Zyk NY, Nimenko EA, Ber AP, Petrov SA, Shafikov RR, Skvortsov DA, Smirnova GB, Borisova YA, Pokrovsky VS, Kolmogorov VS, Vaneev AN, Ivanenkov YA, Khudyakov AD, Kovalev SV, Erofeev AS, Gorelkin PV, Beloglazkina EK, Zyk NV, Khazanova ES, Majouga AG. PSMA-targeted small-molecule docetaxel conjugate: Synthesis and preclinical evaluation. Eur J Med Chem 2021; 227:113936. [PMID: 34717125 DOI: 10.1016/j.ejmech.2021.113936] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 10/17/2021] [Accepted: 10/18/2021] [Indexed: 12/11/2022]
Abstract
Prostate cancer is one of the most commonly diagnosed men's cancers and remains one of the leading causes of cancer death. The development of approaches to the treatment of this oncological disease is an ongoing process. In this work, we have carried out the selection of ligands for the creation of conjugates based on the drug docetaxel and synthesized a series of three docetaxel conjugates. In vitro cytotoxicity of these molecules was evaluated using the MTT assay. Based on the assay results, we selected the conjugate which showed cytotoxic potential close to unmodified docetaxel. At the same time, the molar solubility of the resulting compound increased up to 20 times in comparison with the drug itself. In vivo evaluation on 22Rv1 (PSMA+) xenograft model demonstrated a good potency of the synthesized conjugate to inhibit tumor growth: the inhibition turned out to be more than 80% at a dose of 30 mg/kg. Pharmacokinetic parameters of conjugate distribution were analyzed. Also, it was found that PSMA-targeted docetaxel conjugate is less toxic than docetaxel itself, the decrease of molar acute toxicity in comparison with free docetaxel was up to 20%. Obtained conjugate PSMA-DOC is a good candidate for further expanded preclinical trials because of high antitumor activity, fewer side toxic effects and better solubility.
Collapse
Affiliation(s)
- Aleksei E Machulkin
- Lomonosov Moscow State University, Chemistry Dept., Leninskie Gory, Building 1/3, GSP-1, Moscow, 119991, Russian Federation.
| | - Anastasia A Uspenskaya
- Lomonosov Moscow State University, Chemistry Dept., Leninskie Gory, Building 1/3, GSP-1, Moscow, 119991, Russian Federation
| | - Nikolay Y Zyk
- Lomonosov Moscow State University, Chemistry Dept., Leninskie Gory, Building 1/3, GSP-1, Moscow, 119991, Russian Federation
| | - Ekaterina A Nimenko
- Lomonosov Moscow State University, Chemistry Dept., Leninskie Gory, Building 1/3, GSP-1, Moscow, 119991, Russian Federation
| | - Anton P Ber
- Lomonosov Moscow State University, Chemistry Dept., Leninskie Gory, Building 1/3, GSP-1, Moscow, 119991, Russian Federation
| | - Stanislav A Petrov
- Lomonosov Moscow State University, Chemistry Dept., Leninskie Gory, Building 1/3, GSP-1, Moscow, 119991, Russian Federation
| | - Radik R Shafikov
- Lomonosov Moscow State University, Chemistry Dept., Leninskie Gory, Building 1/3, GSP-1, Moscow, 119991, Russian Federation; Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry Russian Academy of Sciences, GSP-7, Ulitsa Miklukho-Maklaya, 16/10, Moscow, 117997, Russian Federation
| | - Dmitry A Skvortsov
- Lomonosov Moscow State University, Chemistry Dept., Leninskie Gory, Building 1/3, GSP-1, Moscow, 119991, Russian Federation; Faculty of Biology and Biotechnologies, Higher School of Economics, Myasnitskaya 13, Moscow, 101000, Russia
| | - Galina B Smirnova
- N.N. Blokhin Cancer Research Center, 24 Kashirskoye Sh., Moscow, 115478, Russia
| | - Yulia A Borisova
- N.N. Blokhin Cancer Research Center, 24 Kashirskoye Sh., Moscow, 115478, Russia
| | - Vadim S Pokrovsky
- N.N. Blokhin Cancer Research Center, 24 Kashirskoye Sh., Moscow, 115478, Russia; RUDN University, Miklukho-Maklaya Str.6, Moscow, 117198, Russian Federation
| | - Vasilii S Kolmogorov
- National University of Science and Technology MISiS, 9 Leninskiy Pr, Moscow, 119049, Russian Federation
| | - Alexander N Vaneev
- National University of Science and Technology MISiS, 9 Leninskiy Pr, Moscow, 119049, Russian Federation
| | - Yan A Ivanenkov
- Lomonosov Moscow State University, Chemistry Dept., Leninskie Gory, Building 1/3, GSP-1, Moscow, 119991, Russian Federation; Moscow Institute of Physics and Technology (State University), 9 Institutskiy Lane, Dolgoprudny City, Moscow Region, 141700, Russian Federation; National University of Science and Technology MISiS, 9 Leninskiy Pr, Moscow, 119049, Russian Federation; The Federal State Unitary Enterprise Dukhov Automatics Research Institute, Moscow, 127055, Russia; Institute of Biochemistry and Genetics Ufa Science Centre Russian Academy of Sciences (IBG RAS), Oktyabrya Prospekt 71, Ufa, 450054, Russian Federation
| | - Alexander D Khudyakov
- Lomonosov Moscow State University, Chemistry Dept., Leninskie Gory, Building 1/3, GSP-1, Moscow, 119991, Russian Federation
| | - Sergei V Kovalev
- Lomonosov Moscow State University, Chemistry Dept., Leninskie Gory, Building 1/3, GSP-1, Moscow, 119991, Russian Federation
| | - Alexander S Erofeev
- National University of Science and Technology MISiS, 9 Leninskiy Pr, Moscow, 119049, Russian Federation
| | - Petr V Gorelkin
- National University of Science and Technology MISiS, 9 Leninskiy Pr, Moscow, 119049, Russian Federation
| | - Elena K Beloglazkina
- Lomonosov Moscow State University, Chemistry Dept., Leninskie Gory, Building 1/3, GSP-1, Moscow, 119991, Russian Federation
| | - Nikolay V Zyk
- Lomonosov Moscow State University, Chemistry Dept., Leninskie Gory, Building 1/3, GSP-1, Moscow, 119991, Russian Federation
| | - Elena S Khazanova
- LLC Izvarino-Pharma, V. Vnukovskoe, Vnukovskoe Sh., 5th Km., Building 1, Moscow, 108817, Russian Federation
| | - Alexander G Majouga
- Lomonosov Moscow State University, Chemistry Dept., Leninskie Gory, Building 1/3, GSP-1, Moscow, 119991, Russian Federation; National University of Science and Technology MISiS, 9 Leninskiy Pr, Moscow, 119049, Russian Federation; Dmitry Mendeleev University of Chemical Technology of Russia, Miusskaya Sq. 9, Moscow, 125047, Russian Federation
| |
Collapse
|
39
|
Polysorbate-Based Drug Formulations for Brain-Targeted Drug Delivery and Anticancer Therapy. APPLIED SCIENCES-BASEL 2021. [DOI: 10.3390/app11199336] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Polysorbates (PSs) are synthetic nonionic surfactants consisting of polyethoxy sorbitan fatty acid esters. PSs have been widely employed as emulsifiers and stabilizers in various drug formulations and food additives. Recently, various PS-based formulations have been developed for safe and efficient drug delivery. This review introduces the general features of PSs and PS-based drug carriers, summarizes recent progress in the development of PS-based drug formulations, and discusses the physicochemical properties, biological safety, P-glycoprotein inhibitory properties, and therapeutic applications of PS-based drug formulations. Additionally, recent advances in brain-targeted drug delivery using PS-based drug formulations have been highlighted. This review will help researchers understand the potential of PSs as effective drug formulation agents.
Collapse
|
40
|
Konoshenko M, Laktionov P. The miRNAs involved in prostate cancer chemotherapy response as chemoresistance and chemosensitivity predictors. Andrology 2021; 10:51-71. [PMID: 34333834 DOI: 10.1111/andr.13086] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 07/09/2021] [Accepted: 07/30/2021] [Indexed: 12/11/2022]
Abstract
BACKGROUND Reliable molecular markers that allow the rational prescription of an effective chemotherapy type for each prostate cancer patient are still needed. Since microRNAs expression is associated with the response to different types of prostate cancer therapy, microRNAs represent a pool of perspective markers of therapy effectiveness comprising chemotherapy. OBJECTIVES The available data on microRNAs associated with chemotherapy response (resistance and sensitivity) are summarized and analyzed in the article. MATERIALS AND METHODS A review of the published data, as well as their analysis by current bioinformatics resources, was conducted. The molecular targets of microRNAs, as well as the reciprocal relationships between the microRNAs and their targets, were studied using the DIANA, STRING, and TransmiR databases. Special attention was dedicated to the mechanisms of prostate cancer chemoresistance development. RESULTS AND DISCUSSION The combined analysis of bioinformatics resources and the available literature indicated that the expression of eight microRNAs that are associated with different responses to chemotherapy have a high potential for the prediction of the prostate cancer chemotherapy response, as found in the experiments and confirmed by the functions of regulated genes. CONCLUSION An overview on the published data and bioinformatics resources, with respect to predictive microRNA markers of chemotherapy response, is presented in this review. The selected microRNA and gene panel has a high potential for predicting the chemosensitivity or chemoresistance of prostate cancer and could represent a set of markers for subsequent study using samples of cell-free microRNAs from different patient groups.
Collapse
Affiliation(s)
- Maria Konoshenko
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch, Russian Academy of Sciences, Novosibirsk, Russia
| | - Pavel Laktionov
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch, Russian Academy of Sciences, Novosibirsk, Russia
| |
Collapse
|
41
|
Yiding C, Zhongyi H. Differences between the quality aspects of various generic and branded docetaxel formulations. Curr Med Res Opin 2021; 37:1421-1433. [PMID: 33998944 DOI: 10.1080/03007995.2021.1929895] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Docetaxel is a widely prescribed chemotherapy drug in oncology and post expiry of its patent, the drug has been marketed as a generic by multiple manufacturers. It is also classified as a narrow therapeutic window drug. Through enhanced permeability and retention effect, polymeric micelles can passively target agents to tumor tissue, thereby decreasing toxicity of the drug in normal tissue. However, various studies have raised concerns that generic docetaxel leads to greater incidences of toxicities among patients with cancer. Thus, we herein review the pharmaceutical challenges associated with different docetaxel formulations and provide insights into the dissimilarities in the quality and safety of branded and generic docetaxel formulations. Literature review reported that 90% of the generic formulations of docetaxel contain inadequate quantity of active drug and high levels of impurities. Higher amounts of solvents such as polysorbate 80 and ethanol in docetaxel formulation lead to severe toxicities such as febrile neutropenia, hematological, and cutaneous toxicities. In most of the studies, even minor changes in excipients, solvents, unbound fraction of docetaxel were associated with adverse events, while in few, the source of docetaxel remained unidentified. One study reported incidence of febrile neutropenia due to a switch in the formulation from branded to generic. The quality, safety, and efficacy of medicines will directly affect the life of patients, and therefore, use of docetaxel formulations that guarantee safety of patients are the necessity of the hour. Being an injectable anti-cancer drug, it is important to determine the consistency of the various formulations of docetaxel globally, conduct bioequivalence studies as per the regulatory standards, taking into account the permissible limits of the excipients or the presence of such unapproved excipients.
Collapse
Affiliation(s)
- Chen Yiding
- Department of Breast Surgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang, China
| | - Huang Zhongyi
- The GCP Office, The Central Hospital of Jing'an District, Shanghai, China
| |
Collapse
|
42
|
Wu Q, Siddharth S, Sharma D. Triple Negative Breast Cancer: A Mountain Yet to Be Scaled Despite the Triumphs. Cancers (Basel) 2021; 13:3697. [PMID: 34359598 PMCID: PMC8345029 DOI: 10.3390/cancers13153697] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 07/13/2021] [Accepted: 07/18/2021] [Indexed: 12/12/2022] Open
Abstract
Metastatic progression and tumor recurrence pertaining to TNBC are certainly the leading cause of breast cancer-related mortality; however, the mechanisms underlying TNBC chemoresistance, metastasis, and tumor relapse remain somewhat ambiguous. TNBCs show 77% of the overall 4-year survival rate compared to other breast cancer subtypes (82.7 to 92.5%). TNBC is the most aggressive subtype of breast cancer, with chemotherapy being the major approved treatment strategy. Activation of ABC transporters and DNA damage response genes alongside an enrichment of cancer stem cells and metabolic reprogramming upon chemotherapy contribute to the selection of chemoresistant cells, majorly responsible for the failure of anti-chemotherapeutic regime. These selected chemoresistant cells further lead to distant metastasis and tumor relapse. The present review discusses the approved standard of care and targetable molecular mechanisms in chemoresistance and provides a comprehensive update regarding the recent advances in TNBC management.
Collapse
Affiliation(s)
| | - Sumit Siddharth
- Department of Oncology, Johns Hopkins University School of Medicine and the Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD 21231, USA;
| | - Dipali Sharma
- Department of Oncology, Johns Hopkins University School of Medicine and the Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD 21231, USA;
| |
Collapse
|
43
|
Docetaxel encapsulation in nanoscale assembly micelles of folate-PEG-docetaxel conjugates for targeted fighting against metastatic breast cancer in vitro and in vivo. Int J Pharm 2021; 605:120822. [PMID: 34182039 DOI: 10.1016/j.ijpharm.2021.120822] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 06/16/2021] [Accepted: 06/20/2021] [Indexed: 01/17/2023]
Abstract
Due to the high frequency and mortality of breast cancer, developing efficient targeted drug delivery systems for frightening against this malignancy is among cancer research priorities. The aim of this study was to synthesize a targeted micellar formulation of docetaxel (DTX) using DTX, folic acid (FA) and polyethylene glycol (PEG) conjugates as building blocks. In the current study, two therapeutic polymers consisting of FA-PEG-DTX and PEG-DTX conjugates were synthesized and implemented to form folate-targeted and non-targeted micelles. Dissipative particle dynamics (DPD) method was used to simulate the behavior of the nanoparticle. The anti-cancer drug, DTX was loaded in to the micelles via solvent switching method in order to increase its solubility and stability. The cytotoxicity of the targeted and non-targeted formulations was evaluated against 4T1 and CHO cell lines. In vivo therapeutic efficiency was studied using ectopic tumor model of metastatic breast cancer, 4T1, in Female BALB/c mice. The successful synthesis of therapeutic polymers, FA-PEG-DTX and PEG-DTX were confirmed implementing 1HNMR spectral analysis. The size of DTX-loaded non-targeted and targeted micelles were 176.3 ± 8.3 and 181 ± 10.1 nm with PDI of 0.23 and 0.17, respectively. Loading efficiencies of DTX in non-targeted and targeted micelles were obtained to be 85% and 82%, respectively. In vitro release study at pH = 7.4 and pH = 5.4 showed a controlled and continuous drug release for both formulations, that was faster at pH = 5.4 (100% drug release within 120 h) than at pH = 7.4 (80% drug release within 150 h). The targeted formulation showed a significant higher cytotoxicity against 4T1 breast cancer cells (high expression of folate receptor) within the range of 12.5 to 200 μg/mL in comparison with no-targeted one. However, there was no significant difference between the cytotoxicity of the targeted and non-targeted formulations against CHO cell line as low-expressed cell line. In accordance with in vitro investigation, in vivo studies verified the ideal anti-tumor efficacy of the targeted formulation compared to Taxotere and non-targeted formulation. Based on the obtained data, FA-targeted DTX-loaded nano-micelles significantly increased the therapeutic efficacy of DTX and therefore can be considered as a new potent platform for breast cancer chemotherapy.
Collapse
|
44
|
Lee TL, Wei PY, Yang MH, Chang PMH, Wang LW, Tai SK. Tongue conservation treatment for oral tongue squamous cell carcinoma with induction chemotherapy, surgery, and risk-adapted adjuvant therapy: A phase II trial. Cancer Rep (Hoboken) 2021; 5:e1456. [PMID: 34051137 PMCID: PMC8842695 DOI: 10.1002/cnr2.1456] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 03/29/2021] [Accepted: 04/28/2021] [Indexed: 11/07/2022] Open
Abstract
BACKGROUND To assess the feasibility of tongue conservation treatment with induction chemotherapy (ICT), tongue conservation surgery, and risk-adapted postoperative adjuvant therapy in oral tongue squamous cell carcinoma (OTSCC). METHODS Patients with newly diagnosed OTSCC cT2-4 N0-2 M0 were recruited. The ICT with a regimen of docetaxel, cisplatin, and oral tegafur/uracil (DCU) was administrated every 21 days. After the first cycle of ICT (DCU1), patients with a more than 30% decrease in the longest diameter of primary tumor underwent a second cycle of ICT (DCU2). Tongue conservation surgery was performed after ICT, and risk-adapted adjuvant therapy was organized based on pathological features. RESULTS From July 2011 to December 2015, a total of 23 patients were enrolled, 87% of whom were classified as stage III-IV. Clinical responders to DCU1 and DCU2 were determined in 90.5% (19/21) and 88.2% (15/17) of patients. Tongue conservation surgery was performed in 16 responders to ICT. Only one patient had a positive margin (6.3%), and a complete pathologic response was achieved in eight patients (50%). Only one patient developed local recurrence after a median follow-up of 58.6 months (range, 7.9-105.2). The 5-year overall survival (0% vs. 87.5%, P = 0.001) and disease-specific survival (0% vs. 93.3%, P = 0.000) were significantly different between the DCU1 nonresponders and responders. CONCLUSION Tongue conservation treatment with ICT, followed by conservation surgery and risk-adapted adjuvant therapy, is feasible for patients with OTSCC who are good responders to ICT. However, the outcomes of nonresponders are dismal. Further study in a larger patient population is warranted.
Collapse
Affiliation(s)
- Tsung-Lun Lee
- Department of Otolaryngology, National Yang Ming Chiao Tung University, Taipei, Taiwan.,Department of Otolaryngology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Pei-Yin Wei
- Department of Otolaryngology, National Yang Ming Chiao Tung University, Taipei, Taiwan.,Department of Otolaryngology, Taoyuan General Hospital, Ministry of Health and Welfare, Taoyuan, Taiwan
| | - Muh-Hwa Yang
- Infection and Immunity Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan.,Oncology, Division of Medical Oncology, Taipei Veterans General Hospital, Taipei, Taiwan.,Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Peter Mu-Hsin Chang
- Oncology, Division of Medical Oncology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Ling-Wei Wang
- Oncology, Division of Radiation Oncology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Shyh-Kuan Tai
- Department of Otolaryngology, National Yang Ming Chiao Tung University, Taipei, Taiwan.,Department of Otolaryngology, Taipei Veterans General Hospital, Taipei, Taiwan.,Infection and Immunity Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan
| |
Collapse
|
45
|
Choi HY, Ruel I, Genest J. Identification of Docetaxel as a Potential Drug to Promote HDL Biogenesis. Front Pharmacol 2021; 12:679456. [PMID: 34093205 PMCID: PMC8176524 DOI: 10.3389/fphar.2021.679456] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 05/07/2021] [Indexed: 12/13/2022] Open
Abstract
Objective: Our recent studies showed that desmocollin 1 (DSC1) binds to apoA-I in order to inhibit apoA-I-mediated high density lipoprotein (HDL) biogenesis in atherosclerotic plaques. To promote HDL biogenesis in the plaque, here we search for small molecules that block apoA-I-DSC1 interactions. Approach and Results: We combined mutational and computational mapping methods to show that amino acid residues 442-539 in the mature DSC1 protein form an apoA-I binding site (AIBS). Using a crystal structure of the AIBS, we carried out virtual screening of 10 million small molecules to estimate their binding affinities to the AIBS, followed by the selection of 51 high-affinity binding molecules as potential inhibitors of apoA-I-DSC1 interactions. Among the 51, the chemotherapy drug docetaxel showed the highest potency in promoting apoA-I-mediated HDL biogenesis in primary human skin fibroblasts with the half-maximal effective concentration of 0.72 nM. In silico docking studies suggest that the taxane ring in docetaxel binds to the AIBS and that the carbon-13 sidechain of the taxane tightens/stabilizes the binding. The HDL biogenic effect of docetaxel was also observed in two predominant cell types in atherosclerosis, macrophages and smooth muscle cells. Importantly, docetaxel promoted HDL biogenesis at concentrations much lower than those required for inducing cytotoxicity. Conclusion: Determination of the AIBS in DSC1 and AIBS structure-based virtual screening allowed us to identify docetaxel as a strong HDL biogenic agent. With the remarkable potency in promoting HDL biogenesis, a chemotherapy drug docetaxel may be repurposed to enhance atheroprotective HDL functions.
Collapse
Affiliation(s)
- Hong Y Choi
- Cardiovascular Research Laboratories, Research Institute of the McGill University Health Center, Montreal, QC, Canada
| | - Isabelle Ruel
- Cardiovascular Research Laboratories, Research Institute of the McGill University Health Center, Montreal, QC, Canada
| | - Jacques Genest
- Cardiovascular Research Laboratories, Research Institute of the McGill University Health Center, Montreal, QC, Canada
| |
Collapse
|
46
|
Optimum multi-drug regime for compartment model of tumour: cell-cycle-specific dynamics in the presence of resistance. J Pharmacokinet Pharmacodyn 2021; 48:543-562. [PMID: 33751365 DOI: 10.1007/s10928-021-09749-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 03/09/2021] [Indexed: 12/19/2022]
Abstract
This work is focused on multi-objective optimisation of a multi-drug chemotherapy schedule for cell-cycle-specific cancer treatment under the influence of drug resistance. The acquired drug resistance to chemotherapeutic agents is incorporated into the existing compartmental model of breast cancer. Furthermore, the toxic effect of drugs on healthy cells and overall drug concentration in the patient body are also constrained in the proposed model. The objective is to determine the optimal drug schedule according to the patient's physiological condition so that the tumour burden is minimised. A multi-objective optimisation algorithm, non-dominated sorting genetic algorithm-II (NSGA-II) is utilised to solve the problem. The obtained results are thoroughly analysed to illustrate the impact of drug resistance on the treatment. The capability of optimised schedules to deal with parametric uncertainty is also analysed. The drug schedules obtained in this work align well with the clinical standards. It is also revealed that the NSGA-II optimised drug schedule with proper rest period between successive dosages yields the minimum cancer load at the end of the treatment.
Collapse
|
47
|
Motono N, Ueda Y, Shimasaki M, Iwai S, Iijima Y, Usuda K, Uramoto H. Prognostic Impact of Sphingosine Kinase 1 in Nonsmall Cell Lung Cancer. CLINICAL PATHOLOGY 2021; 14:2632010X20988531. [PMID: 33623898 PMCID: PMC7879003 DOI: 10.1177/2632010x20988531] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Accepted: 12/25/2020] [Indexed: 12/02/2022]
Abstract
Bioactive sphingolipid is clearly relevant to lung physiology. The relationship of the bioactive sphingolipid pathway to pulmonary disease has been studied in cellular, tissue, and animal model, including lung cancer models. The samples of 53 patients diagnosed with nonsmall cell lung carcinoma (NSCLC) between June 2009 and May 2014 at our hospital were analyzed. Immunohistochemical (IHC) analysis was performed. The degree of immunostaining was reviewed and scored. Using this method of assessment, we evaluated the IHC score of sphingosine kinase 1 (SPHK1), vimentin, E-cadherin, and Ki-67. Both invasive adenocarcinoma cell and squamous cell carcinoma cell were well stained by SPHK1, and fibroblasts were also well stained by SPHK1. Although the IHC score of SPHK1 was not significantly differed between invasive adenocarcinoma and squamous cell carcinoma, the IHC scores of fibroblast, vimentin, and Ki-67 were higher in squamous cell carcinoma than invasive adenocarcinoma. Correlation among IHC scores in each of invasive adenocarcinoma and squamous cell carcinoma was performed. SPHK1 had positive correlation with both fibroblast and Ki-67, and fibroblast and Ki-67 had also positive correlation in invasive adenocarcinoma. On the contrary, SPHK1 had no significant correlation with fibroblast, and had negative correlation with Ki-67 in squamous cell carcinoma. Although there was not significant prognostic difference in SPHK1 score (P = .09), IHC score high group tended to be worse on relapse-free survival. SPHK1 might be prognostic factor in lung-invasive adenocarcinoma and novel target for drug against lung-invasive adenocarcinoma.
Collapse
Affiliation(s)
- Nozomu Motono
- Department of Thoracic Surgery, Kanazawa Medical University, Uchinada, Japan
| | - Yoshimichi Ueda
- Department of Pathology II, Kanazawa Medical University, Uchinada, Japan
| | - Miyako Shimasaki
- Department of Pathology II, Kanazawa Medical University, Uchinada, Japan
| | - Shun Iwai
- Department of Thoracic Surgery, Kanazawa Medical University, Uchinada, Japan
| | - Yoshihito Iijima
- Department of Thoracic Surgery, Kanazawa Medical University, Uchinada, Japan
| | - Katsuo Usuda
- Department of Thoracic Surgery, Kanazawa Medical University, Uchinada, Japan
| | - Hidetaka Uramoto
- Department of Thoracic Surgery, Kanazawa Medical University, Uchinada, Japan
| |
Collapse
|
48
|
A. Razak SA, Mohd Gazzali A, Fisol FA, M. Abdulbaqi I, Parumasivam T, Mohtar N, A. Wahab H. Advances in Nanocarriers for Effective Delivery of Docetaxel in the Treatment of Lung Cancer: An Overview. Cancers (Basel) 2021; 13:400. [PMID: 33499040 PMCID: PMC7865793 DOI: 10.3390/cancers13030400] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 12/09/2020] [Accepted: 12/24/2020] [Indexed: 12/24/2022] Open
Abstract
Docetaxel (DCX) is a highly effective chemotherapeutic drug used in the treatment of different types of cancer, including non-small cell lung cancer (NSCLC). The drug is known to have low oral bioavailability due to its low aqueous solubility, poor membrane permeability and susceptibility to hepatic first-pass metabolism. To mitigate these problems, DCX is administered via the intravenous route. Currently, DCX is commercially available as a single vial that contains polysorbate 80 and ethanol to solubilize the poorly soluble drug. However, this formulation causes short- and long-term side effects, including hypersensitivity, febrile neutropenia, fatigue, fluid retention, and peripheral neuropathy. DCX is also a substrate to the drug efflux pump P-glycoprotein (P-gp) that would reduce its concentration within the vicinity of the cells and lead to the development of drug resistance. Hence, the incorporation of DCX into various nanocarrier systems has garnered a significant amount of attention in recent years to overcome these drawbacks. The surfaces of these drug-delivery systems indeed can be functionalized by modification with different ligands for smart targeting towards cancerous cells. This article provides an overview of the latest nanotechnological approaches and the delivery systems that were developed for passive and active delivery of DCX via different routes of administration for the treatment of lung cancer.
Collapse
Affiliation(s)
- S. Aishah A. Razak
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, Minden, Penang 11800, Malaysia; (S.A.A.R.); (F.A.F.); (I.M.A.); (T.P.); (N.M.)
| | - Amirah Mohd Gazzali
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, Minden, Penang 11800, Malaysia; (S.A.A.R.); (F.A.F.); (I.M.A.); (T.P.); (N.M.)
| | - Faisalina Ahmad Fisol
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, Minden, Penang 11800, Malaysia; (S.A.A.R.); (F.A.F.); (I.M.A.); (T.P.); (N.M.)
- Malaysian Institute of Pharmaceuticals and Nutraceuticals (IPharm), National Institute of Biotechnology Malaysia (NIBM), Ministry of Science, Technology and Innovation (MOSTI), Gelugor, Penang 11700, Malaysia
| | - Ibrahim M. Abdulbaqi
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, Minden, Penang 11800, Malaysia; (S.A.A.R.); (F.A.F.); (I.M.A.); (T.P.); (N.M.)
| | - Thaigarajan Parumasivam
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, Minden, Penang 11800, Malaysia; (S.A.A.R.); (F.A.F.); (I.M.A.); (T.P.); (N.M.)
| | - Noratiqah Mohtar
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, Minden, Penang 11800, Malaysia; (S.A.A.R.); (F.A.F.); (I.M.A.); (T.P.); (N.M.)
| | - Habibah A. Wahab
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, Minden, Penang 11800, Malaysia; (S.A.A.R.); (F.A.F.); (I.M.A.); (T.P.); (N.M.)
| |
Collapse
|
49
|
Muth M, Ojara FW, Kloft C, Joerger M. Role of TDM-based dose adjustments for taxane anticancer drugs. Br J Clin Pharmacol 2020; 87:306-316. [PMID: 33247980 DOI: 10.1111/bcp.14678] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 09/10/2020] [Accepted: 11/03/2020] [Indexed: 01/14/2023] Open
Abstract
The classical taxanes (paclitaxel, docetaxel), the newer taxane cabazitaxel and the nanoparticle-bound nab-paclitaxel are among the most widely used anticancer drugs. Still, the optimal use and the value of pharmacological personalization of the taxanes is still controversial. We give an overview on the pharmacological properties of the taxanes, including metabolism, pharmacokinetics-pharmacodynamic relations and aspects in the clinical use of taxanes. The latter includes the ongoing debate on the most effective and safe regimen, the recommended initial dose, and pharmacological dosing individualization. The taxanes are among the most widely used anticancer drugs in patients with solid malignancies. Despite their longtime use in clinical routine, the optimal dosing strategy (weekly versus 3-weekly) or optimal average dose (cabazitaxel, nab-paclitaxel) has not been fully resolved, as it may differ according to tumour entity and line of treatment. The value of pharmacological individualization of the taxanes (TDM, TCI) has been partly explored for 3-weekly paclitaxel and docetaxel, but remains mostly unexplored for cabazitaxel and nab-paclitaxel at present.
Collapse
Affiliation(s)
- Marsilla Muth
- Department of Oncology & Hematology, Cantonal Hospital, St. Gallen, Switzerland
| | - Francis Williams Ojara
- Department of Clinical Pharmacy and Biochemistry, Institute of Pharmacy, Freie Universität Berlin, Germany.,Graduate Research Training Program PharMetrX, Germany
| | - Charlotte Kloft
- Department of Clinical Pharmacy and Biochemistry, Institute of Pharmacy, Freie Universität Berlin, Germany
| | - Markus Joerger
- Department of Oncology & Hematology, Cantonal Hospital, St. Gallen, Switzerland
| |
Collapse
|
50
|
Priyadarshini R, Raj GM, Sundaram R, Kayal S, Ramesh A, Shewade DG. Association of plasma docetaxel levels with ABCB1 gene polymorphisms and tumour response in locally advanced breast cancer patients of South India on neo-adjuvant chemotherapy. Breast Cancer 2020; 28:414-423. [PMID: 33125673 DOI: 10.1007/s12282-020-01177-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Accepted: 10/13/2020] [Indexed: 11/29/2022]
Abstract
BACKGROUND Genetic factors could be attributed to the variability in docetaxel plasma levels and its subsequent therapeutic response. The objectives of this study were to assess the effect of ABCB1 gene polymorphisms [SNPs rs1045642 (C3435T) and rs1128503 (C1236T)] on docetaxel plasma levels and also to analyze the influence of docetaxel plasma levels on tumour response in the ethnically distinct South Indian population. METHODS 104 locally advanced breast cancer (LABC) patients on docetaxel-based neo-adjuvant chemotherapy (NACT) were included. The plasma docetaxel levels were estimated using the validated reverse phase liquid chromatography with mass spectrometry (LC-MS/MS). DNA was extracted (phenol-chloroform extraction method) and the real-time PCR system using validated TaqMan® SNP genotyping assay method was used for genotyping. Tumour response was assessed by RECIST criteria based on the MRI images. RESULTS Patients with "CT/TT" genotype of the SNP C1236T had a C0/Ct ratio of 1.6 times higher than those with "CC" genotype (13.5 ± 6.5 vs 8.3 ± 3.1, p = 0.002). Though not significant, patients with "CT/TT" genotype had greater initial plasma concentration (C0) and area under the plasma concentration-time curve (AUC0-t). Conversely, the SNP C3435T was not associated with the plasma docetaxel levels. Furthermore, the C0 and normalized C0 were found to be higher in tumour responders compared to non-responders (p < 0.05). CONCLUSIONS The plasma levels of docetaxel were significantly influenced by the SNP C1236T of ABCB1 gene coding for the MDR1 transporter (P-glycoprotein). The plasma levels of docetaxel were also found to influence its therapeutic effect.
Collapse
Affiliation(s)
- Rekha Priyadarshini
- Department of Pharmacology, Indira Gandhi Medical College & Research Institute (IGMC & RI), Puducherry, 605009, India.
| | - Gerard Marshall Raj
- Department of Pharmacology, Sri Venkateshwaraa Medical College Hospital and Research Centre (SVMCH & RC), Puducherry, 605102, India
| | - Rajan Sundaram
- Department of Pharmacology, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Puducherry, 605006, India
| | - Smita Kayal
- Department of Medical Oncology, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Puducherry, 605006, India
| | - Ananthakrishnan Ramesh
- Department of Radio-diagnosis, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Puducherry, 605006, India
| | - Deepak Gopal Shewade
- Department of Pharmacology, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Puducherry, 605006, India
| |
Collapse
|