1
|
Sarwar S, Morozov VM, Newcomb MA, Yan B, Brant JO, Opavsky R, Guryanova OA, Ishov AM. Overcoming ABCB1 mediated multidrug resistance in castration resistant prostate cancer. Cell Death Dis 2024; 15:558. [PMID: 39090086 PMCID: PMC11294535 DOI: 10.1038/s41419-024-06949-3] [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: 04/08/2024] [Revised: 07/19/2024] [Accepted: 07/24/2024] [Indexed: 08/04/2024]
Abstract
Prostate cancer (PCa) is the second leading cause of cancer-related death in American men. PCa that relapses after hormonal therapies, referred to as castration resistant PCa (CRPC), often presents with metastases (mCRPC) that are the major cause of mortality. The few available therapies for mCRPC patients include taxanes docetaxel (DTX) and cabazitaxel (CBZ). However, development of resistance limits their clinical use. Mechanistically, resistance arises through upregulation of multidrug resistance (MDR) proteins such as MDR1/ABCB1, making ABCB1 an attractive therapeutic target. Yet, ABCB1 inhibitors failed to be clinically useful due to low specificity and toxicity issues. To study taxanes resistance, we produced CBZ resistant C4-2B cells (RC4-2B) and documented resistance to both CBZ and DTX in cell culture and in 3D prostaspheres settings. RNAseq identified increased expression of ABCB1 in RC4-2B, that was confirmed by immunoblotting and immunofluorescent analysis. ABCB1-specific inhibitor elacridar reversed CBZ and DTX resistance in RC4-2B cells, confirming ABCB1-mediated resistance mechanism. In a cell-based screen using a curated library of cytotoxic drugs, we found that DNA damaging compounds Camptothecin (CPT) and Cytarabine (Ara-C) overcame resistance as seen by similar cytotoxicity in parental C4-2B and resistant RC4-2B. Further, these compounds were cytotoxic to multiple PC cells resistant to taxanes with high ABCB1 expression and, therefore, can be used to conquer the acquired resistance to taxanes in PCa. Finally, inhibition of cyclin-dependent kinases 4/6 (CDK4/6) with small molecule inhibitors (CDK4/6i) potentiated cytotoxic effect of CPT or Ara-C in both parental and resistant cells. Overall, our findings indicate that DNA damaging agents CPT and Ara-C alone or in combination with CDK4/6i can be suggested as a new treatment regimen in CRPC patients, including those that are resistant to taxanes.
Collapse
Affiliation(s)
- Sadia Sarwar
- Department of Anatomy and Cell Biology, University of Florida College of Medicine, Gainesville, FL, USA
| | - Viacheslav M Morozov
- Department of Anatomy and Cell Biology, University of Florida College of Medicine, Gainesville, FL, USA
| | - Mallory A Newcomb
- Department of Anatomy and Cell Biology, University of Florida College of Medicine, Gainesville, FL, USA
| | - Bowen Yan
- Department of Pharmacology and Therapeutics, University of Florida College of Medicine, Gainesville, FL, USA
| | - Jason O Brant
- Department of Biostatistics, University of Florida College of Medicine, Gainesville, FL, USA
- University of Florida Health Cancer Center, Gainesville, FL, USA
| | - Rene Opavsky
- Department of Anatomy and Cell Biology, University of Florida College of Medicine, Gainesville, FL, USA
- University of Florida Health Cancer Center, Gainesville, FL, USA
| | - Olga A Guryanova
- Department of Pharmacology and Therapeutics, University of Florida College of Medicine, Gainesville, FL, USA
- University of Florida Health Cancer Center, Gainesville, FL, USA
| | - Alexander M Ishov
- Department of Anatomy and Cell Biology, University of Florida College of Medicine, Gainesville, FL, USA.
- University of Florida Health Cancer Center, Gainesville, FL, USA.
| |
Collapse
|
2
|
VAGELI DIMITRAP, DOUKAS PANAGIOTISG, GOUPOU KERASIA, BENOS ANTONIOSD, ASTARA KYRIAKI, ZACHAROULI KONSTANTINA, SOTIRIOU SOTIRIS, IOANNOU MARIA. Hypoxia-inducible factor 1alpha and vascular endothelial growth factor in Glioblastoma Multiforme: a systematic review going beyond pathologic implications. Oncol Res 2024; 32:1239-1256. [PMID: 39055895 PMCID: PMC11267112 DOI: 10.32604/or.2024.052130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Accepted: 05/23/2024] [Indexed: 07/28/2024] Open
Abstract
Glioblastoma multiforme (GBM) is an aggressive primary brain tumor characterized by extensive heterogeneity and vascular proliferation. Hypoxic conditions in the tissue microenvironment are considered a pivotal player leading tumor progression. Specifically, hypoxia is known to activate inducible factors, such as hypoxia-inducible factor 1alpha (HIF-1α), which in turn can stimulate tumor neo-angiogenesis through activation of various downward mediators, such as the vascular endothelial growth factor (VEGF). Here, we aimed to explore the role of HIF-1α/VEGF immunophenotypes alone and in combination with other prognostic markers or clinical and image analysis data, as potential biomarkers of GBM prognosis and treatment efficacy. We performed a systematic review (Medline/Embase, and Pubmed database search was completed by 16th of April 2024 by two independent teams; PRISMA 2020). We evaluated methods of immunoassays, cell viability, or animal or patient survival methods of the retrieved studies to assess unbiased data. We used inclusion criteria, such as the evaluation of GBM prognosis based on HIF-1α/VEGF expression, other biomarkers or clinical and imaging manifestations in GBM related to HIF-1α/VEGF expression, application of immunoassays for protein expression, and evaluation of the effectiveness of GBM therapeutic strategies based on HIF-1α/VEGF expression. We used exclusion criteria, such as data not reporting both HIF-1α and VEGF or prognosis. We included 50 studies investigating in total 1319 GBM human specimens, 18 different cell lines or GBM-derived stem cells, and 6 different animal models, to identify the association of HIF-1α/VEGF immunophenotypes, and with other prognostic factors, clinical and macroscopic data in GBM prognosis and therapeutic approaches. We found that increased HIF-1α/VEGF expression in GBM correlates with oncogenic factors, such as miR-210-3p, Oct4, AKT, COX-2, PDGF-C, PLDO3, M2 polarization, or ALK, leading to unfavorable survival. Reduced HIF-1α/VEGF expression correlates with FIH-1, ADNP, or STAT1 upregulation, as well as with clinical manifestations, like epileptogenicity, and a favorable prognosis of GBM. Based on our data, HIF-1α or VEGF immunophenotypes may be a useful tool to clarify MRI-PET imaging data distinguishing between GBM tumor progression and pseudoprogression. Finally, HIF-1α/VEGF immunophenotypes can reflect GBM treatment efficacy, including combined first-line treatment with histone deacetylase inhibitors, thimerosal, or an active metabolite of irinotecan, as well as STAT3 inhibitors alone, and resulting in a favorable tumor prognosis and patient survival. These data were supported by a combination of variable methods used to evaluate HIF-1α/VEGF immunophenotypes. Data limitations may include the use of less sensitive detection methods in some cases. Overall, our data support HIF-1α/VEGF's role as biomarkers of GBM prognosis and treatment efficacy.
Collapse
Affiliation(s)
- DIMITRA P. VAGELI
- Department of Surgery, Yale University, New Haven, CT 06510, USA
- Department of Pathology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, 41500, Greece
| | - PANAGIOTIS G. DOUKAS
- Department of Medicine, Rutgers/Saint Peter’s University Hospital, New Brunswick, NJ08901, USA
| | - KERASIA GOUPOU
- Department of Pathology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, 41500, Greece
| | - ANTONIOS D. BENOS
- Department of Pathology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, 41500, Greece
| | - KYRIAKI ASTARA
- Department of Pathology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, 41500, Greece
- Department of Neurology, Army Share Fund Hospital (NIMTS), Athens, 11521, Greece
| | - KONSTANTINA ZACHAROULI
- Department of Pathology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, 41500, Greece
| | - SOTIRIS SOTIRIOU
- Laboratory of Embryology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, 41500, Greece
| | - MARIA IOANNOU
- Department of Pathology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, 41500, Greece
| |
Collapse
|
3
|
Rehan F, Zhang M, Fang J, Greish K. Therapeutic Applications of Nanomedicine: Recent Developments and Future Perspectives. Molecules 2024; 29:2073. [PMID: 38731563 PMCID: PMC11085487 DOI: 10.3390/molecules29092073] [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: 04/08/2024] [Revised: 04/24/2024] [Accepted: 04/26/2024] [Indexed: 05/13/2024] Open
Abstract
The concept of nanomedicine has evolved significantly in recent decades, leveraging the unique phenomenon known as the enhanced permeability and retention (EPR) effect. This has facilitated major advancements in targeted drug delivery, imaging, and individualized therapy through the integration of nanotechnology principles into medicine. Numerous nanomedicines have been developed and applied for disease treatment, with a particular focus on cancer therapy. Recently, nanomedicine has been utilized in various advanced fields, including diagnosis, vaccines, immunotherapy, gene delivery, and tissue engineering. Multifunctional nanomedicines facilitate concurrent medication delivery, therapeutic monitoring, and imaging, allowing for immediate responses and personalized treatment plans. This review concerns the major advancement of nanomaterials and their potential applications in the biological and medical fields. Along with this, we also mention the various clinical translations of nanomedicine and the major challenges that nanomedicine is currently facing to overcome the clinical translation barrier.
Collapse
Affiliation(s)
- Farah Rehan
- Department of Molecular Medicine, Al-Jawhara Centre for Molecular Medicine, College of Medicine and Medical Sciences, Arabian Gulf University, Manama 323, Bahrain;
| | - Mingjie Zhang
- Faculty of Pharmaceutical Sciences, Sojo University, Ikeda 4-22-1, Nishi-ku, Kumamoto 860-0082, Japan;
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Jun Fang
- Faculty of Pharmaceutical Sciences, Sojo University, Ikeda 4-22-1, Nishi-ku, Kumamoto 860-0082, Japan;
| | - Khaled Greish
- Department of Molecular Medicine, Al-Jawhara Centre for Molecular Medicine, College of Medicine and Medical Sciences, Arabian Gulf University, Manama 323, Bahrain;
| |
Collapse
|
4
|
Ahmad A, Rashid S, Chaudhary AA, Alawam AS, Alghonaim MI, Raza SS, Khan R. Nanomedicine as potential cancer therapy via targeting dysregulated transcription factors. Semin Cancer Biol 2023; 89:38-60. [PMID: 36669712 DOI: 10.1016/j.semcancer.2023.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 01/02/2023] [Accepted: 01/15/2023] [Indexed: 01/19/2023]
Abstract
Cancer as a disease possess quite complicated pathophysiological implications and is among the prominent causes of morbidity and mortality on global scales. Anti-cancer chemotherapy, surgery, and radiation therapy are some of the present-day conventional treatment options. However, these therapeutic paradigms own several retreats, including lack of specificity, non-targeted toxicological implications, inefficient drug delivery to targeted cells, and emergence of cancer resistance, ultimately causing ineffective cancer management. Owing to the advanced and better biophysical characteristic features and potentiality for the tailoring and customizations and in several fashions, nanotechnology can entirely transubstantiate the cancer identification and its managements. Additionally, nanotechnology also renders several answers to present-day mainstream limitations springing-up in anti-cancer therapeutics. Nanocarriers, owing to their outstanding physicochemical features including but not limited to their particle size, surface morphological features viz. shape etc., have been employed in nanomedicinal platforms for targeting various transcription factors leading to worthy pharmacological outcomes. This transcription targeting activates the wide array of cellular and molecular events like antioxidant enzyme-induction, apoptotic cell death, cell-cycle arrest etc. These outcomes are obtained after the activation or inactivation of several transcription factors and cellular pathways. Further, nanoformulations have been precisely calibrated and functionalized with peculiar targeting groups for improving their efficiency to deliver the drug-payload to specified and targeted cancerous cells and tissues. This review undertakes an extensive, across-the-board and all-inclusive approach consisting of various studies encompassing different types of tailored and customized nanoformulations and nanomaterials designed for targeting the transcription factors implicated in the process of carcinogenesis, tumor-maturation, growth and metastasis. Various transcription factors viz. nuclear factor kappa (NF-κB), signal transducer and activators of transcription (STAT), Cmyc and Twist-related protein 1 (TWIST1) along with several types of nanoparticles targeting these transcription factors have been summarized here. A section has also been dedicated to the different types of nanoparticles targeting the hypoxia inducing factors. Efforts have been made to summarize several other transcription factors implicated in various stages of cancer development, growth, progression and invasion, and their targeting with different kinds of nanomedicinal agents.
Collapse
Affiliation(s)
- Anas Ahmad
- Julia McFarlane Diabetes Research Centre (JMDRC), Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - Summya Rashid
- Department of Pharmacology & Toxicology, College of Pharmacy, Prince Sattam Bin Abdulaziz University, P.O. Box 173, Al-Kharj 11942, Saudi Arabia
| | - Anis Ahmad Chaudhary
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh 11623, Saudi Arabia
| | - Abdullah S Alawam
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh 11623, Saudi Arabia
| | - Mohammad Ibrahim Alghonaim
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh 11623, Saudi Arabia
| | - Syed Shadab Raza
- Laboratory for Stem Cell and Restorative Neurology, Department of Biotechnology, Era's Lucknow Medical College Hospital, Sarfarazganj, Lucknow 226003, India
| | - Rehan Khan
- Chemical Biology Unit, Institute of Nano Science and Technology (INST), Knowledge City, Sector 81, Mohali, Punjab 140306, India.
| |
Collapse
|
5
|
Schmidt KT, Karzai F, Bilusic M, Cordes LM, Chau CH, Peer CJ, Wroblewski S, Huitema ADR, Schellens JHM, Gulley JL, Dahut WL, Figg WD, Madan RA. A Single-arm Phase II Study Combining NLG207, a Nanoparticle Camptothecin, with Enzalutamide in Advanced Metastatic Castration-resistant Prostate Cancer Post-Enzalutamide. Oncologist 2022; 27:718-e694. [PMID: 35640474 PMCID: PMC9438911 DOI: 10.1093/oncolo/oyac100] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 04/06/2022] [Indexed: 07/30/2023] Open
Abstract
BACKGROUND Despite the clinical efficacy of enzalutamide monotherapy in patients with advanced prostate cancer, therapeutic resistance and disease progression are inevitable. We proposed a study to evaluate NLG207, a nanoparticle-drug conjugate (NDC) of the potent topoisomerase I inhibitor camptothecin, in combination with enzalutamide, in patients with metastatic castration-resistant prostate cancer (mCRPC) following progression on enzalutamide. METHODS This was a single-arm, optimal two-stage, phase II study to evaluate the efficacy of NLG207 in combination with enzalutamide in patients with mCRPC who received prior enzalutamide. A lead-in dose escalation evaluated the recommended phase 2 dose of NLG207 in combination with enzalutamide. Patients received NLG207 via IV infusion every 2 weeks and enzalutamide 160 mg orally once daily. RESULTS Between March 2019 and June 2021, four patients were accrued to the lead-in dose escalation. Two of the four patients were evaluable and both experienced DLTs at the NLG207 12 mg/m2 dose level; one DLT was related to a dose delay for noninfective cystitis and myelosuppression, the other a grade 3 noninfective cystitis. Further evaluation of NLG207 in combination with enzalutamide was halted and the study was ultimately terminated. PSA declines from baseline were observed in two patients. CONCLUSION NLG207 12 mg/m2 in combination with enzalutamide was not well tolerated in patients with mCRPC following several lines of the standard of care therapy. CLINICALTRIALS.GOV IDENTIFIER NCT03531827.
Collapse
Affiliation(s)
- Keith T Schmidt
- Clinical Pharmacology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Fatima Karzai
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Marijo Bilusic
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Lisa M Cordes
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Cindy H Chau
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Cody J Peer
- Clinical Pharmacology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Susan Wroblewski
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Alwin D R Huitema
- Department Pharmacy & Pharmacology, Netherlands Cancer Institute, Amsterdam, The Netherlands
- Department of Clinical Pharmacy, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Department of Pharmacology, Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Jan H M Schellens
- Utrecht Institute of Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - James L Gulley
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - William L Dahut
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - William D Figg
- Corresponding author: William D. Figg, 9000 Rockville Pike, Building 10/Room 5A03, Bethesda, MD 20892, USA. Tel: +1 240 760 6179;
| | | |
Collapse
|
6
|
Buchelnikova VA, Rogozhin EA, Barashkova AS, Buchelnikov AS, Evstigneev MP. C 60 Fullerene Clusters Stabilize the Biologically Inactive Form of Topotecan. Chem Res Toxicol 2022; 35:1482-1492. [PMID: 35980010 DOI: 10.1021/acs.chemrestox.2c00071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
There is a range of experimental proofs that biologically relevant compounds change their activity in the presence of C60 fullerene clusters in aqueous solution, which most frequently act as a nanoplatform for drug delivery. Inspired by this evidence, we made an effort to investigate the interaction of fullerene clusters with the antibiotic topotecan (TPT). This study proceeded in three steps, namely, UV/vis titration to confirm complexation and in vitro assays on proliferating and nonproliferating cells to elucidate the role of C60 fullerene in the putative change in TPT activity. Surprisingly, although the nonproliferating cell assay is consistent with the titration data and confirms complex formation, it contradicted the results of the proliferating cell assay. The latter showed that the mixture of TPT and fullerene affects the cells in the same way as pure TPT, as if there were no fullerenes in solution at all, whereas the action of TPT was expected to be enhanced. We explained this contradiction by the specific stabilization of the biologically inactive carboxylate form of the antibiotic adsorbed in the alkaline shell of large fullerene clusters, which leads to neutralization of the drug delivery function and almost zero net biological effect of the antibiotic in vitro. The practical outcome of the work is that fullerene clusters can be used for the selective delivery of pH-sensitive drug forms.
Collapse
Affiliation(s)
| | - Eugene A Rogozhin
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry Russian Academy of Sciences, Moscow 117997, Russia.,Gause Institute of New Antibiotics, Moscow 119021, Russia
| | - Anna S Barashkova
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry Russian Academy of Sciences, Moscow 117997, Russia
| | | | | |
Collapse
|
7
|
Nguyen-Trinh QN, Trinh KXT, Trinh NT, Vo VT, Li N, Nagasaki Y, Vong LB. A silica-based antioxidant nanoparticle for oral delivery of Camptothecin which reduces intestinal side effects while improving drug efficacy for colon cancer treatment. Acta Biomater 2022; 143:459-470. [PMID: 35235866 DOI: 10.1016/j.actbio.2022.02.036] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Revised: 02/17/2022] [Accepted: 02/22/2022] [Indexed: 12/20/2022]
Abstract
Camptothecin (CPT) is a potent anticancer agent for the treatment of colorectal cancer; however, it exhibits some limitations, including poor solubility, low stability, and low bioavailability via oral administration, which restrict its usability in clinical treatments. In addition, overproduction of reactive oxygen species (ROS) during chemotherapy induces drug resistance and severe intestinal side effects. In this study, silica-installed ROS scavenging nanoparticles (siRNP) with 50-60 nm in diameter were employed to overcome the aforementioned drawbacks of CPT. The solubility of CPT was significantly improved by incorporating it into the core of the nanoparticle, forming CPT-loaded siRNP (CPT@siRNP). The anticancer activity of CPT@siRNP against colorectal cancer cells (C-26) in vitro was significantly improved as compared to free CPT through higher efficiency of intracellular internalization and induction of apoptosis. Owing to its antioxidant properties, CPT@siRNP reduced cytotoxicity to normal endothelial cells, which was in sharp contrast to the high toxicity of free CPT. Oral administration of CPT and CPT@siRNP to the C-26 tumor-bearing mice exhibited antitumor activity, accompanied by effective suppression of tumor growth. Although CPT treatment suppressed tumor progression, it caused severe side effects, including intestinal damage and significant bodyweight loss. Interestingly, such noticeable side effects were not observed in the mice treated with CPT@siRNP, and the effect of tumor growth inhibition tended to be similar to or higher than that of CPT treatment. The results obtained in this study indicate that CPT@siRNP is a potential therapeutic nanomedicine for the treatment of colon cancer. STATEMENT OF SIGNIFICANCE: Here we employed silica-containing antioxidant nanoparticle (siRNP) as promising oral delivery nanocarrier of campothecin (CPT) to treat colon cancer. The design of siRNP via covalent conjugation of antioxidant nitroxide radicals and the silanol groups in the polymer backbone contributes to a significant increase in the absorption of hydrophobic drug molecules inside the core and enhances the stability of nanoparticles in the gastrointestinal environment for oral drug delivery. CPT-loaded siRNP (CPT@siRNP) significantly improved solubility of CPT. As compared to free CTP, the CPT@siRNP treatment showed a significantly higher toxicity to colon cancer cell, inhibition of cancer cell migration, and induction of apopotosis. With the antioxidant feature, siRNP also significantly suppressed the intestinal side effects caused by CPT treatment in tumor-bearing mouse model.
Collapse
|
8
|
Zhang Y, Coleman M, Brekken RA. Perspectives on Hypoxia Signaling in Tumor Stroma. Cancers (Basel) 2021; 13:3070. [PMID: 34202979 PMCID: PMC8234221 DOI: 10.3390/cancers13123070] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 06/11/2021] [Accepted: 06/17/2021] [Indexed: 12/12/2022] Open
Abstract
Hypoxia is a well-known characteristic of solid tumors that contributes to tumor progression and metastasis. Oxygen deprivation due to high demand of proliferating cancer cells and standard of care therapies induce hypoxia. Hypoxia signaling, mainly mediated by the hypoxia-inducible transcription factor (HIF) family, results in tumor cell migration, proliferation, metabolic changes, and resistance to therapy. Additionally, the hypoxic tumor microenvironment impacts multiple cellular and non-cellular compartments in the tumor stroma, including disordered tumor vasculature, homeostasis of ECM. Hypoxia also has a multifaceted and often contradictory influence on immune cell function, which contributes to an immunosuppressive environment. Here, we review the important function of HIF in tumor stromal components and summarize current clinical trials targeting hypoxia. We provide an overview of hypoxia signaling in tumor stroma that might help address some of the challenges associated with hypoxia-targeted therapies.
Collapse
Affiliation(s)
- Yuqing Zhang
- Hamon Center for Therapeutic Oncology Research, UT Southwestern, Dallas, TX 75390, USA; (Y.Z.); (M.C.)
- Department of Surgery, UT Southwestern, Dallas, TX 75390, USA
- Cancer Biology Graduate Program, UT Southwestern, Dallas, TX 75390, USA
| | - Morgan Coleman
- Hamon Center for Therapeutic Oncology Research, UT Southwestern, Dallas, TX 75390, USA; (Y.Z.); (M.C.)
- Division of Pediatric Hematology and Oncology, UT Southwestern, Dallas, TX 75390, USA
| | - Rolf A. Brekken
- Hamon Center for Therapeutic Oncology Research, UT Southwestern, Dallas, TX 75390, USA; (Y.Z.); (M.C.)
- Department of Surgery, UT Southwestern, Dallas, TX 75390, USA
- Cancer Biology Graduate Program, UT Southwestern, Dallas, TX 75390, USA
| |
Collapse
|
9
|
Schmidt KT, Chau CH, Strope JD, Huitema ADR, Sissung TM, Price DK, Figg WD. Antitumor Activity of NLG207 (Formerly CRLX101) in Combination with Enzalutamide in Preclinical Prostate Cancer Models. Mol Cancer Ther 2021; 20:915-924. [PMID: 33632874 PMCID: PMC8102325 DOI: 10.1158/1535-7163.mct-20-0228] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 06/06/2020] [Accepted: 02/11/2021] [Indexed: 11/16/2022]
Abstract
Effective treatments for patients with metastatic castration-resistant prostate cancer following disease progression on enzalutamide are currently an unmet clinical need. Simultaneous inhibition of the hypoxia-inducible factor (HIF)-1α and androgen receptor (AR) pathways has been previously shown to overcome enzalutamide resistance in vitro Combination treatment with NLG207, a nanoparticle-drug conjugate of camptothecin and inhibitor of HIF-1α, and enzalutamide was evaluated in preclinical prostate cancer models of enzalutamide resistance. The effect of NLG207 and enzalutamide on average tumor volume and tumor re-growth after 3 weeks of treatment was evaluated in vivo using the subcutaneous 22Rv1 xenograft and castrated subcutaneous VCaP xenograft models. Correlative assessments of antitumor activity were evaluated in vitro using cell proliferation and qPCR assays. NLG207 8 mg/kg alone and in combination with enzalutamide reduced average tumor volume by 93% after 3 weeks of treatment (P < 0.05) in comparison with vehicle control in the subcutaneous 22Rv1 xenograft model. Notably, the addition of NLG207 also enhanced the efficacy of enzalutamide alone in the castrated subcutaneous VCaP xenograft model, decreasing the median rate of tumor growth by 51% (P = 0.0001) in comparison with enzalutamide alone. In vitro assessments of cell proliferation and gene expression further demonstrated antitumor activity via AR-HIF-1α crosstalk inhibition. Combination treatment with NLG207 and enzalutamide was shown to be effective in preclinical prostate cancer models of enzalutamide resistance. Clinical investigation of this treatment combination is ongoing (NCT03531827).
Collapse
Affiliation(s)
- Keith T Schmidt
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Cindy H Chau
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Jonathan D Strope
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Alwin D R Huitema
- Department Pharmacy and Pharmacology, Netherlands Cancer Institute, Amsterdam, The Netherlands
- Department of Clinical Pharmacy, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Tristan M Sissung
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Douglas K Price
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - William D Figg
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland.
| |
Collapse
|
10
|
A phase Ib/II and pharmacokinetic study of EP0057 (formerly CRLX101) in combination with weekly paclitaxel in patients with recurrent or persistent epithelial ovarian, fallopian tube, or primary peritoneal cancer. Gynecol Oncol 2020; 160:688-695. [PMID: 33390325 DOI: 10.1016/j.ygyno.2020.12.025] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 12/19/2020] [Indexed: 11/20/2022]
Abstract
BACKGROUND EP0057 (formerly CRLX101) is an investigational nanoparticle-drug conjugate (NDC) of a cyclodextrin-based polymer backbone plus camptothecin, a topoisomerase-1 inhibitor. Prior studies showed efficacy in recurrent or persistent, epithelial ovarian, fallopian tube or primary peritoneal cancer (EOC). METHODS This phase Ib/2 trial assessed safety and efficacy of EP0057 Q2W plus weekly paclitaxel in patients with EOC. The recommended phase 2 dose (RP2D) was identified using a 3+3 design. The single-arm phase 2 assessed overall response (ORR) per RECIST 1.1 in patients previously treated with bevacizumab. Secondary objectives included progression free survival (PFS) and duration of response. RESULTS The RP2D was established as 15 mg/m2 EP0057 Q2W plus 80 mg/m2 paclitaxel administered 3 weeks on/1 week off. Nine patients enrolled on phase 1b, with no DLTs; 21 additional patients enrolled on phase 2. All completed >1 cycle. Median age was 62 (44-76) years, 57% ≥3 prior therapies. For the primary analysis, 6/19 patients with prior bevacizumab had confirmed responses (ORR=31.6% (95% CI: 15.4% to 54.0%)) including one complete response (CR). Median PFS was 5.4 months. Most common grade 3/4 adverse events attributed to treatment were decreased neutrophil count (13, 43%) and anemia (3, 10%). CONCLUSIONS Although the observed ORR was not statistically better than the historical control rate, EP0057 remains an interesting option for treatment of recurrent EOC. EP0057 exhibits high plasma drug retention, slow clearance, and controlled slow release of CPT from the polymer when administered alone and with paclitaxel. (NCT02389985) 242 words.
Collapse
|
11
|
Menon MP, Hua KF. The Long Non-coding RNAs: Paramount Regulators of the NLRP3 Inflammasome. Front Immunol 2020; 11:569524. [PMID: 33101288 PMCID: PMC7546312 DOI: 10.3389/fimmu.2020.569524] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 08/24/2020] [Indexed: 12/17/2022] Open
Abstract
The NOD LRR pyrin domain containing protein 3 (NLRP3) inflammasome is a cytosolic multi-proteins conglomerate with intrinsic ATPase activity. Their predominant presence in the immune cells emphasizes its significant role in immune response. The downstream effector proteins IL-1β and IL-18 are responsible for the biological functions of the NLRP3 inflammasome upon encountering the alarmins and microbial ligands. Although the NLRP3 inflammasome is essential for host defense during infections, uncontrolled activation and overproduction of IL-1β and IL-18 increase the risk of developing autoimmune and metabolic disorders. Emerging evidences suggest the action of lncRNAs in regulating the activity of NLRP3 inflammasome in various disease conditions. The long non-coding RNA (lncRNA) is an emerging field of study and evidence on their regulatory role in various diseases is grabbing attention. Recent studies emphasize the functions of lncRNAs in the fine control of the NLRP3 inflammasome at nuclear and cytoplasmic levels by interfering in chromatin architecture, gene transcription and translation. Recently, lncRNAs are also found to control the activity of various regulators of NLRP3 inflammasome. Understanding the precise role of lncRNA in controlling the activity of NLRP3 inflammasome helps us to design targeted therapies for multiple inflammatory diseases. The present review is a novel attempt to consolidate the substantial role of lncRNAs in the regulation of the NLRP3 inflammasome. A deeper insight on the NLRP3 inflammasome regulation by lncRNAs will help in developing targeted and beneficial therapeutics in the future.
Collapse
Affiliation(s)
- Mridula P. Menon
- Department of Biotechnology and Animal Science, National Ilan University, Yilan, Taiwan
| | - Kuo-Feng Hua
- Department of Biotechnology and Animal Science, National Ilan University, Yilan, Taiwan
- Department of Pathology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan
| |
Collapse
|
12
|
Particulate systems for improving therapeutic efficacy of pharmaceuticals against central nervous system-related diseases. J Taiwan Inst Chem Eng 2020. [DOI: 10.1016/j.jtice.2020.09.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
13
|
Kargozar S, Baino F, Hamzehlou S, Hamblin MR, Mozafari M. Nanotechnology for angiogenesis: opportunities and challenges. Chem Soc Rev 2020; 49:5008-5057. [PMID: 32538379 PMCID: PMC7418030 DOI: 10.1039/c8cs01021h] [Citation(s) in RCA: 122] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Angiogenesis plays a critical role within the human body, from the early stages of life (i.e., embryonic development) to life-threatening diseases (e.g., cancer, heart attack, stroke, wound healing). Many pharmaceutical companies have expended huge efforts on both stimulation and inhibition of angiogenesis. During the last decade, the nanotechnology revolution has made a great impact in medicine, and regulatory approvals are starting to be achieved for nanomedicines to treat a wide range of diseases. Angiogenesis therapies involve the inhibition of angiogenesis in oncology and ophthalmology, and stimulation of angiogenesis in wound healing and tissue engineering. This review aims to summarize nanotechnology-based strategies that have been explored in the broad area of angiogenesis. Lipid-based, carbon-based and polymeric nanoparticles, and a wide range of inorganic and metallic nanoparticles are covered in detail. Theranostic and imaging approaches can be facilitated by nanoparticles. Many preparations have been reported to have a bimodal effect where they stimulate angiogenesis at low dose and inhibit it at higher doses.
Collapse
Affiliation(s)
- Saeid Kargozar
- Tissue Engineering Research Group (TERG), Department of Anatomy and Cell Biology, School of Medicine, Mashhad University of Medical Sciences, 917794-8564 Mashhad, Iran
| | - Francesco Baino
- Institute of Materials Physics and Engineering, Applied Science and Technology Department, Politecnico di Torino, Corso Duca degli Abruzzi 24, 101 29 Torino, Italy
| | - Sepideh Hamzehlou
- Hematology/Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Michael R. Hamblin
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Dermatology, Harvard Medical School, Boston, MA 02115, USA
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein 2028, South Africa
| | - Masoud Mozafari
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
14
|
Nery de Albuquerque Rego G, da Hora Alves A, Penteado Nucci M, Bustamante Mamani J, Anselmo de Oliveira F, Gamarra LF. Antiangiogenic Targets for Glioblastoma Therapy from a Pre-Clinical Approach, Using Nanoformulations. Int J Mol Sci 2020; 21:ijms21124490. [PMID: 32599834 PMCID: PMC7349965 DOI: 10.3390/ijms21124490] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Revised: 06/12/2020] [Accepted: 06/18/2020] [Indexed: 12/13/2022] Open
Abstract
Glioblastoma (GBM) is the most aggressive tumor type whose resistance to conventional treatment is mediated, in part, by the angiogenic process. New treatments involving the application of nanoformulations composed of encapsulated drugs coupled to peptide motifs that direct drugs to specific targets triggered in angiogenesis have been developed to reach and modulate different phases of this process. We performed a systematic review with the search criterion (Glioblastoma OR Glioma) AND (Therapy OR Therapeutic) AND (Nanoparticle) AND (Antiangiogenic OR Angiogenesis OR Anti-angiogenic) in Pubmed, Scopus, and Cochrane databases, in which 312 articles were identified; of these, only 27 articles were included after selection and analysis of eligibility according to the inclusion and exclusion criteria. The data of the articles were analyzed in five contexts: the characteristics of the tumor cells; the animal models used to induce GBM for antiangiogenic treatment; the composition of nanoformulations and their physical and chemical characteristics; the therapeutic anti-angiogenic process; and methods for assessing the effects on antiangiogenic markers caused by therapies. The articles included in the review were heterogeneous and varied in practically all aspects related to nanoformulations and models. However, there was slight variance in the antiangiogenic effect analysis. CD31 was extensively used as a marker, which does not provide a view of the effects on the most diverse aspects involved in angiogenesis. Therefore, the present review highlighted the need for standardization between the different approaches of antiangiogenic therapy for the GBM model that allows a more effective meta-analysis and that helps in future translational studies.
Collapse
Affiliation(s)
| | - Arielly da Hora Alves
- Hospital Israelita Albert Einstein, São Paulo 05652-900, Brazil; (G.N.d.A.R.); (A.d.H.A.); (J.B.M.); (F.A.d.O.)
| | - Mariana Penteado Nucci
- LIM44-Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo 01246-903, Brazil;
| | - Javier Bustamante Mamani
- Hospital Israelita Albert Einstein, São Paulo 05652-900, Brazil; (G.N.d.A.R.); (A.d.H.A.); (J.B.M.); (F.A.d.O.)
| | | | - Lionel Fernel Gamarra
- Hospital Israelita Albert Einstein, São Paulo 05652-900, Brazil; (G.N.d.A.R.); (A.d.H.A.); (J.B.M.); (F.A.d.O.)
- Correspondence: ; Tel.: +55-11-2151-0243
| |
Collapse
|
15
|
Allen J, Wang J, Zolotarskaya OY, Sule A, Mohammad S, Arslan S, Wynne KJ, Yang H, Valerie K. PEAMOtecan, a novel chronotherapeutic polymeric drug for brain cancer. J Control Release 2020; 321:36-48. [PMID: 32027939 DOI: 10.1016/j.jconrel.2020.02.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 01/26/2020] [Accepted: 02/02/2020] [Indexed: 12/12/2022]
Abstract
Glioblastoma multiforme (GBM) is an aggressive and difficult to treat form of brain cancer. In this work, we report on a novel chronotherapeutic polymeric drug, PEAMOtecan, for GBM therapy. PEAMOtecan was synthesized by conjugating camptothecin, a topoisomerase I inhibitor, to our proprietary, 'clickable' and modular polyoxetane polymer platform consisting of acetylene-functionalized 3-ethyl-3-(hydroxymethyl)oxetane (EAMO) repeat units (Patent No.: US 9,421,276) via the linker 3,3'-dithiodipropionic acid (DDPA) with a disulfide bond (SS) extended by short-chain polyethylene glycol (PEG). We show that PEAMOtecan is a highly modular polymer nanoformulation that protects covalently bound CPT until slowly being released over extended periods of time dependent on the cleavage of the disulfide and ester linkages. PEAMOtecan kills glioma cells by mitotic catastrophe with p53 mutant/knockdown cells being more sensitive than matched wild type cells potentially providing cancer-specific targeting. To establish proof-of-principle therapeutic effects, we tested PEAMOtecan as monotherapy for efficacy in a mouse orthotopic glioma model. PEAMOtecan was administered by one-time, convection-enhanced delivery (CED) intra-tumorally to achieve superior distribution and extended drug release over time. In addition, the near-infrared (NIR) dye Cy5.5 was coupled to the polymer providing live-animal imaging capability to track tissue distribution and clearance of the injected polymer over time. We show that PEAMOtecan significantly improves the survival of mice harboring intra-cranial tumors (p = .0074 compared to untreated group). Altogether, these results support further development and testing of our nanoconjugate platform.
Collapse
Affiliation(s)
- Jasmine Allen
- Department of Radiation Oncology, Virginia Commonwealth University, Richmond, Virginia 23298, United States of America
| | - Juan Wang
- Department of Radiation Oncology, Virginia Commonwealth University, Richmond, Virginia 23298, United States of America
| | - Olga Yu Zolotarskaya
- Department of Chemical and Life Science Engineering, Virginia Commonwealth University, Richmond, Virginia 23298, United States of America
| | - Amrita Sule
- Department of Radiation Oncology, Virginia Commonwealth University, Richmond, Virginia 23298, United States of America
| | - Sajjad Mohammad
- Department of Radiation Oncology, Virginia Commonwealth University, Richmond, Virginia 23298, United States of America
| | - Shukaib Arslan
- Department of Radiation Oncology, Virginia Commonwealth University, Richmond, Virginia 23298, United States of America
| | - Kenneth J Wynne
- Department of Chemical and Life Science Engineering, Virginia Commonwealth University, Richmond, Virginia 23298, United States of America
| | - Hu Yang
- Department of Chemical and Life Science Engineering, Virginia Commonwealth University, Richmond, Virginia 23298, United States of America; Department of Pharmaceutics, Virginia Commonwealth University, Richmond, Virginia 23298, United States of America; Massey Cancer Center, Virginia Commonwealth University, Richmond, Virginia 23298, United States of America.
| | - Kristoffer Valerie
- Department of Radiation Oncology, Virginia Commonwealth University, Richmond, Virginia 23298, United States of America; Massey Cancer Center, Virginia Commonwealth University, Richmond, Virginia 23298, United States of America.
| |
Collapse
|
16
|
Hsu FT, Chiang I, Wang W. Induction of apoptosis through extrinsic/intrinsic pathways and suppression of ERK/NF-κB signalling participate in anti-glioblastoma of imipramine. J Cell Mol Med 2020; 24:3982-4000. [PMID: 32149465 PMCID: PMC7171418 DOI: 10.1111/jcmm.15022] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Revised: 11/27/2019] [Accepted: 01/06/2020] [Indexed: 12/11/2022] Open
Abstract
Glioblastomas are the most aggressive type of brain tumour, with poor prognosis even after standard treatment such as surgical resection, temozolomide and radiation therapy. The overexpression of the nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) in glioblastomas is recognized as an important treatment target. Thus, an urgent need regarding glioblastomas is the development of a new, suitable agent that may show potential for the inhibition of extracellular signal-regulated kinase (ERK)/NF-κB-mediated glioblastoma progression. Imipramine, a tricyclic antidepressant, has anti-inflammatory actions against inflamed glial cells; additionally, imipramine can induce glioblastoma toxicity via the activation of autophagy. However, whether imipramine can suppress glioblastoma progression via the induction of apoptosis and blockage of ERK/NF-κB signalling remains unclear. The main purpose of this study was to investigate the effects of imipramine on apoptotic signalling and ERK/NF-κB-mediated glioblastoma progression by using cell proliferation (3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide [MTT] assay), flow cytometry, Western blotting, and cell invasion/migration assay analysis in vitro. The ERK and NF-κB inhibitory capacity of imipramine is detected by NF-κB reporter gene assay and Western blotting. Additionally, a glioblastoma-bearing animal model was used to validate the therapeutic efficacy and general toxicity of imipramine. Our results demonstrated that imipramine successfully triggered apoptosis through extrinsic/intrinsic pathways and suppressed the invasion/migration ability of glioblastoma cells. Furthermore, imipramine effectively suppressed glioblastoma progression in vivo via the inhibition of the ERK/NF-κB pathway. In summary, imipramine is a potential anti-glioblastoma drug which induces apoptosis and has the capacity to inhibit ERK/NF-κB signalling.
Collapse
Affiliation(s)
- Fei-Ting Hsu
- Department of Biological Science and TechnologyChina Medical UniversityTaichungTaiwan
| | - I‐Tsang Chiang
- Department of Radiation OncologyShow Chwan Memorial HospitalChanghuaTaiwan
- Department of Radiation OncologyChang Bing Show Chwan Memorial HospitalLukangTaiwan
- Department of Medical Imaging and Radiological SciencesCentral Taiwan University of Science and TechnologyTaichungTaiwan
| | - Wei‐Shu Wang
- Department of MedicineNational Yang‐Ming University HospitalYilanTaiwan
- School of MedicineNational Yang‐Ming UniversityTaipeiTaiwan
| |
Collapse
|
17
|
Major Contribution of Caspase-9 to Honokiol-Induced Apoptotic Insults to Human Drug-Resistant Glioblastoma Cells. Molecules 2020; 25:molecules25061450. [PMID: 32210117 PMCID: PMC7145301 DOI: 10.3390/molecules25061450] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2019] [Revised: 12/12/2019] [Accepted: 03/20/2020] [Indexed: 12/12/2022] Open
Abstract
Temozolomide (TMZ)-induced chemoresistance to human glioblastomas is a critical challenge now. Our previous studies showed that honokiol, a major bioactive constituent of Magnolia officinalis (Houpo), can kill human glioblastoma cells and suppresses glioblastoma growth. This study was further aimed to evaluate the effects of honokiol on human drug-resistant glioblastoma cells and the possible mechanisms. The results by data mining in the cancer genome atlas (TCGA) database and immunohistochemistry displayed that expression of caspase-9 mRNA and protein in human glioblastomas was induced. Human TMZ-resistant U87-MG-R9 glioblastoma cells were selected and prepared from human drug-sensitive U87-MG cells. Compared to human drug-sensitive U87-MG cells, TMZ did not affect viability of U87-MG-R9 glioblastoma cells. Interestingly, treatment with honokiol suppressed proliferation and survival of human drug-resistant glioblastoma cells in concentration- and time-dependent manners. Compared to caspase-8 activation, honokiol chiefly increased activity of caspase-9 in U87-MG-R9 cells. Successively, levels of cleaved caspase-3 and activities of caspase-3 and caspase-6 in human TMZ-tolerant glioblastoma cells were augmented following honokiol administration. In parallel, honokiol triggered DNA fragmentation of U87-MG-R9 cells. Accordingly, treatment of human TMZ-resistant glioblastoma cells with honokiol induced cell apoptosis but did not affect cell necrosis. Fascinatingly, suppressing caspase-9 activity using its specific inhibitors repressed honokiol-induced caspase-6 activation, DNA fragmentation, and cell apoptosis. Taken together, this study has shown the major roles of caspase-9 in transducing honokiol-induced mitochondria-dependent apoptosis in human drug-resistant glioblastoma cells. Thus, honokiol may be clinically applied as a drug candidate for treatment of glioblastoma patients with chemoresistance.
Collapse
|
18
|
Ramadan WS, Zaher DM, Altaie AM, Talaat IM, Elmoselhi A. Potential Therapeutic Strategies for Lung and Breast Cancers through Understanding the Anti-Angiogenesis Resistance Mechanisms. Int J Mol Sci 2020; 21:ijms21020565. [PMID: 31952335 PMCID: PMC7014257 DOI: 10.3390/ijms21020565] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 12/16/2019] [Accepted: 01/03/2020] [Indexed: 02/07/2023] Open
Abstract
Breast and lung cancers are among the top cancer types in terms of incidence and mortality burden worldwide. One of the challenges in the treatment of breast and lung cancers is their resistance to administered drugs, as observed with angiogenesis inhibitors. Based on clinical and pre-clinical findings, these two types of cancers have gained the ability to resist angiogenesis inhibitors through several mechanisms that rely on cellular and extracellular factors. This resistance is mediated through angiogenesis-independent vascularization, and it is related to cancer cells and their microenvironment. The mechanisms that cancer cells utilize include metabolic symbiosis and invasion, and they also take advantage of neighboring cells like macrophages, endothelial cells, myeloid and adipose cells. Overcoming resistance is of great interest, and researchers are investigating possible strategies to enhance sensitivity towards angiogenesis inhibitors. These strategies involved targeting multiple players in angiogenesis, epigenetics, hypoxia, cellular metabolism and the immune system. This review aims to discuss the mechanisms of resistance to angiogenesis inhibitors and to highlight recently developed approaches to overcome this resistance.
Collapse
Affiliation(s)
- Wafaa S. Ramadan
- College of Medicine, University of Sharjah, Sharjah 27272, UAE; (W.S.R.); (D.M.Z.); (A.M.A.); (A.E.)
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah 27272, UAE
| | - Dana M. Zaher
- College of Medicine, University of Sharjah, Sharjah 27272, UAE; (W.S.R.); (D.M.Z.); (A.M.A.); (A.E.)
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah 27272, UAE
| | - Alaa M. Altaie
- College of Medicine, University of Sharjah, Sharjah 27272, UAE; (W.S.R.); (D.M.Z.); (A.M.A.); (A.E.)
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah 27272, UAE
| | - Iman M. Talaat
- College of Medicine, University of Sharjah, Sharjah 27272, UAE; (W.S.R.); (D.M.Z.); (A.M.A.); (A.E.)
- Pathology Department, Faculty of Medicine, Alexandria University, 21526 Alexandria, Egypt
- Correspondence: ; Tel.: +971-65057221
| | - Adel Elmoselhi
- College of Medicine, University of Sharjah, Sharjah 27272, UAE; (W.S.R.); (D.M.Z.); (A.M.A.); (A.E.)
- Department of Physiology, Michigan State University, East Lansing, MI 48824, USA
| |
Collapse
|
19
|
Measurement of NLG207 (formerly CRLX101) nanoparticle-bound and released camptothecin in human plasma. J Pharm Biomed Anal 2019; 181:113073. [PMID: 31927166 DOI: 10.1016/j.jpba.2019.113073] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Revised: 11/27/2019] [Accepted: 12/21/2019] [Indexed: 12/16/2022]
Abstract
Camptothecin (CPT), a potent inhibitor of topoisomerase I and HIF-1α, failed to demonstrate utility as an anti-cancer agent in early clinical trial investigations, primarily due to limited clinical activity and significant toxicity attributable to unfavorable physicochemical properties (e.g. low plasma solubility, pH-labile lactone ring). NLG207 (formerly CRLX101), a nanoparticle-drug conjugate (NDC) of CPT designed to optimize plasma pharmacokinetics and facilitate drug delivery to tumors, is included as part of combination treatment in two Phase II clinical trials ongoing at the National Cancer Institute (NCT02769962 and NCT03531827). To better understand the potential for drug-drug interactions and to correlate drug exposure to clinical outcomes and pharmacodynamic biomarkers, a robust analytical method was developed to measure CPT in human plasma. Two sample processing methods were developed to quantify both NDC-bound CPT and free CPT, primarily via alteration of pH conditions. A solid-phase extraction recovered >79 % of CPT prior to quantitative analysis by ultra HPLC-MS/MS. Dynamic calibration ranges of 10 to 10,000 ng/mL and 1 to 1000 ng/mL for total and free CPT, respectively were utilized to capture clinical ranges. NLG207 NDCs demonstrated significant rates of CPT release in human plasma at room temperature after 2 h but were shown to be stable at 4 °C for 24 h and through 4 freeze/thaw cycles. This assay was used to quantitate CPT plasma concentrations in clinical samples to confirm clinical utility following NLG207 treatment in subjects with advanced prostate cancer.
Collapse
|
20
|
Fakhri S, Abbaszadeh F, Jorjani M, Pourgholami MH. The effects of anticancer medicinal herbs on vascular endothelial growth factor based on pharmacological aspects: a review study. Nutr Cancer 2019; 73:1-15. [DOI: 10.1080/01635581.2019.1673451] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
- Sajad Fakhri
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Fatemeh Abbaszadeh
- Department of Neuroscience, Faculty of Advanced Technologies in Medical Sciences, Iran University of Medical Sciences, Tehran, Iran
- Neurobiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Masoumeh Jorjani
- Neurobiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | |
Collapse
|
21
|
Chen YF, Wang YH, Lei CS, Changou CA, Davis ME, Yen Y. Host immune response to anti-cancer camptothecin conjugated cyclodextrin-based polymers. J Biomed Sci 2019; 26:85. [PMID: 31647037 PMCID: PMC6806548 DOI: 10.1186/s12929-019-0583-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 10/10/2019] [Indexed: 12/11/2022] Open
Abstract
Introduction Efficacy and safety are critical concerns when designing drug carriers. Nanoparticles are a particular type of carrier that has gained recent attention in cancer therapeutics. Methods In this study, we assess the safety profile of IT-101, a nanoparticle formed by self-assembly of camptothecin (CPT) conjugated cyclodextrin-based polymers. IT-101 delivers CPT to target cancer cells in animal models of numerous human cancers and in humans. Previous data from preclinical and clinical trials indicate that IT-101 has no notable immunological side effects. However, there have been no published studies focused on evaluating the effects of IT-101 on host immune systems. Results In this work, we demonstrate that IT-101 diminished initial host immune response following first injection of the nanopharmaceutical and induced NK cell activation and T cell proliferation upon further IT-101 exposure. Additionally, IT-101 could attenuate tumor growth more efficiently than CPT treatment only. Conclusions Drugs administration in whole-body circulation may lead to poorly bioavailable in central nervous system and often has toxic effects on peripheral tissues. Conjugated with cyclodextrin-based polymers not only reduce adverse effects but also modulate the immune responses to elevate drug efficacy. These immune responses may potentially facilitate actions of immune blockage, such as PD1/PDL1 in cancer treatment.
Collapse
Affiliation(s)
- Yi-Fan Chen
- The Ph.D. Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, 11031, Taipei, Taiwan.,Master Program for Clinical Pharmacogenomics and Pharmacoproteomics, Taipei Medical University, 11031, Taipei, Taiwan
| | - Yen-Hsin Wang
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, 11031, Taipei, Taiwan
| | - Cing-Syuan Lei
- The Ph.D. Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, 11031, Taipei, Taiwan
| | - Chun A Changou
- Ph.D. Program of Cancer Biology and Drug Discovery, Taipei Medical University, 11031, Taipei, Taiwan.,Integrated Laboratory, Center of Translational Medicine, Taipei Medical University, 11031, Taipei, Taiwan.,Core Facility, Taipei Medical University, 11031, Taipei, Taiwan
| | - Mark E Davis
- Chemical Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Yun Yen
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, 11031, Taipei, Taiwan. .,Integrated Laboratory, Center of Translational Medicine, Taipei Medical University, 11031, Taipei, Taiwan. .,Graduate Institute of Cancer Biology and Drug Discovery, Taipei Medical University, 11031, Taipei, Taiwan. .,Cancer Center, Taipei Municipal WanFang Hospital, 11696, Taipei, Taiwan.
| |
Collapse
|
22
|
Saghafi T, Taheri RA, Parkkila S, Emameh RZ. Phytochemicals as Modulators of Long Non-Coding RNAs and Inhibitors of Cancer-Related Carbonic Anhydrases. Int J Mol Sci 2019; 20:E2939. [PMID: 31208095 PMCID: PMC6627131 DOI: 10.3390/ijms20122939] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 05/29/2019] [Accepted: 05/30/2019] [Indexed: 01/17/2023] Open
Abstract
Long non-coding RNAs (lncRNAs) are classified as a group of transcripts which regulate various biological processes, such as RNA processing, epigenetic control, and signaling pathways. According to recent studies, lncRNAs are dysregulated in cancer and play an important role in cancer incidence and spreading. There is also an association between lncRNAs and the overexpression of some tumor-associated proteins, including carbonic anhydrases II, IX, and XII (CA II, CA IX, and CA XII). Therefore, not only CA inhibition, but also lncRNA modulation, could represent an attractive strategy for cancer prevention and therapy. Experimental studies have suggested that herbal compounds regulate the expression of many lncRNAs involved in cancer, such as HOTAIR (HOX transcript antisense RNA), H19, MALAT1 (metastasis-associated lung adenocarcinoma transcript 1), PCGEM1 (Prostate cancer gene expression marker 1), PVT1, etc. These plant-derived drugs or phytochemicals include resveratrol, curcumin, genistein, quercetin, epigallocatechin-3-galate, camptothcin, and 3,3'-diindolylmethane. More comprehensive information about lncRNA modulation via phytochemicals would be helpful for the administration of new herbal derivatives in cancer therapy. In this review, we describe the state-of-the-art and potential of phytochemicals as modulators of lncRNAs in different types of cancers.
Collapse
Affiliation(s)
- Tayebeh Saghafi
- Department of Energy and Environmental Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), 14965/161, Tehran, Iran.
| | - Ramezan Ali Taheri
- Nanobiotechnology Research Center, Baqiyatallah University of Medical Sciences, P.O.Box 14965/161 Tehran, Iran.
| | - Seppo Parkkila
- Faculty of Medicine and Health Technology, Tampere University, FI-33520 Tampere, Finland.
- Fimlab Laboratories Ltd. and Tampere University Hospital, FI-33520 Tampere, Finland.
| | - Reza Zolfaghari Emameh
- Department of Energy and Environmental Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), 14965/161, Tehran, Iran.
| |
Collapse
|
23
|
Lin MC, Lee YW, Tseng YY, Lin YW, Chen JT, Liu SH, Chen RM. Honokiol Induces Autophagic Apoptosis in Neuroblastoma Cells through a P53-Dependent Pathway. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2019; 47:895-912. [DOI: 10.1142/s0192415x19500472] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
In children, neuroblastomas are the most common and deadly solid tumor. Our previous studies showed that honokiol can cross the blood–brain barrier and kill neuroblastoma cells. In this study, we further evaluated if exposure to honokiol for short periods could induce autophagy and subsequent apoptosis of neuroblastoma cells and possible mechanisms. Exposure of neuroblastoma neuro-2a cells to honokiol for 24[Formula: see text]h induced morphological shrinkage and cell death. As to the mechanisms, honokiol consecutively induced cytochrome c release from mitochondria, caspase-3 activation, DNA fragmentation and cell apoptosis. Separately, honokiol time-dependently augmented the proportion of autophagic cells and the ratio of light chain 3 (LC3)-II/LC3-I. Pretreatment of neuro-2a cells with 3-methyladenine, an inhibitor of autophagy, attenuated honokiol-induced cell autophagy, caspase-3 activation, DNA damage and cell apoptosis. In contrast, stimulation of autophagy by rapamycin, an inducer of autophagy, significantly enhanced honokiol-induced cell apoptosis. Furthermore, honokiol-induced autophagic apoptosis was confirmed in neuroblastoma NB41A3 cells. Knocking down translation of p53 using RNA interference attenuated honokiol-induced autophagy and apoptosis in neuro-2a and NB41A3 cells. Taken together, this study showed that at early periods, honokiol can induce autophagic apoptosis of neuroblastoma cells through activating a p53-dependent mechanism. Consequently, honokiol has the potential to be a therapeutic option for neuroblastomas.
Collapse
Affiliation(s)
- Ming-Chung Lin
- Department of Anesthesiology, Chi Mei Medical Center, Tainan, Taiwan
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yuan-Wen Lee
- Department of Anesthesiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Anesthesiology and Health Policy Research Center, Taipei Medical University Hospital, Taipei, Taiwan
| | - Yuan-Yun Tseng
- Department of Neurosurgery, Shuang-Ho Hospital, Taipei Medical University, Taipei, Taiwan
| | - Yung-Wei Lin
- Department of Urology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Cell Biology and Molecular Image Research Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Jui-Tai Chen
- Department of Anesthesiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Cell Biology and Molecular Image Research Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Shing-Hwa Liu
- Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Ruei-Ming Chen
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Cell Biology and Molecular Image Research Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
- Anesthesiology and Health Policy Research Center, Taipei Medical University Hospital, Taipei, Taiwan
- TMU Research Center of Cancer Translational Medicine, Taipei, Taiwan
| |
Collapse
|
24
|
Abstract
The systemic delivery of drugs to the body via circulation after oral administration is a preferred method of drug administration during cancer treatment given its ease of implementation. However, the physicochemical properties of many current anticancer drugs limit their effectiveness when delivered by systemic routes. The use of nanoparticles (NPs) has emerged as an effective means of overcoming the inherent limitations of systemic drug delivery. We provide herein an overview of various NP formulations that facilitate improvements in the efficacy of various anticancer drugs compared with the free drug. This review will be useful to the reader who is interested in the role NP technology is playing in shaping the future of chemotherapeutic drug delivery and disease treatment.
Collapse
|
25
|
Mebendazole and radiation in combination increase survival through anticancer mechanisms in an intracranial rodent model of malignant meningioma. J Neurooncol 2018; 140:529-538. [PMID: 30414098 DOI: 10.1007/s11060-018-03009-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Accepted: 09/20/2018] [Indexed: 12/16/2022]
Abstract
PURPOSE Meningiomas are a frequent tumor of the central nervous system. Although mostly benign, approximately 5% present as atypical or malignant tumors. Treatments for atypical meningiomas include gross total resection and radiotherapy, but about 33% of patients have recurrent tumors, sometimes as a higher grade. Recently, the brain penetrant anthelmintic drug, mebendazole, has shown promise as an anticancer agent in rodent models of glioblastoma and medulloblastoma. METHODS The half maximal inhibitory concentration (IC50) effect on colony formation, cell proliferation, and caspase-3/7 markers of apoptosis of mebendazole with and without radiation was measured in vitro. Mice intracranially implanted with KT21MG1 human meningioma were administered mebendazole alone or in combination with radiation. Survival benefit was evaluated, while tumors were investigated by immunohistochemical staining for apoptosis, cell proliferation, and vascular density. RESULTS In vitro experiments on meningioma cell lines showed the IC50 for mebendazole in the range of 0.26-0.42 µM. Mebendazole alone induced cytotoxicity, however the combination had a greater reduction in colony formation and resulted in higher levels of cleaved caspase-3. The in vivo study showed both, mebendazole alone and the combination, to have a survival benefit with an increase in apoptosis, and decreases in tumor cell and vascular proliferation. CONCLUSION These preclinical findings indicate that mebendazole alone or in combination with radiation can be considered for the treatment of malignant meningioma. The mechanism of action for this combination may include an increase in apoptosis, a reduction in proliferation and angiogenesis, or a combination of these effects.
Collapse
|
26
|
Yen TH, Hsieh CL, Liu TT, Huang CS, Chen YC, Chuang YC, Lin SS, Hsu FT. Amentoflavone Induces Apoptosis and Inhibits NF-ĸB-modulated Anti-apoptotic Signaling in Glioblastoma Cells. ACTA ACUST UNITED AC 2018; 32:279-285. [PMID: 29475910 DOI: 10.21873/invivo.11235] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Revised: 12/14/2017] [Accepted: 12/18/2017] [Indexed: 01/08/2023]
Abstract
The goal of the present study was to investigate anticancer effect of amentoflavone on glioblastoma cells in vitro. Our results demonstrated that amentoflavone not only significantly reduced cell viability, nuclear factor-ĸappa B (NF-ĸB) activation, and protein expression of cellular Fas-associated protein with death domain-like interleukin 1 beta-converting enzyme inhibitory protein (C-FLIP) and myeloid cell leukemia 1 (MCL1), but significantly triggered cell accumulation at the sub-G1 phase, loss of mitochondrial membrane potential, and expression of active caspase-3 and -8. In order to verify the effect of NF-ĸB inhibitor on expression of anti-apoptotic proteins, we performed western blotting. We found that the of NF-ĸB inhibitor or amentoflavone markedly diminished protein levels of MCL1 and C-FLIP. Taken all together, our findings show that amentoflavone induces intrinsic and extrinsic apoptosis and inhibits NF-ĸB-modulated anti-apoptotic signaling in U-87 MG cells in vitro.
Collapse
Affiliation(s)
- Tsung-Hsien Yen
- Department of Radiology, Cheng Ching General Hospital, Taichung, Taiwan, R.O.C
| | - Chia-Ling Hsieh
- The Ph.D. Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan, R.O.C
| | - Tsu-Te Liu
- Division of Gastroenterology, Department of Internal Medicine, National Yang-Ming University Hospital, Yilan, Taiwan, R.O.C
| | - Chih-Sheng Huang
- Department of Surgery, National Yang-Ming University Hospital, Yilan, Taiwan, R.O.C.,Department of Surgery, School of Medicine, National Yang-Ming University, Taipei, Taiwan, R.O.C
| | - Yen-Chung Chen
- Department of Pathology, National Yang-Ming University Hospital, Yilan, Taiwan, R.O.C
| | | | - Song-Shei Lin
- Department of Medical Imaging and Radiological Sciences, Central-Taiwan University of Science and Technology, Taichung, Taiwan, R.O.C.
| | - Fei-Ting Hsu
- Department of Radiology, School of Medicine, Taipei Medical University, Taipei, Taiwan, R.O.C. .,Department of Medical Imaging, Taipei Medical University Hospital, Taipei, Taiwan, R.O.C.,Research Center of Translational Imaging, College of Medicine, Taipei Medical University, Taipei, Taiwan, R.O.C.,Translational Laboratory, Department of Medical Research, Taipei Medical University Hospital, Taipei, Taiwan, R.O.C
| |
Collapse
|
27
|
He MH, Chen L, Zheng T, Tu Y, He Q, Fu HL, Lin JC, Zhang W, Shu G, He L, Yuan ZX. Potential Applications of Nanotechnology in Urological Cancer. Front Pharmacol 2018; 9:745. [PMID: 30038573 PMCID: PMC6046453 DOI: 10.3389/fphar.2018.00745] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Accepted: 06/19/2018] [Indexed: 01/16/2023] Open
Abstract
Nowadays, the potential scope of nanotechnology in uro-oncology (cancers of the prostate, bladder, and kidney) is broad, ranging from drug delivery, prevention, and diagnosis to treatment. Novel drug delivery methods using magnetic nanoparticles, gold nanoparticles, and polymeric nanoparticles have been investigated in prostate cancer. Additionally, renal cancer treatment may be profoundly influenced by applications of nanotechnology principles. Various nanoparticle-based strategies for kidney cancer therapy have been proposed. Partly due to the dilution of drug concentrations by urine production, causing inadequate drug delivery to tumor cells in the treatment of bladder cancer, various multifunctional bladder-targeted nanoparticles have been developed to enhance therapeutic efficiency. In each of these cancer research fields, nanotechnology has shown several advantages over widely used traditional methods. Different types of nanoparticles improve the solubility of poorly soluble drugs, and multifunctional nanoparticles have good specificity toward prostate, renal, and bladder cancer. Moreover, nanotechnology can also combine with other novel technologies to further enhance effectivity. As our understanding of nanotechnologies grows, additional opportunities to improve the diagnosis and treatment of urological cancer are excepted to arise. In this review, we focus on nanotechnologies with potential applications in urological cancer therapy and highlight clinical areas that would benefit from nanoparticle therapy.
Collapse
Affiliation(s)
- Ming-Hui He
- Department of Pharmacy, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Li Chen
- Department of Pharmacy, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Ting Zheng
- Department of Pharmacy, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Yu Tu
- Department of Pharmacy, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Qian He
- Department of Pharmacy, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Hua-Lin Fu
- Department of Pharmacy, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Ju-Chun Lin
- Department of Pharmacy, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Wei Zhang
- Department of Pharmacy, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Gang Shu
- Department of Pharmacy, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Lili He
- College of Pharmacy, Southwest Minzu University, Chengdu, China
| | - Zhi-Xiang Yuan
- Department of Pharmacy, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| |
Collapse
|
28
|
Li M, Zhang F, Su Y, Zhou J, Wang W. Nanoparticles designed to regulate tumor microenvironment for cancer therapy. Life Sci 2018; 201:37-44. [PMID: 29577880 DOI: 10.1016/j.lfs.2018.03.044] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Revised: 03/12/2018] [Accepted: 03/21/2018] [Indexed: 02/08/2023]
Abstract
Increasing understanding in tumor pathology reveals that tumor microenvironment (TME), which supports tumor progression and poses barriers for available therapies, takes a great responsibility in inefficient treatment and poor prognosis. In recent years, the versatile nanotechnology employed in TME regulation has made great progress. The nanoparticles (NPs) can be tailored as needed to accurately target TME components by distinguishing healthy tissues from malignancy, and to regulate TME to promote tumor regression. Meanwhile, the emerging microRNAs (miRNAs) demonstrate great potentials for TME regulation, but are regrettably restricted by quick degradation. NPs systems enable the successful delivery of miRNA to TME without the limitation, expanding the application of nucleic acid drug. In this review, we summarized recent NPs-based strategies aiming at regulating TME in different ways, including anti-angiogenesis, extracellular matrix (ECM) remodeling, tumor-associated fibroblasts (TAFs) treatment and tumor-associated macrophages (TAMs) treatment, along with the miRNAs-loaded NPs for TME regulation. Catching and utilizing the features of TME for NPs design can contribute to reversing drug-resistance, optimized drug distribution, and eventually more efficient cancer therapy.
Collapse
Affiliation(s)
- Min Li
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, China
| | - Fangrong Zhang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, China
| | - Yujie Su
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, China
| | - Jianping Zhou
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, China.
| | - Wei Wang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, China.
| |
Collapse
|
29
|
Chio CC, Chen KY, Chang CK, Chuang JY, Liu CC, Liu SH, Chen RM. Improved effects of honokiol on temozolomide-induced autophagy and apoptosis of drug-sensitive and -tolerant glioma cells. BMC Cancer 2018; 18:379. [PMID: 29614990 PMCID: PMC5883267 DOI: 10.1186/s12885-018-4267-z] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 03/20/2018] [Indexed: 01/08/2023] Open
Abstract
Background Temozolomide (TMZ)-induced side effects and drug tolerance to human gliomas are still challenging issues now. Our previous studies showed that honokiol, a major bioactive constituent of Magnolia officinalis (Houpo), is safe for normal brain cells and can kill human glioma cells. This study was further aimed to evaluate the improved effects of honokiol and TMZ on drug-sensitive and -resistant glioma cells and the possible mechanisms. Methods TMZ-sensitive human U87-MG and murine GL261 glioma cells and TMZ-resistant human U87-MR-R9 glioma cells were exposed to honokiol and TMZ, and cell viability and LC50 of honokiol were assayed. To determine the death mechanisms, caspase-3 activity, DNA fragmentation, apoptotic cells, necrotic cells, cell cycle, and autophagic cells. The glioma cells were pretreated with 3-methyladenine (3-MA) and chloroquine (CLQ), two inhibitors of autophagy, and then exposed to honokiol or TMZ. Results Exposure of human U87-MG glioma cells to honokiol caused cell death and significantly enhanced TMZ-induced insults. As to the mechanism, combined treatment of human U87-MG cells with honokiol and TMZ induced greater caspase-3 activation, DNA fragmentation, cell apoptosis, and cell-cycle arrest at the G1 phase but did not affect cell necrosis. The improved effects of honokiol on TMZ-induced cell insults were further verified in mouse GL261 glioma cells. Moreover, exposure of drug-tolerant human U87-MG-R9 cells to honokiol induced autophagy and consequent apoptosis. Pretreatments with 3-MA and CLQ caused significant attenuations in honokiol- and TMZ-induced cell autophagy and apoptosis in human TMZ-sensitive and -tolerant glioma cells. Conclusions Taken together, this study demonstrated the improved effects of honokiol with TMZ on autophagy and subsequent apoptosis of drug-sensitive and -tolerant glioma cells. Thus, honokiol has the potential to be a drug candidate for treating human gliomas.
Collapse
Affiliation(s)
- Chung-Ching Chio
- Department of Neurosurgery, Chi-Mei Medical Center, Tainan, Taiwan.,Graduate Institute of Medical Sciences, College of Medicine and Comprehensive Cancer Center, Taipei Medical University, 250 Wu-Hsing St., Taipei, 110, Taiwan
| | - Kung-Yen Chen
- Graduate Institute of Medical Sciences, College of Medicine and Comprehensive Cancer Center, Taipei Medical University, 250 Wu-Hsing St., Taipei, 110, Taiwan.,Cellular Physiology and Molecular Image Research Center and Department of Anesthesiology, Wan-Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Cheng-Kuei Chang
- Department of Neurosurgery, Shuang Ho Hospital, Taipei Medical University, Taipei, Taiwan
| | - Jian-Ying Chuang
- Graduate Institute of Neural Regenerative Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chih-Chung Liu
- Anesthesiology and Health Policy Research Center and Department of Anesthesiology, Taipei Medical University Hospital, Taipei Medical University, Taipei, Taiwan
| | - Shing-Hwa Liu
- Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Ruei-Ming Chen
- Graduate Institute of Medical Sciences, College of Medicine and Comprehensive Cancer Center, Taipei Medical University, 250 Wu-Hsing St., Taipei, 110, Taiwan. .,Cellular Physiology and Molecular Image Research Center and Department of Anesthesiology, Wan-Fang Hospital, Taipei Medical University, Taipei, Taiwan. .,Anesthesiology and Health Policy Research Center and Department of Anesthesiology, Taipei Medical University Hospital, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
30
|
Lin PI, Tai YT, Chan WP, Lin YL, Liao MH, Chen RM. Estrogen/ERα signaling axis participates in osteoblast maturation via upregulating chromosomal and mitochondrial complex gene expressions. Oncotarget 2017; 9:1169-1186. [PMID: 29416685 PMCID: PMC5787428 DOI: 10.18632/oncotarget.23453] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Accepted: 12/09/2017] [Indexed: 01/08/2023] Open
Abstract
Estrogen deficiency usually leads to bone loss and osteoporosis in postmenopausal women. Osteoblasts play crucial roles in bone formation. However, osteoblast functions are influenced by mitochondrial bioenergetic conditions. In this study, we investigated the roles of the estrogen and estrogen receptor alpha (ERα) axis in mitochondrial energy metabolism and subsequent osteoblast mineralization. Exposure of rat calvarial osteoblasts to estradiol caused substantial improvements in alkaline phosphatase activities and cell calcification. In parallel, treatment of human osteoblast-like U2OS cells, derived from a female osteosarcoma patient, with estradiol specifically augmented ERα levels. Sequentially, estradiol stimulated translocation of ERα to nuclei in human osteoblasts and induced expressions of genomic respiratory chain complex NDUFA10, UQCRC1, cytochrome c oxidase (COX)8A, COX6A2, COX8C, COX6C, COX6B2, COX412, and ATP12A genes. Concurrently, estradiol stimulated translocation of ERα to mitochondria from the cytoplasm. A bioinformatic search found the existence of four estrogen response elements in the 5’-promoter region of the mitochondrial cox i gene. Interestingly, estradiol induced COX I mRNA and protein expressions in human osteoblasts or rat calvarial osteoblasts. Knocking-down ERα translation concurrently downregulated estradiol-induced COX I mRNA expression. Consequently, exposure to estradiol led to successive increases in the mitochondrial membrane potential, the mitochondrial enzyme activity, and cellular adenosine triphosphate levels. Taken together, this study showed the roles of the estradiol/ERα signaling axis in improving osteoblast maturation through upregulating the mitochondrial bioenergetic system due to induction of definite chromosomal and mitochondrial complex gene expressions. Our results provide novel insights elucidating the roles of the estrogen/ERα alliance in regulating bone formation.
Collapse
Affiliation(s)
- Pei-I Lin
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yu-Ting Tai
- Cell Physiology and Molecular Image Research Center and Department of Anesthesiology, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Wing P Chan
- Department of Radiology, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Yi-Ling Lin
- Cell Physiology and Molecular Image Research Center and Department of Anesthesiology, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Mei-Hsiu Liao
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Cell Physiology and Molecular Image Research Center and Department of Anesthesiology, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Ruei-Ming Chen
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Cell Physiology and Molecular Image Research Center and Department of Anesthesiology, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan.,Anesthesiology and Health Policy Research Center, Taipei Medical University Hospital, Taipei, Taiwan
| |
Collapse
|
31
|
Li F, Jiang T, Li Q, Ling X. Camptothecin (CPT) and its derivatives are known to target topoisomerase I (Top1) as their mechanism of action: did we miss something in CPT analogue molecular targets for treating human disease such as cancer? Am J Cancer Res 2017; 7:2350-2394. [PMID: 29312794 PMCID: PMC5752681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Accepted: 11/07/2017] [Indexed: 06/07/2023] Open
Abstract
Camptothecin (CPT) was discovered from plant extracts more than 60 years ago. Since then, only two CPT analogues (irinotecan and topotecan) have been approved for cancer treatment, although several thousand CPT derivatives have been synthesized and many of them were actively studied in our research community over the past 6+ decades. In this review article, we briefly summarize: (1) the discovery and early development of CPTs, (2) the recognized CPT mechanism of action (MOA), (3) the synthesis of CPT and CPT analogues, and (4) the structure-activity relationship (SAR) of CPT and its analogues. Next, we provide evidence that certain CPT analogues can exert improved efficacy with low toxicity independently of topoisomerase I (Top1) inhibition; instead, these CPT analogues use novel MOAs by targeting important cancer survival-associated oncogenic proteins and/or by bypassing various treatment-resistant mechanisms. We then present a comprehensive review of the most advanced CPT analogues in clinical development, with the goal of resolving why no new CPTs have been FDA approved for cancer treatment, beyond irinotecan and topotecan. We argue that new CPT Top1 inhibitor drugs are unlikely being found to be significantly better than irinotecan and/or topotecan in terms of the overall antitumor activity and toxicity. The significance of CPT analogues that possess novel MOAs has not been sufficiently recognized so far. In our opinion, this is a research area with great potential to make a breakthrough for development of the next generation of CPT analogues that possess high efficacy (due to novel targets) and low toxicity (due to low inhibition of Top1 activity/function) for effective treatment of human disease, including cancer.
Collapse
Affiliation(s)
- Fengzhi Li
- Department of Pharmacology & Therapeutics, Roswell Park Cancer InstituteBuffalo, New York, USA
| | - Tao Jiang
- Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology, School of Medicine and Pharmacy, Ocean University of ChinaQingdao, China
| | - Qingyong Li
- Collaborative Innovation Center of Yangtze River Region Green Pharmaceuticals, Zhejiang University of TechnologyHangzhou, China
| | - Xiang Ling
- Department of Pharmacology & Therapeutics, Roswell Park Cancer InstituteBuffalo, New York, USA
- Canget BioTekpharmaBuffalo, New York, USA
| |
Collapse
|
32
|
Voss MH, Hussain A, Vogelzang N, Lee JL, Keam B, Rha SY, Vaishampayan U, Harris WB, Richey S, Randall JM, Shaffer D, Cohn A, Crowell T, Li J, Senderowicz A, Stone E, Figlin R, Motzer RJ, Haas NB, Hutson T. A randomized phase II trial of CRLX101 in combination with bevacizumab versus standard of care in patients with advanced renal cell carcinoma. Ann Oncol 2017; 28:2754-2760. [PMID: 28950297 DOI: 10.1093/annonc/mdx493] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Nanoparticle-drug conjugates enhance drug delivery to tumors. Gradual payload release inside cancer cells augments antitumor activity while reducing toxicity. CRLX101 is a novel nanoparticle-drug conjugate containing camptothecin, a potent inhibitor of topoisomerase I and the hypoxia-inducible factors 1α and 2α. In a phase Ib/2 trial, CRLX101 + bevacizumab was well tolerated with encouraging activity in metastatic renal cell carcinoma (mRCC). We conducted a randomized phase II trial comparing CRLX101 + bevacizumab versus standard of care (SOC) in refractory mRCC. PATIENTS AND METHODS Patients with mRCC and 2-3 prior lines of therapy were randomized 1 : 1 to CRLX101 + bevacizumab versus SOC, defined as investigator's choice of any approved regimen not previously received. The primary end point was progression-free survival (PFS) by blinded independent radiological review in patients with clear cell mRCC. Secondary end points included overall survival, objective response rate and safety. RESULTS In total, 111 patients were randomized and received ≥1 dose of drug (CRLX101 + bevacizumab, 55; SOC, 56). Within the SOC arm, patients received single-agent bevacizumab (19), axitinib (18), everolimus (7), pazopanib (4), sorafenib (4), sunitinib (2), or temsirolimus (2). In the clear cell population, the median PFS on the CRLX101 + bevacizumab and SOC arms was 3.7 months (95% confidence interval, 2.0-4.3) and 3.9 months (95% confidence interval 2.2-5.4), respectively (stratified log-rank P = 0.831). The objective response rate by IRR was 5% with CRLX101 + bevacizumab versus 14% with SOC (Mantel-Haenszel test, P = 0.836). Consistent with previous studies, the CRLX101 + bevacizumab combination was generally well tolerated, and no new safety signal was identified. CONCLUSIONS Despite promising efficacy data on the earlier phase Ib/2 trial of mRCC, this randomized trial did not demonstrate improvement in PFS for the CRLX101 + bevacizumab combination when compared with approved agents in patients with heavily pretreated clear cell mRCC. Further development in this disease is not planned. CLINICAL TRIAL IDENTIFICATION NCT02187302 (NIH).
Collapse
Affiliation(s)
- M H Voss
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York. mailto:
| | - A Hussain
- Department of Medicine, Greenebaum Cancer Center, University of Maryland, Baltimore
| | - N Vogelzang
- Department of Hematology/Oncology, Comprehensive Cancer Centers of Nevada, Las Vegas; US Oncology Research, USA
| | - J L Lee
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul
| | - B Keam
- Department of Internal Medicine, Seoul National University Hospital, Seoul
| | - S Y Rha
- Department of Medicine, Severance Hospital, Seoul, Korea
| | - U Vaishampayan
- Department of Oncology, Karmanos Cancer Institute, Detroit
| | - W B Harris
- Department of Hematology/Oncology, Emory University Winship Cancer Institute, Atlanta
| | - S Richey
- US Oncology Research, USA; Department of Medicine, Texas Oncology, Fort Worth
| | - J M Randall
- Department of Medicine, University of California, San Diego, La Jolla
| | - D Shaffer
- US Oncology Research, USA; Department of Medicine, Albany Medical Center, NYOH, Albany
| | - A Cohn
- US Oncology Research, USA; Department of Clinical Research, Rocky Mountain Cancer Centers, Denver
| | - T Crowell
- Department of Medicine, Cerulean Pharma Inc., Waltham
| | - J Li
- Department of Medicine, Cerulean Pharma Inc., Waltham
| | - A Senderowicz
- Department of Medicine, Cerulean Pharma Inc., Waltham
| | - E Stone
- Department of Medicine, Cerulean Pharma Inc., Waltham
| | - R Figlin
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles
| | - R J Motzer
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York
| | - N B Haas
- Department of Medicine, Abramson Cancer Center, University of Pennsylvania, Philadelphia
| | - T Hutson
- US Oncology Research, USA; Department of Medicine, Texas Oncology, Dallas, USA
| |
Collapse
|
33
|
Wu GJ, Chen JT, Tsai HC, Chen TL, Liu SH, Chen RM. Protection of Dexmedetomidine Against Ischemia/Reperfusion-Induced Apoptotic Insults to Neuronal Cells Occurs Via an Intrinsic Mitochondria-Dependent Pathway. J Cell Biochem 2017; 118:2635-2644. [DOI: 10.1002/jcb.25847] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2016] [Accepted: 12/15/2016] [Indexed: 01/26/2023]
Affiliation(s)
- Gong-Jhe Wu
- Department of Anesthesiology; Shin Kong Wu Ho-Su Memorial Hospital; Taipei Taiwan
- Comprehensive Cancer Center; Taipei Medical University; Taipei Taiwan
| | - Jui-Tai Chen
- Graduate Institute of Medical Sciences; College of Medicine; Taipei Medical University; Taipei Taiwan
| | - Hsiao-Chien Tsai
- Anesthesiology and Health Policy Research Center; Taipei Medical University; Taipei Taiwan
| | - Ta-Liang Chen
- Anesthesiology and Health Policy Research Center; Taipei Medical University; Taipei Taiwan
| | - Shing-Hwa Liu
- Institute of Toxicology, College of Medicine; National Taiwan University; Taipei Taiwan
| | - Ruei-Ming Chen
- Comprehensive Cancer Center; Taipei Medical University; Taipei Taiwan
- Graduate Institute of Medical Sciences; College of Medicine; Taipei Medical University; Taipei Taiwan
- Anesthesiology and Health Policy Research Center; Taipei Medical University; Taipei Taiwan
| |
Collapse
|
34
|
Cheng BC, Chen JT, Yang ST, Chio CC, Liu SH, Chen RM. Cobalt chloride treatment induces autophagic apoptosis in human glioma cells via a p53-dependent pathway. Int J Oncol 2017; 50:964-974. [PMID: 28197638 DOI: 10.3892/ijo.2017.3861] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Accepted: 01/13/2017] [Indexed: 12/09/2022] Open
Abstract
Malignant glioma is the most aggressive brain tumor. Hypoxic condition has been explored for killing cancer stem cells or drug-resistant tumor cells. This study investigated the effects of hypoxia on autophagic death and the possible mechanisms. Exposure of human malignant glioma U87-MG cells to cobalt chloride (CoCl2) increased cellular hypoxia-inducible factor-1α levels and concurrently decreased cell viability concentration- and time-dependently. In parallel, treatment with CoCl2 suppressed proliferation of human U87-MG cells. Autophagic cells and levels of LC3-II were concentration- and time-dependently induced in human U87-MG cells after exposure to CoCl2. However, pretreatment with 3-mehyladenine (3-MA) and chloroquine, inhibitors of cell autophagy, caused significant alleviations in CoCl2-induced cell autophagy. In contrast, exposure to rapamycin, an inducer of cell autophagy, synergistically induced hypoxia-induced autophagy of U87-MG cells. Administration of human U87-MG cells with CoCl2 triggered caspase-3 activation and cell apoptosis. Interestingly, pretreatment with 3-MA and chloroquine remarkably suppressed CoCl2-induced caspase-3 activation and cell apoptosis. Application of p53 small interference (si)RNA into human U87-MG cells downregulated levels of this protein and simultaneously lowered hypoxia- and 3-MA-induced alterations in cell autophagy, apoptosis, and death. The hypoxia-induced autophagy and apoptosis of DBTRG-05MG cells were significantly lowered by 3-MA pretreatment and p53 knockdown. Therefore, the present study shows that CoCl2 treatment can induce autophagy of human glioma cells and subsequent autophagic apoptosis via a p53-dependent pathway. Hypoxia-induced autophagic apoptosis may be applied as a therapeutic strategy for treatment of glioma patients.
Collapse
Affiliation(s)
- Bor-Chin Cheng
- Department of Surgery, Chi-Mei Medical Center, Tainan, Taiwan, R.O.C
| | - Jui-Tai Chen
- Graduate Institute of Medical Sciences, Taipei Medical University, Taipei, Taiwan, R.O.C
| | - Shun-Tai Yang
- Department of Neurosurgery, Shuang-Ho Hospital, Taipei Medical University, New Taipei City, Taiwan, R.O.C
| | - Chung-Ching Chio
- Department of Neurosurgery, Chi-Mei Medical Center, Tainan, Taiwan, R.O.C
| | - Shing-Hwa Liu
- Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, Taiwan, R.O.C
| | - Ruei-Ming Chen
- Comprehensive Cancer Center, Taipei Medical University, Taipei, Taiwan, R.O.C
| |
Collapse
|
35
|
Martino E, Della Volpe S, Terribile E, Benetti E, Sakaj M, Centamore A, Sala A, Collina S. The long story of camptothecin: From traditional medicine to drugs. Bioorg Med Chem Lett 2016; 27:701-707. [PMID: 28073672 DOI: 10.1016/j.bmcl.2016.12.085] [Citation(s) in RCA: 183] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2016] [Revised: 12/28/2016] [Accepted: 12/29/2016] [Indexed: 12/11/2022]
Abstract
20-(S)-Camptothecin (CPT) is a natural alkaloid extracted from the bark of Camptotheca acuminata (Chinese happy tree). It acts as a DNA topoisomerase 1 poison with an interesting antitumor activity and its use is limited by low stability and solubility and unpredictable drug-drug interactions. Since the late 20th century, it has been widely used in cancer therapy and, since extraction yields from plant tissues are very low, various synthetic routes have been developed to satisfy the increase in demand for CPT. Moreover, SAR studies have allowed for the development of more potent CPT analogues topotecan and irinotecan. Unfortunately, resistance has already occurred in several tumour lines. Additional studies are needed to better understand the relationship between substituents and resistance, its clinical relevance and the impact of related gene polymorphism. One of the latest research approaches focuses on modifying the delivery mode to improve tumour cell uptake and reduce toxicity.
Collapse
Affiliation(s)
- Emanuela Martino
- Department of Earth and Environmental Sciences, University of Pavia, Via S. Epifanio 14, 27100 Pavia, Italy
| | - Serena Della Volpe
- Department of Drug Sciences, Medicinal Chemistry and Pharmaceutical Technology Section, University of Pavia, Viale Taramelli 12, 27100 Pavia, Italy
| | - Elisa Terribile
- Department of Drug Sciences, Medicinal Chemistry and Pharmaceutical Technology Section, University of Pavia, Viale Taramelli 12, 27100 Pavia, Italy
| | - Emanuele Benetti
- Department of Drug Sciences, Medicinal Chemistry and Pharmaceutical Technology Section, University of Pavia, Viale Taramelli 12, 27100 Pavia, Italy
| | - Mirena Sakaj
- Department of Drug Sciences, Medicinal Chemistry and Pharmaceutical Technology Section, University of Pavia, Viale Taramelli 12, 27100 Pavia, Italy
| | - Adriana Centamore
- Department of Drug Sciences, Medicinal Chemistry and Pharmaceutical Technology Section, University of Pavia, Viale Taramelli 12, 27100 Pavia, Italy
| | - Andrea Sala
- Department of Drug Sciences, Medicinal Chemistry and Pharmaceutical Technology Section, University of Pavia, Viale Taramelli 12, 27100 Pavia, Italy
| | - Simona Collina
- Department of Drug Sciences, Medicinal Chemistry and Pharmaceutical Technology Section, University of Pavia, Viale Taramelli 12, 27100 Pavia, Italy.
| |
Collapse
|
36
|
Yuan ZX, Mo J, Zhao G, Shu G, Fu HL, Zhao W. Targeting Strategies for Renal Cell Carcinoma: From Renal Cancer Cells to Renal Cancer Stem Cells. Front Pharmacol 2016; 7:423. [PMID: 27891093 PMCID: PMC5103413 DOI: 10.3389/fphar.2016.00423] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Accepted: 10/25/2016] [Indexed: 12/14/2022] Open
Abstract
Renal cell carcinoma (RCC) is a common form of urologic tumor that originates from the highly heterogeneous epithelium of renal tubules. Over the last decade, targeting therapies to renal cancer cells have transformed clinical care for RCC. Recently, it was proposed that renal cancer stem cells (CSCs) isolated from renal carcinomas were responsible for driving tumor growth and resistance to conventional chemotherapy and radiotherapy, according to the theory of CSCs; this has provided the rationale for therapies targeting this aggressive cell population. Precise identification of renal CSC populations and the complete cell hierarchy will accurately inform characterization of disease subtypes. This will ultimately contribute to more personalized and targeted therapies. Here, we summarize potential targeting strategies for renal cancer cells and renal CSCs, including tyrosine kinase inhibitors, mammalian target of rapamycin inhibitors (mTOR), interleukins, CSC marker inhibitors, bone morphogenetic protein-2, antibody drug conjugates, and nanomedicine. In conclusion, targeting therapies for RCC represent new directions for exploration and clinical investigation and they plant a seed of hope for advanced clinical care.
Collapse
Affiliation(s)
- Zhi-Xiang Yuan
- Department of Pharmacy, College of Veterinary Medicine, Sichuan Agricultural University Chengdu, China
| | - Jingxin Mo
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen UniversityGuangzhou, China; Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen UniversityGuangzhou, China
| | - Guixian Zhao
- Department of Pharmacy, College of Veterinary Medicine, Sichuan Agricultural University Chengdu, China
| | - Gang Shu
- Department of Pharmacy, College of Veterinary Medicine, Sichuan Agricultural University Chengdu, China
| | - Hua-Lin Fu
- Department of Pharmacy, College of Veterinary Medicine, Sichuan Agricultural University Chengdu, China
| | - Wei Zhao
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen UniversityGuangzhou, China; Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen UniversityGuangzhou, China
| |
Collapse
|