1
|
Karimi S, Bakhshali R, Bolandi S, Zahed Z, Mojtaba Zadeh SS, Kaveh Zenjanab M, Jahanban Esfahlan R. For and against tumor microenvironment: Nanoparticle-based strategies for active cancer therapy. Mater Today Bio 2025; 31:101626. [PMID: 40124335 PMCID: PMC11926801 DOI: 10.1016/j.mtbio.2025.101626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 02/22/2025] [Accepted: 02/27/2025] [Indexed: 03/25/2025] Open
Abstract
Cancer treatment is challenged by the tumor microenvironment (TME), which promotes drug resistance and cancer cell growth. This review offers a comprehensive and innovative perspective on how nanomedicine can modify the TME to enhance therapy. Strategies include using nanoparticles to improve oxygenation, adjust acidity, and alter the extracellular matrix, making treatments more effective. Additionally, nanoparticles can enhance immune responses by activating immune cells and reducing suppression within tumors. By integrating these approaches with existing therapies, such as chemotherapy and radiotherapy, nanoparticles show promise in overcoming traditional treatment barriers. The review discusses how changes in the TME can enhance the effectiveness of nanomedicine itself, creating a reciprocal relationship that boosts overall efficacy. We also highlight novel strategies aimed at exploiting and overcoming the TME, leveraging nanoparticle-based approaches for targeted cancer therapy through precise TME modulation.
Collapse
Affiliation(s)
- Soroush Karimi
- Nano Drug Delivery Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | | | | | - Zahra Zahed
- Department of Medical Sciences, Ardabil University of Medical Sciences, Ardabil, Iran
| | | | - Masoumeh Kaveh Zenjanab
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Rana Jahanban Esfahlan
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
2
|
Qaderi K, Shahmoradi A, Thyagarajan A, Sahu RP. Impact of targeting the platelet-activating factor and its receptor in cancer treatment. Mil Med Res 2025; 12:10. [PMID: 40033370 PMCID: PMC11877967 DOI: 10.1186/s40779-025-00597-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 01/26/2025] [Indexed: 03/05/2025] Open
Abstract
The lipid mediator platelet-activating factor (PAF) and its receptor (PAFR) signaling play critical roles in a wide range of physiological and pathophysiological conditions, including cancer growth and metastasis. The ability of PAFR to interact with other oncogenic signaling cascades makes it a promising target for cancer treatment. Moreover, numerous natural and synthetic compounds, characterized by diverse pharmacological activities such as anti-inflammatory and anti-tumor effects, have been explored for their potential as PAF and PAFR antagonists. In this review, we provide comprehensive evidence regarding the PAF/PAFR signaling pathway, highlighting the effectiveness of various classes of PAF and PAFR inhibitors and antagonists across multiple cancer models. Notably, the synergistic effects of PAF and PAFR antagonists in enhancing the efficacy of chemotherapy and radiation therapy in several experimental cancer models are also discussed. Overall, the synthesis of literature review indicates that targeting the PAF/PAFR axis represents a promising approach for cancer treatment and also exerts synergy with chemotherapy and radiation therapy.
Collapse
Affiliation(s)
- Kimya Qaderi
- Department of Molecular and Cell Biology, College of Life Sciences, University of Leicester, University Road, Leicester, LE1 7RH, UK
| | - Arvin Shahmoradi
- Department of Laboratory Medicine, Faculty of Paramedical, Kurdistan University of Medical Sciences, Sanandaj, 66177-13446, Kurdistan, Iran
| | - Anita Thyagarajan
- Department of Pharmacology and Toxicology, Boonshoft School of Medicine Wright State University, Dayton, OH, 45435, USA
| | - Ravi P Sahu
- Department of Pharmacology and Toxicology, Boonshoft School of Medicine Wright State University, Dayton, OH, 45435, USA.
| |
Collapse
|
3
|
Panchal I, Tripathi RKP, Yadav MR, Valera M, Parmar K. Design, Synthesis, Biological and in silico Evaluation of Novel Indazole-pyridine Hybrids for the Treatment of Breast Cancer. Curr Comput Aided Drug Des 2025; 21:211-225. [PMID: 39108123 DOI: 10.2174/0115734099308839240724100224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 06/12/2024] [Accepted: 06/13/2024] [Indexed: 01/03/2025]
Abstract
INTRODUCTION The prevalence of breast cancer presents a substantial global health concern, underscoring the ongoing need for the development of inventive therapeutic remedies. METHODS In this investigation, an array of novel indazole-pyridine hybrids (5a-h) have been designed and synthesized to assess their potential as candidates for treating breast cancer. Subsequently, we have conducted biological evaluations to determine their cytotoxic effects on the human MCF-7 breast cancer cell line. Furthermore, in silico analysis was conducted to estimate the inhibition potential of the compounds against TrkA (Tropomyosin receptor kinase A), a specific molecular target associated with breast cancer, through molecular docking. In silico physicochemical and pharmacokinetic predictions were made to assess the compounds' drug-like properties. RESULTS Compound 5a emerged as the most active compound among the others with GI50 < 10 μg/ml. Besides, compound 5a showed high binding energy (BE -10.7 kcal/mol) against TrkA and was stabilized within the TrkA binding pocket through hydrophobic, H-bonding, and van der Waals interactions. In silico physicochemical and pharmacokinetic prediction studies indicated that compound 5a obeyed both Lipinski's and Veber's rule and displayed a versatile pharmacokinetic profile, implying compound 5a to appear as a viable candidate and that it could be further refined to develop therapeutic agents for potentially treating breast cancer. CONCLUSION This study offers a promising direction for the advancement of innovative breast cancer treatments, highlighting the effectiveness of indazole-pyridine hybrids as potential anticancer agents. Further optimization and preclinical development are necessary to advance these compounds to clinical trials.
Collapse
Affiliation(s)
- Ishan Panchal
- Department of Pharmaceutical Chemistry, Parul Institute of Pharmacy, Parul University, Vadodara, Gujarat, 391760, India
- Department of Pharmaceutical Chemistry, Arihant School of Pharmacy and Bio-Research Institute, Gandhinagar, Gujarat, 382421, India
| | - Rati Kailash Prasad Tripathi
- Department of Pharmaceutical Sciences, Sushruta School of Medical and Paramedical Sciences, Assam University (A Central University), Silchar, Assam, 788011, India
| | - Mange Ram Yadav
- Director (R & D), Research and Development Cell, Parul University, Vadodara, Gujarat, 391760, India
| | - Meet Valera
- Department of Pharmaceutical Chemistry, Parul Institute of Pharmacy, Parul University, Vadodara, Gujarat, 391760, India
| | - Kinjal Parmar
- Department of Quality Assurance, Parul Institute of Pharmacy & Research, Parul University, Vadodara, Gujarat, 391760, India
| |
Collapse
|
4
|
Awad MG, Hanafy NAN, Ali RA, Abd El-Monem DD, El-Shafiey SH, El-Magd MA. Exploring the therapeutic applications of nano-therapy of encapsulated cisplatin and anthocyanin-loaded multiwalled carbon nanotubes coated with chitosan-conjugated folic acid in targeting breast and liver cancers. Int J Biol Macromol 2024; 280:135854. [PMID: 39307483 DOI: 10.1016/j.ijbiomac.2024.135854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 09/14/2024] [Accepted: 09/19/2024] [Indexed: 09/26/2024]
Abstract
This study aimed to assess the targeted nano-therapy of encapsulated cisplatin (Cis) and anthocyanin (Ant)-loaded multiwalled carbon nanotubes (CNT) coated with chitosan conjugated folic acid on breast MCF7 and liver HepG2 cancer cells. Zeta potential, UV-spectroscopy, FTIR, TEM, and SEM were used to evaluate CNT, its modified form (CNT Mod), CNT-loaded Cis NPs, CNT-loaded Ant NPs, and CNT- Cis + Ant NPs. All treatments induced apoptosis-dependent cytotoxicity in both cell lines as revealed functionally by the MTT assay, morphologically (DNA degradation) by acridine orange/ethidium bromide (AO/EB) double staining, and molecularly (Bax upregulation and Bcl2 downregulation) by real-time PCR, with best effect for the combined treatment (CNT- Cis + Ant NPs). This combined treatment also significantly reduced inflammation (low TNFα), migration (low MMP9 and high TIMP1), and angiogenesis (low VEGF), while significantly increasing antioxidant status (high Nrf2 and OH-1) in MCF7 and HepG2 cells compared to other treatments. Interestingly, cells treated with CNT Mod exhibited higher cytotoxic, apoptotic, anti-migratory, and anti-angiogenic potentials relative to CNT-treated cells. In conclusion, targeted nano-therapy of encapsulated cisplatin and anthocyanin-loaded carbon nanotubes coated with chitosan conjugated folic acid can efficiently combat breast and liver cancers by sustained release, in addition to its apoptotic, antioxidant, anti-inflammatory, anti-metastatic, and anti-angiogenic effects.
Collapse
Affiliation(s)
- Mai G Awad
- Zoology Department, Faculty of Women for Arts Science and Education, Ain Shams University, 11757 Cairo, Egypt
| | - Nemany A N Hanafy
- Group of Bionanotechnology and Molecular Cell Biology, Institute of Nanoscience and Nanotechnology, Kafrelsheikh University, 33516 Kafrelsheikh, Egypt
| | - Ramadan A Ali
- Zoology Department, Faculty of Women for Arts Science and Education, Ain Shams University, 11757 Cairo, Egypt
| | - Dalia D Abd El-Monem
- Zoology Department, Faculty of Women for Arts Science and Education, Ain Shams University, 11757 Cairo, Egypt
| | - Sara H El-Shafiey
- Zoology Department, Faculty of Women for Arts Science and Education, Ain Shams University, 11757 Cairo, Egypt
| | - Mohammed A El-Magd
- Department of Anatomy, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafrelsheikh 33516, Egypt.
| |
Collapse
|
5
|
Jan K, Hassan N, James A, Hussain I, Rashid SM. Exploring molecular targets in cancer: Unveiling the anticancer potential of Paeoniflorin through a comprehensive analysis of diverse signaling pathways and recent advances. J Biol Methods 2024; 11:e99010014. [PMID: 39323487 PMCID: PMC11423941 DOI: 10.14440/jbm.2024.0003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 06/14/2024] [Accepted: 06/20/2024] [Indexed: 09/27/2024] Open
Abstract
Tumors have posed significant threats to human health for over 250 years, emerging as the foremost cause of death. While chemotherapeutic drugs are effective in treating tumors, their side effects can sometimes be challenging to manage during therapy. Nonetheless, there is growing interest in exploring natural compounds as alternatives, which potentially achieve therapeutic outcomes comparable to conventional chemotherapeutics with fewer adverse effects. Paeoniflorin (PF), a monoterpene glycoside derived from the root of Paeonia lactiflora, has garnered significant attention lately due to its promising anti-cancer properties. This review offers an updated outline of the molecular mechanisms underlying PF's anti-tumor function, with a focus on its modulation of various signaling pathways. PF exerts its anti-tumor activity by regulating crucial cellular processes including apoptosis, angiogenesis, proliferation, and metastasis. We explored the multifaceted impact of PF while modulating through signaling pathways, encompassing nuclear factor kappa B, NOTCH, caspase cascade, transforming growth factor-β, NEDD4, P53/14-3-3, STAT 3, MAPK, MMP-9, and SKP2 signaling pathways, highlighting its versatility in targeting diverse malignancies. Furthermore, we discuss future research directions aimed at exploring innovative and targeted cancer therapies facilitated by PF.
Collapse
Affiliation(s)
- Kounser Jan
- Division of Veterinary Biochemistry, Faculty of Veterinary Science and Animal Husbandry, Srinagar, Jammu and Kashmir, 190006, India
| | - Neelofar Hassan
- Division of Veterinary Biochemistry, Faculty of Veterinary Science and Animal Husbandry, Srinagar, Jammu and Kashmir, 190006, India
| | - Antonisamy James
- Departments of Medicinal and Biological Chemistry, The University of Toledo, Toledo, Ohio, 43614, United States of America
| | - Ishraq Hussain
- Division of Veterinary Biochemistry, Faculty of Veterinary Science and Animal Husbandry, Srinagar, Jammu and Kashmir, 190006, India
| | - Shahzada Mudasir Rashid
- Division of Veterinary Biochemistry, Faculty of Veterinary Science and Animal Husbandry, Srinagar, Jammu and Kashmir, 190006, India
- Departments of Medicinal and Biological Chemistry, The University of Toledo, Toledo, Ohio, 43614, United States of America
| |
Collapse
|
6
|
Zhai Y, Wang B, Han W, Yu B, Ci J, An F. Green synthesis of AgNPs using plant extract and investigation of its anti-human colorectal cancer application. OPEN CHEM 2023; 21. [DOI: 10.1515/chem-2023-0174] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025] Open
Abstract
Abstract
Recently, the plant extracts used to synthesize nanoparticles (NPs) have been considered an excellent alternative to physical and chemical ways. The applications of NPs in the fields of agriculture, industry, and medicine are so many and diverse that they cannot be counted. In recent years, silver nanoparticles (AgNPs) have attracted the consideration of several scientists because of their special characteristics and many applications in various fields, including optoelectronic catalysts, biological markers, and pharmaceutical and medical applications. In the current experiment, the cytotoxic potential of the properties of AgNPs green formulation using green tea on human colorectal cancer cells were determined. The NPs characterization was done by field emission-scanning electron microscopes, Fourier transform infrared spectroscopy, and X-ray diffraction analysis. The average diameter of the particles was about 35 nm. The presence of (111), (200), (220), and (311) peaks at the positions of 38°, 44°, 63°, and 77° indicate the presence of AgNPs, which confirms the correct synthesis of AgNPs. The 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-2H-tetrazolium bromide (MTT) assay was used to measure anti-colorectal carcinoma (on HCT-8, HT-29, MDST8, HCA-7 Colony 29, HCT 116, and Ramos.2G6.4C10 cells) properties of AgNPs. The findings indicate that in 3 days, the cancer cell survival percentage in various dilations reduced as much as the NPs concentration increased. The best anticancer effect was reported at 1,000 μg/mL dilation. The IC50 was 141, 46, 149, 125, 125, and 44 µg/mL against HCT-8, HT-29, MDST8, HCA-7 Colony 29, HCT 116, and Ramos.2G6.4C10 colorectal cancer cells, respectively. The results indicated that these NPs could inhibit colorectal cancer cells more strongly than normal cells. After doing the clinical trial studies, the recent AgNPs are a suitable option for colorectal cancer treatment.
Collapse
Affiliation(s)
- Yuze Zhai
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine , No. 16369, Jingshi Road , Jinan 250000, Shandong , China
| | - Benjun Wang
- Department of Anorectal Surgery, Affiliated Hospital of Shandong University of Traditional Chinese Medicine , No. 16369, Jingshi Road , Jinan 250000, Shandong , China
| | - Weiwei Han
- Department of Anorectal Surgery, Affiliated Hospital of Shandong University of Traditional Chinese Medicine , No. 16369, Jingshi Road , Jinan 250000, Shandong , China
| | - Bianfang Yu
- Department of Anorectal Surgery, Affiliated Hospital of Shandong University of Traditional Chinese Medicine , No. 16369, Jingshi Road , Jinan 250000, Shandong , China
| | - Jichen Ci
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine , No. 16369, Jingshi Road , Jinan 250000, Shandong , China
| | - Fan An
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine , No. 16369, Jingshi Road , Jinan 250000, Shandong , China
| |
Collapse
|
7
|
Abdullahi A, Wong TWL, Ng SSM. Putative role of non-invasive vagus nerve stimulation in cancer pathology and immunotherapy: Can this be a hidden treasure, especially for the elderly? Cancer Med 2023; 12:19081-19090. [PMID: 37587897 PMCID: PMC10557911 DOI: 10.1002/cam4.6466] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 07/09/2023] [Accepted: 07/16/2023] [Indexed: 08/18/2023] Open
Abstract
Cancer is globally a disease of significant public health concern owing to its prevalence, and association with morbidity and mortality. Thus, cost-effective treatments for cancer are important to help reduce its significant morbidity and mortality. However, the current therapeutic options for cancer such as chemotherapy, radiotherapy, and surgery may produce serious adverse events such as nausea, vomiting, fatigue, and peripheral neuropathy, especially in the long term. In addition, these therapeutic options may not be well tolerated by the elderly especially those who are frail. The current article is aimed at discussing an alternative therapeutic option, non-invasive vagus nerve stimulation (VNS), and the roles it plays in cancer pathology and immunotherapy. The VNS does this by reducing oxidative stress via silent information regulator 1 (SIRT1); inhibiting inflammation via both hypothalamic-pituitary-axis (HPA) and the release of corticosteroid from the adrenal gland, and cholinergic anti-inflammatory pathway (CAP), and increasing vagal activity which helps in the regulation of cell proliferation, differentiation, apoptosis, and metabolism, and increase chance of survival. Furthermore, it helps with reducing complications due to cancer or its treatments such as postoperative ileus and severity of peripheral neuropathy induced by chemotherapy, and improves cancer-related fatigue, lymphopenia, and quality of life. These suggest that the importance of non-invasive VNS in cancer pathology and immunotherapy cannot be overemphasized. Therefore, considering the safety of non-invasive VNS and its cost-effectiveness, it is a therapeutic option worth trying for these patients, especially in combination with other therapies.
Collapse
Affiliation(s)
- Auwal Abdullahi
- Department of Rehabilitation SciencesThe Hong Kong Polytechnic UniversityKowloonHong Kong Special Administrative RegionChina
| | - Thomson W. L. Wong
- Department of Rehabilitation SciencesThe Hong Kong Polytechnic UniversityKowloonHong Kong Special Administrative RegionChina
| | - Shamay S. M. Ng
- Department of Rehabilitation SciencesThe Hong Kong Polytechnic UniversityKowloonHong Kong Special Administrative RegionChina
| |
Collapse
|
8
|
Firman A, Warli SM, Sihombing B, Lelo A, Indharty RS, Nasution IPA, Muhar AM. Changes in thyroid function in prostate cancer patients receiving docetaxel chemotherapy at Haji Adam Malik Hospital, Indonesia. Rep Pract Oncol Radiother 2023; 28:522-528. [PMID: 37795233 PMCID: PMC10547421 DOI: 10.5603/rpor.a2023.0057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 07/24/2023] [Indexed: 10/06/2023] Open
Abstract
Background Prostate cancer treatment is determined based on several factors, namely tumor grading, staging, co-morbidity, patient preferences, life expectancy at diagnosis. Today, taxanes are commonly prescribed to treat several types of cancer and have been shown to have antitumor effects in many cancers. This research has never been done in prostate cancer patients but similar studies have been done before in breast cancer patients. Materials and methods The research design was observational analytic where this type of research was a prospective cohort where data was collected to record prostate cancer patients who received docetaxel chemotherapy which were then examined for thyroid function in cancer patients at the Adam Malik Hospital, Medan, Indonesia. Result In this study, data were collected regarding the thyroid function of the study sample in the form of free thyroxine (fT4) and thyroid-stimulating hormone (TSH) levels before chemotherapy with the docetaxel regimen. The mean of fT4 in all research subjects was 1.05 with a standard deviation of 0.26. The mean TSH in all study subjects was 1.52 with a standard deviation of 1.21. Thyroid function was examined after 3 cycles of docetaxel chemotherapy. The mean of fT4 in all research subjects was 0.91 with a standard deviation of 0.23. The mean TSH in all study subjects was 1.69 with a standard deviation of 1.09. Conclusion There are traces of the use of docetaxel chemotherapy in prostate cancer patients on decreased thyroid function at the Adam Malik Hospital in the form of decreased fT4 levels and increased TSH.
Collapse
Affiliation(s)
- Al Firman
- Department of Surgery, Faculty of Medicine, Universitas Sumatera Utara, Haji Adam Malik Hospital, Medan, Indonesia
| | - Syah Mirsya Warli
- Department of Urology, Faculty of Medicine, Universitas Sumatera Utara, Haji Adam Malik Hospital, Medan, Indonesia
- Division of Urology, Department of Surgery Faculty of Medicine, Universitas Sumatera Utara–Haji Adam Malik Hospital, Medan, Indonesia
| | - Bungaran Sihombing
- Division of Urology, Department of Surgery Faculty of Medicine, Universitas Sumatera Utara–Haji Adam Malik Hospital, Medan, Indonesia
| | - Aznan Lelo
- Department of Clinical Pharmacology, Faculty of Medicine, Universitas Sumatera Utara, Haji Adam Malik Hospital, Medan, Indonesia
| | - Rr. Suzy Indharty
- Department of Neurosurgery, Faculty of Medicine, Universitas Sumatera Utara, Haji Adam Malik Hospital, Medan, Indonesia
| | - Iqbal Pahlevi Adeputera Nasution
- Division of Pediatric Surgery, Department of Surgery, Faculty of Medicine, Universitas Sumatera Utara, Haji Adam Malik Hospital, Medan, Indonesia
| | - Adi Muradi Muhar
- Division of Digestive Surgery, Department of Surgery, Faculty of Medicine, Universitas Sumatera Utara, Haji Adam Malik Hospital, Medan, Indonesia
| |
Collapse
|
9
|
Pankowska KA, Będkowska GE, Chociej-Stypułkowska J, Rusak M, Dąbrowska M, Osada J. Crosstalk of Immune Cells and Platelets in an Ovarian Cancer Microenvironment and Their Prognostic Significance. Int J Mol Sci 2023; 24:ijms24119279. [PMID: 37298230 DOI: 10.3390/ijms24119279] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 05/17/2023] [Accepted: 05/23/2023] [Indexed: 06/12/2023] Open
Abstract
Ovarian cancer (OC) is one of the deadliest gynecological cancers, largely due to the fast development of metastasis and drug resistance. The immune system is a critical component of the OC tumor microenvironment (TME) and immune cells such as T cells, NK cells, and dendritic cells (DC) play a key role in anti-tumor immunity. However, OC tumor cells are well known for evading immune surveillance by modulating the immune response through various mechanisms. Recruiting immune-suppressive cells such as regulatory T cells (Treg cells), macrophages, or myeloid-derived suppressor cells (MDSC) inhibit the anti-tumor immune response and promote the development and progression of OC. Platelets are also involved in immune evasion by interaction with tumor cells or through the secretion of a variety of growth factors and cytokines to promote tumor growth and angiogenesis. In this review, we discuss the role and contribution of immune cells and platelets in TME. Furthermore, we discuss their potential prognostic significance to help in the early detection of OC and to predict disease outcome.
Collapse
Affiliation(s)
- Katarzyna Aneta Pankowska
- Department of Haematological Diagnostics, Medical University of Bialystok, Waszyngtona 15A Street, 15-269 Bialystok, Poland
| | - Grażyna Ewa Będkowska
- Department of Haematological Diagnostics, Medical University of Bialystok, Waszyngtona 15A Street, 15-269 Bialystok, Poland
| | - Joanna Chociej-Stypułkowska
- Department of Haematological Diagnostics, Medical University of Bialystok, Waszyngtona 15A Street, 15-269 Bialystok, Poland
| | - Małgorzata Rusak
- Department of Haematological Diagnostics, Medical University of Bialystok, Waszyngtona 15A Street, 15-269 Bialystok, Poland
| | - Milena Dąbrowska
- Department of Haematological Diagnostics, Medical University of Bialystok, Waszyngtona 15A Street, 15-269 Bialystok, Poland
| | - Joanna Osada
- Department of Haematological Diagnostics, Medical University of Bialystok, Waszyngtona 15A Street, 15-269 Bialystok, Poland
| |
Collapse
|
10
|
Swetha KL, Maravajjala KS, Li SD, Singh MS, Roy A. Breaking the niche: multidimensional nanotherapeutics for tumor microenvironment modulation. Drug Deliv Transl Res 2023; 13:105-134. [PMID: 35697894 DOI: 10.1007/s13346-022-01194-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/01/2022] [Indexed: 12/13/2022]
Abstract
Most of the current antitumor therapeutics were developed targeting the cancer cells only. Unfortunately, in the majority of tumors, this single-dimensional therapy is found to be ineffective. Advanced research has shown that cancer is a multicellular disorder. The tumor microenvironment (TME), which is made by a complex network of the bulk tumor cells and other supporting cells, plays a crucial role in tumor progression. Understanding the importance of the TME in tumor growth, different treatment modalities have been developed targeting these supporting cells. Recent clinical results suggest that simultaneously targeting multiple components of the tumor ecosystem with drug combinations can be highly effective. This type of "multidimensional" therapy has a high potential for cancer treatment. However, tumor-specific delivery of such multi-drug combinations remains a challenge. Nanomedicine could be utilized for the tumor-targeted delivery of such multidimensional therapeutics. In this review, we first give a brief overview of the major components of TME. We then highlight the latest developments in nanoparticle-based combination therapies, where one drug targets cancer cells and other drug targets tumor-supporting components in the TME for a synergistic effect. We include the latest preclinical and clinical studies and discuss innovative nanoparticle-mediated targeting strategies.
Collapse
Affiliation(s)
- K Laxmi Swetha
- Department of Pharmacy, Birla Institute of Technology & Science, Vidya Vihar, Pilani, Rajasthan, 333031, India
| | - Kavya Sree Maravajjala
- Department of Pharmacy, Birla Institute of Technology & Science, Vidya Vihar, Pilani, Rajasthan, 333031, India
| | - Shyh-Dar Li
- Faculty of Pharmaceutical Sciences, The University of British Columbia, 2405 Westbrook Mall, Vancouver, BC, Canada
| | - Manu Smriti Singh
- Department of Biotechnology, Bennett University, Greater Noida, Uttar Pradesh, 201310, India. .,Center of Excellence for Nanosensors and Nanomedicine, Bennett University, Greater Noida, Uttar Pradesh, 201310, India.
| | - Aniruddha Roy
- Department of Pharmacy, Birla Institute of Technology & Science, Vidya Vihar, Pilani, Rajasthan, 333031, India.
| |
Collapse
|
11
|
Cui X, Zhang F, Zhao Y, Li P, Wang T, Xu Z, Zhang J, Zhang W. A novel ligand-modified nanocomposite microparticles improved efficiency of quercetin and paclitaxel delivery in the non-small cell lung cancer. Drug Deliv 2022; 29:3123-3133. [PMID: 36151722 PMCID: PMC9848416 DOI: 10.1080/10717544.2022.2120567] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Chemotherapy is the first choice for the treatment of cancer but it is still limited by insufficient kill efficiency and drug resistance. These problems urgently need to be overcome in a way that minimizes damage to the body. In this study, we designed the nanocomposite microparticles (NMPs) modified by cetuximab (Cet) and loaded anti-tumor agents- quercetin (QUE) and paclitaxel (PTX)- for eliciting specific drugs homing and enhancing the killing efficiency of chemotherapy drugs (P/Q@CNMPs). Physicochemical characteristics results presented that P/Q@CNMPs have a suitable aerodynamic diameter and uniform morphology that could meet the requirements of particles deposition in the lung. And it also had the characteristics of sustained-release and pH-responsive which could release the agents in the right place and has a continuous effect. In vitro and in vivo analysis results presented that P/Q@CNMPs have the accuracy targeting ability and killing effect on non-small cell lung cancer (NSCLC) which express positive epidermal growth factor receptor (EGFR) on the membrane. Furthermore, this system also has low toxicity and good biocompatibility. These results demonstrated that P/Q@CNMPs could be a potential intelligent targeting strategy used for chemo-resistant NSCLC therapies.
Collapse
Affiliation(s)
- Xiaoming Cui
- College of Pharmacy, Weifang Medical University, Weifang, P.R. China
| | - Fang Zhang
- College of Pharmacy, Weifang Medical University, Weifang, P.R. China
| | - Yanyan Zhao
- College of Pharmacy, Weifang Medical University, Weifang, P.R. China
| | - Pan Li
- College of Pharmacy, Weifang Medical University, Weifang, P.R. China
| | - Ting Wang
- College of Pharmacy, Weifang Medical University, Weifang, P.R. China
| | - Zhilu Xu
- College of Pharmacy, Weifang Medical University, Weifang, P.R. China
| | - Jingjing Zhang
- College of Basic Medical, Qingdao Binhai University, Qingdao, P.R. China,CONTACT Jingjing Zhang College of Basic Medical, Qingdao Binhai University, Qingdao, 266000, P.R. China; Weifen Zhang College of Pharmacy, Weifang Medical University, 7166# Baotong West Street, Weifang261053, Shandong, P.R. China
| | - Weifen Zhang
- College of Pharmacy, Weifang Medical University, Weifang, P.R. China,Shandong Intelligent Materials and Regenerative Medicine Engineering Technology Research Center, Weifang, P.R. China
| |
Collapse
|
12
|
Goda MS, Elhady SS, Nafie MS, Bogari HA, Malatani RT, Hareeri RH, Badr JM, Donia MS. Phragmanthera austroarabica A.G.Mill. and J.A.Nyberg Triggers Apoptosis in MDA-MB-231 Cells In Vitro and In Vivo Assays: Simultaneous Determination of Selected Constituents. Metabolites 2022; 12:metabo12100921. [PMID: 36295823 PMCID: PMC9611470 DOI: 10.3390/metabo12100921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Revised: 09/26/2022] [Accepted: 09/27/2022] [Indexed: 11/16/2022] Open
Abstract
Phragmanthera austroarabica (Loranthaceae), a semi-parasitic plant, is well known for its high content of polyphenols that are responsible for its antioxidant and anti-inflammatory activities. Gallic acid, catechin, and methyl gallate are bioactive metabolites of common occurrence in the family of Loranthaceae. Herein, the concentrations of these bioactive metabolites were assessed using high-performance thin layer chromatography (HPTLC). Methyl gallate, catechin, and gallic acid were scanned at 280 nm. Their concentrations were assessed as 14.5, 6.5 and 43.6 mg/g of plant dry extract, respectively. Phragmanthera austroarabica extract as well as the three pure compounds were evaluated regarding the cytotoxic activity. The plant extract exhibited promising cytotoxic activity against MDA-MB-231 breast cells with the IC50 value of 19.8 μg/mL while the tested pure compounds displayed IC50 values in the range of 21.26–29.6 μg/mL. For apoptosis investigation, P. austroarabica induced apoptotic cell death by 111-fold change and necrosis by 9.31-fold change. It also activated the proapoptotic genes markers and inhibited the antiapoptotic gene, validating the apoptosis mechanism. Moreover, in vivo studies revealed a significant reduction in the breast tumor volume and weight in solid Ehrlich carcinoma (SEC) mice. The treatment of SEC mice with P. austroarabica extract improved both hematological and biochemical parameters with amelioration in the liver and kidney histopathology to near normal. Taken together, P. austroarabica extract exhibited promising anti-cancer activity through an apoptosis-induction.
Collapse
Affiliation(s)
- Marwa S. Goda
- Department of Pharmacognosy, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt
- Department of Pharmacognosy, Faculty of Pharmacy, Galala University, New Galala 43713, Egypt
| | - Sameh S. Elhady
- Department of Natural Products, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Correspondence: (S.S.E.); (J.M.B.); Tel.: +966-544512552 (S.S.E.); +20-1091332451 (J.M.B.)
| | - Mohamed S. Nafie
- Department of Chemistry, Faculty of Science, Suez Canal University, Ismailia 41522, Egypt
| | - Hanin A. Bogari
- Department of Pharmacy Practice, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Raina T. Malatani
- Department of Pharmacy Practice, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Rawan H. Hareeri
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Jihan M. Badr
- Department of Pharmacognosy, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt
- Correspondence: (S.S.E.); (J.M.B.); Tel.: +966-544512552 (S.S.E.); +20-1091332451 (J.M.B.)
| | - Marwa S. Donia
- Department of Pharmacognosy, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt
| |
Collapse
|
13
|
Wu KC, Liao KS, Yeh LR, Wang YK. Drug Repurposing: The Mechanisms and Signaling Pathways of Anti-Cancer Effects of Anesthetics. Biomedicines 2022; 10:biomedicines10071589. [PMID: 35884894 PMCID: PMC9312706 DOI: 10.3390/biomedicines10071589] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 06/20/2022] [Accepted: 07/01/2022] [Indexed: 12/14/2022] Open
Abstract
Cancer is one of the leading causes of death worldwide. There are only limited treatment strategies that can be applied to treat cancer, including surgical resection, chemotherapy, and radiotherapy, but these have only limited effectiveness. Developing a new drug for cancer therapy is protracted, costly, and inefficient. Recently, drug repurposing has become a rising research field to provide new meaning for an old drug. By searching a drug repurposing database ReDO_DB, a brief list of anesthetic/sedative drugs, such as haloperidol, ketamine, lidocaine, midazolam, propofol, and valproic acid, are shown to possess anti-cancer properties. Therefore, in the current review, we will provide a general overview of the anti-cancer mechanisms of these anesthetic/sedative drugs and explore the potential underlying signaling pathways and clinical application of these drugs applied individually or in combination with other anti-cancer agents.
Collapse
Affiliation(s)
- King-Chuen Wu
- Department of Anesthesiology, Chang Gung Memorial Hospital, Chiayi 61363, Taiwan;
- Department of Nursing, Chang Gung University of Science and Technology, Chiayi 61363, Taiwan
| | - Kai-Sheng Liao
- Department of Pathology, Ditmanson Medical Foundation, Chia-Yi Christian Hospital, Chiayi 60002, Taiwan;
| | - Li-Ren Yeh
- Department of Anesthesiology, E-Da Cancer Hospital, Kaohsiung 82445, Taiwan
- Department of Medical Imaging and Radiology, Shu-Zen College of Medicine and Management, Kaohsiung 82144, Taiwan
- Correspondence: (L.-R.Y.); (Y.-K.W.); Tel.: +886-7-6150-022 (L.-R.Y.); +886-6-2353-535 (ext. 5333) (Y.-K.W.)
| | - Yang-Kao Wang
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan
- Correspondence: (L.-R.Y.); (Y.-K.W.); Tel.: +886-7-6150-022 (L.-R.Y.); +886-6-2353-535 (ext. 5333) (Y.-K.W.)
| |
Collapse
|
14
|
Pincela Lins PM, Ribovski L, Corsi Antonio L, Altei WF, Sobreiro Selistre-de-Araújo H, Cancino-Bernardi J, Zucolotto V. Comparing extracellular vesicles and cell membranes as biocompatible coatings for gold nanorods: Implications for targeted theranostics. Eur J Pharm Biopharm 2022; 176:168-179. [PMID: 35643369 DOI: 10.1016/j.ejpb.2022.05.018] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 05/20/2022] [Accepted: 05/23/2022] [Indexed: 11/22/2022]
Abstract
Extracellular vesicles (EVs) and cell membrane nanoghosts are excellent coatings for nanomaterials, providing enhanced delivery in the target sites and evasion of the immune system. These cell-derived coatings allow the exploration of the delivery properties of the nanoparticles without stimulation of the immune system. Despite the advances reported on the use of EVs and cell-membrane coatings for nanomedicine applications, there are no standards to compare the benefits and main differences between these technologies. Here we investigated macrophage-derived EVs and cell membranes-coated gold nanorods and compared both systems in terms of target delivery in cancer and stromal cells. Our results reveal a higher tendency of EV-coated nanorods to interact with macrophages yet both EV and cell membrane-coated nanorods were internalized in the metastatic breast cancer cells. The main differences between these nanoparticles are related to the presence or absence of CD47 in the coating material, not usually addressed in EVs characterization. Our findings highlight important delivery differences exhibited by EVs- or cell membranes- coated nanorods which understanding may be important to the design and development of theragnostic nanomaterials using these coatings for target delivery.
Collapse
Affiliation(s)
- Paula Maria Pincela Lins
- Nanomedicine and Nanotoxicology Group, Physics Institute of São Carlos, University of São Paulo, São Carlos, SP, Brazil.
| | - Laís Ribovski
- Nanomedicine and Nanotoxicology Group, Physics Institute of São Carlos, University of São Paulo, São Carlos, SP, Brazil
| | - Luana Corsi Antonio
- Nanomedicine and Nanotoxicology Group, Physics Institute of São Carlos, University of São Paulo, São Carlos, SP, Brazil
| | - Wanessa Fernanda Altei
- Laboratory of Biochemistry and Molecular Biology, Department of Physiological Sciences, Federal University of São Carlos, SP, Brazil; Molecular Oncology Research Center, Barretos Cancer Hospital, SP, Brazil
| | | | - Juliana Cancino-Bernardi
- Nanomedicine and Nanotoxicology Group, Physics Institute of São Carlos, University of São Paulo, São Carlos, SP, Brazil
| | - Valtencir Zucolotto
- Nanomedicine and Nanotoxicology Group, Physics Institute of São Carlos, University of São Paulo, São Carlos, SP, Brazil; Institute of Advanced Studies, University of São Paulo, São Carlos, SP, Brazil
| |
Collapse
|
15
|
Maravajjala KS, Swetha KL, Roy A. pH-responsive nanoparticles for multidimensional combined chemo-immunotherapy of cancer. J Pharm Sci 2022; 111:2353-2368. [DOI: 10.1016/j.xphs.2022.05.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 05/11/2022] [Accepted: 05/11/2022] [Indexed: 10/18/2022]
|
16
|
Emerging Nanotherapeutic Approaches to Overcome Drug Resistance in Cancers with Update on Clinical Trials. Pharmaceutics 2022; 14:pharmaceutics14040866. [PMID: 35456698 PMCID: PMC9028322 DOI: 10.3390/pharmaceutics14040866] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 04/08/2022] [Accepted: 04/12/2022] [Indexed: 02/04/2023] Open
Abstract
A key issue with modern cancer treatments is the emergence of resistance to conventional chemotherapy and molecularly targeted medicines. Cancer nanotherapeutics were created in order to overcome the inherent limitations of traditional chemotherapeutics. Over the last few decades, cancer nanotherapeutics provided unparalleled opportunities to understand and overcome drug resistance through clinical assessment of rationally designed nanoparticulate delivery systems. In this context, various design strategies such as passive targeting, active targeting, nano-drug, and multimodal nano-drug combination therapy provided effective cancer treatment. Even though cancer nanotherapy has made great technological progress, tumor biology complexity and heterogeneity and a lack of comprehensive knowledge of nano-bio interactions remain important roadblocks to future clinical translation and commercialization. The current developments and advancements in cancer nanotherapeutics employing a wide variety of nanomaterial-based platforms to overcome cancer treatment resistance are discussed in this article. There is also a review of various nanotherapeutics-based approaches to cancer therapy, including targeting strategies for the tumor microenvironment and its components, advanced delivery systems for specific targeting of cancer stem cells (CSC), as well as exosomes for delivery strategies, and an update on clinical trials. Finally, challenges and the future perspective of the cancer nanotherapeutics to reverse cancer drug resistance are discussed.
Collapse
|
17
|
Rawding PA, Bu J, Wang J, Kim D, Drelich AJ, Kim Y, Hong S. Dendrimers for cancer immunotherapy: Avidity-based drug delivery vehicles for effective anti-tumor immune response. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2022; 14:e1752. [PMID: 34414690 PMCID: PMC9485970 DOI: 10.1002/wnan.1752] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 07/25/2021] [Accepted: 07/29/2021] [Indexed: 12/19/2022]
Abstract
Cancer immunotherapy, or the utilization of a patient's own immune system to treat cancer, has shifted the paradigm of cancer treatment. Despite meaningful responses being observed in multiple studies, currently available immunotherapy platforms have only proven effective to a small subset of patients. To address this, nanoparticles have been utilized as a novel carrier for immunotherapeutic drugs, achieving robust anti-tumor effects with increased adaptive and durable responses. Specifically, dendrimer nanoparticles have attracted a great deal of scientific interest due to their versatility in various therapeutic applications, resulting from their unique physicochemical properties and chemically well-defined architecture. This review offers a comprehensive overview of dendrimer-based immunotherapy technologies, including their formulations, biological functionalities, and therapeutic applications. Common formulations include: (1) modulators of cytokine secretion of immune cells (adjuvants); (2) facilitators of the recognition of tumorous antigens (vaccines); (3) stimulators of immune effectors to selectively attack cells expressing specific antigens (antibodies); and (4) inhibitors of immune-suppressive responses (immune checkpoint inhibitors). On-going works and prospects of dendrimer-based immunotherapies are also discussed. Overall, this review provides a critical overview on rapidly growing dendrimer-based immunotherapy technologies and serves as a guideline for researchers and clinicians who are interested in this field. This article is categorized under: Nanotechnology Approaches to Biology > Nanoscale Systems in Biology Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease Therapeutic Approaches and Drug Discovery > Emerging Technologies.
Collapse
Affiliation(s)
- Piper A Rawding
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA,Wisconsin Center for NanoBioSystems, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Jiyoon Bu
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA,Wisconsin Center for NanoBioSystems, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Jianxin Wang
- Wisconsin Center for NanoBioSystems, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - DaWon Kim
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA,Wisconsin Center for NanoBioSystems, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Adam J Drelich
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA,Wisconsin Center for NanoBioSystems, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Youngsoo Kim
- Wisconsin Center for NanoBioSystems, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Seungpyo Hong
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA,Wisconsin Center for NanoBioSystems, University of Wisconsin-Madison, Madison, WI 53705, USA,Yonsei Frontier Lab and Department of Pharmacy, Yonsei University, Seoul 03722, Republic of Korea
| |
Collapse
|
18
|
Li J, Hu ZE, We YJ, Liu YH, Wang N, Yu XQ. Multifunctional carbon quantum dots as a theranostic nanomedicine for fluorescence imaging-guided glutathione depletion to improve chemodynamic therapy. J Colloid Interface Sci 2022; 606:1219-1228. [PMID: 34492460 DOI: 10.1016/j.jcis.2021.08.114] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 08/15/2021] [Accepted: 08/16/2021] [Indexed: 12/18/2022]
Abstract
To minimize unwanted reactions with high concentrations of reduced glutathione (GSH) in the tumor microenvironment (TME) during chemodynamic therapy (CDT), a simple and effective strategy was developed to fabricate a TME stimuli-responsive theranostic nanomedicine (Fe-CD) for fluorescence imaging-guided GSH depletion and cancer therapy by combining fluorescent imaging carbon dots (CD) and Fe(III). Introducing Fe(III) into Fe-CD not only quenched the fluorescence of CD while reacting with and consuming intracellular GSH for fluorescence imaging of the depletion of GSH but also provided a source of metal ions to generate more abundant hydroxyl radicals (•OH) with hydrogen peroxide (H2O2) through the Fenton reaction to improve CDT. Fe-CD showed promising •OH generation under H2O2 to effectively degrade methylene blue in vitro and obviously activate the green fluorescence of the reactive oxygen species (ROS) probe in cells. Benefiting from the fluorescence enhancement in response to TME stimulation, Fe-CD greatly enhanced CDT cytotoxicity while monitoring successful GSH depletion by fluorescence imaging. Fe-CD has the potential to act as a theranostic nanomedicine for fluorescence imaging-guided GSH depletion to amplify CDT.
Collapse
Affiliation(s)
- Jun Li
- Key Laboratory of Green Chemistry and Technology (Ministry of Education), College of Chemistry, Sichuan University, Chengdu, 610064, China
| | - Zu-E Hu
- Key Laboratory of Green Chemistry and Technology (Ministry of Education), College of Chemistry, Sichuan University, Chengdu, 610064, China
| | - Yun-Jie We
- Key Laboratory of Green Chemistry and Technology (Ministry of Education), College of Chemistry, Sichuan University, Chengdu, 610064, China
| | - Yan-Hong Liu
- Key Laboratory of Green Chemistry and Technology (Ministry of Education), College of Chemistry, Sichuan University, Chengdu, 610064, China
| | - Na Wang
- Key Laboratory of Green Chemistry and Technology (Ministry of Education), College of Chemistry, Sichuan University, Chengdu, 610064, China.
| | - Xiao-Qi Yu
- Key Laboratory of Green Chemistry and Technology (Ministry of Education), College of Chemistry, Sichuan University, Chengdu, 610064, China.
| |
Collapse
|
19
|
Foglietta F, Serpe L, Canaparo R. The Effective Combination between 3D Cancer Models and Stimuli-Responsive Nanoscale Drug Delivery Systems. Cells 2021; 10:3295. [PMID: 34943803 PMCID: PMC8699241 DOI: 10.3390/cells10123295] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 11/17/2021] [Accepted: 11/22/2021] [Indexed: 12/12/2022] Open
Abstract
Stimuli-responsive drug-delivery systems (DDSs) have emerged as a potential tool for applications in healthcare, mainly in the treatment of cancer where versatile nanocarriers are co-triggered by endogenous and exogenous stimuli. Two-dimensional (2D) cell cultures are the most important in vitro model used to evaluate the anticancer activity of these stimuli-responsive DDSs due to their easy manipulation and versatility. However, some limitations suggest that these in vitro models poorly predict the outcome of in vivo studies. One of the main drawbacks of 2D cell cultures is their inadequate representation of the 3D environment's physiological complexity, which sees cells interact with each other and the extracellular matrix (ECM) according to their specific cellular organization. In this regard, 3D cancer models are a promising approach that can overcome the main shortcomings of 2D cancer cell cultures, as these in vitro models possess many peculiarities by which they mimic in vivo tumors, including physiologically relevant cell-cell and cell-ECM interactions. This is, in our opinion, even more relevant when a stimuli-responsive DDS is being investigated. In this review, we therefore report and discuss endogenous and exogenous stimuli-responsive DDSs whose effectiveness has been tested using 3D cancer cell cultures.
Collapse
Affiliation(s)
| | - Loredana Serpe
- Department of Drug Science and Technology, University of Torino, Via Pietro Giuria 13, 10125 Torino, Italy; (F.F.); (R.C.)
| | | |
Collapse
|
20
|
Debela DT, Muzazu SGY, Heraro KD, Ndalama MT, Mesele BW, Haile DC, Kitui SK, Manyazewal T. New approaches and procedures for cancer treatment: Current perspectives. SAGE Open Med 2021; 9:20503121211034366. [PMID: 34408877 PMCID: PMC8366192 DOI: 10.1177/20503121211034366] [Citation(s) in RCA: 596] [Impact Index Per Article: 149.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 07/05/2021] [Indexed: 01/11/2023] Open
Abstract
Cancer is a global health problem responsible for one in six deaths worldwide. Treating cancer has been a highly complex process. Conventional treatment approaches, such as surgery, chemotherapy, and radiotherapy, have been in use, while significant advances are being made in recent times, including stem cell therapy, targeted therapy, ablation therapy, nanoparticles, natural antioxidants, radionics, chemodynamic therapy, sonodynamic therapy, and ferroptosis-based therapy. Current methods in oncology focus on the development of safe and efficient cancer nanomedicines. Stem cell therapy has brought promising efficacy in regenerating and repairing diseased or damaged tissues by targeting both primary and metastatic cancer foci, and nanoparticles brought new diagnostic and therapeutic options. Targeted therapy possessed breakthrough potential inhibiting the growth and spread of specific cancer cells, causing less damage to healthy cells. Ablation therapy has emerged as a minimally invasive procedure that burns or freezes cancers without the need for open surgery. Natural antioxidants demonstrated potential tracking down free radicals and neutralizing their harmful effects thereby treating or preventing cancer. Several new technologies are currently under research in clinical trials, and some of them have already been approved. This review presented an update on recent advances and breakthroughs in cancer therapies.
Collapse
Affiliation(s)
- Dejene Tolossa Debela
- Center for Innovative Drug Development and Therapeutic Trials for Africa (CDT-Africa), College of Health Sciences, Addis Ababa University, Addis Ababa, Ethiopia
| | - Seke GY Muzazu
- Center for Innovative Drug Development and Therapeutic Trials for Africa (CDT-Africa), College of Health Sciences, Addis Ababa University, Addis Ababa, Ethiopia
- Enteric Diseases and Vaccines Research Unit, Centre for Infectious Disease Research in Zambia (CIDRZ), Lusaka, Zambia
| | - Kidist Digamo Heraro
- Center for Innovative Drug Development and Therapeutic Trials for Africa (CDT-Africa), College of Health Sciences, Addis Ababa University, Addis Ababa, Ethiopia
- Wachemo University, Hossana, Ethiopia
| | - Maureen Tayamika Ndalama
- Center for Innovative Drug Development and Therapeutic Trials for Africa (CDT-Africa), College of Health Sciences, Addis Ababa University, Addis Ababa, Ethiopia
| | - Betelhiem Woldemedhin Mesele
- Center for Innovative Drug Development and Therapeutic Trials for Africa (CDT-Africa), College of Health Sciences, Addis Ababa University, Addis Ababa, Ethiopia
- Kotebe Metropolitan University, Addis Ababa, Ethiopia
| | - Dagimawi Chilot Haile
- Center for Innovative Drug Development and Therapeutic Trials for Africa (CDT-Africa), College of Health Sciences, Addis Ababa University, Addis Ababa, Ethiopia
- University of Gondar, Gondar, Ethiopia
| | - Sophia Khalayi Kitui
- Center for Innovative Drug Development and Therapeutic Trials for Africa (CDT-Africa), College of Health Sciences, Addis Ababa University, Addis Ababa, Ethiopia
| | - Tsegahun Manyazewal
- Center for Innovative Drug Development and Therapeutic Trials for Africa (CDT-Africa), College of Health Sciences, Addis Ababa University, Addis Ababa, Ethiopia
| |
Collapse
|
21
|
Briolay T, Petithomme T, Fouet M, Nguyen-Pham N, Blanquart C, Boisgerault N. Delivery of cancer therapies by synthetic and bio-inspired nanovectors. Mol Cancer 2021; 20:55. [PMID: 33761944 PMCID: PMC7987750 DOI: 10.1186/s12943-021-01346-2] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 03/05/2021] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND As a complement to the clinical development of new anticancer molecules, innovations in therapeutic vectorization aim at solving issues related to tumor specificity and associated toxicities. Nanomedicine is a rapidly evolving field that offers various solutions to increase clinical efficacy and safety. MAIN: Here are presented the recent advances for different types of nanovectors of chemical and biological nature, to identify the best suited for translational research projects. These nanovectors include different types of chemically engineered nanoparticles that now come in many different flavors of 'smart' drug delivery systems. Alternatives with enhanced biocompatibility and a better adaptability to new types of therapeutic molecules are the cell-derived extracellular vesicles and micro-organism-derived oncolytic viruses, virus-like particles and bacterial minicells. In the first part of the review, we describe their main physical, chemical and biological properties and their potential for personalized modifications. The second part focuses on presenting the recent literature on the use of the different families of nanovectors to deliver anticancer molecules for chemotherapy, radiotherapy, nucleic acid-based therapy, modulation of the tumor microenvironment and immunotherapy. CONCLUSION This review will help the readers to better appreciate the complexity of available nanovectors and to identify the most fitting "type" for efficient and specific delivery of diverse anticancer therapies.
Collapse
Affiliation(s)
- Tina Briolay
- Université de Nantes, Inserm, CRCINA, F-44000, Nantes, France
| | | | - Morgane Fouet
- Université de Nantes, Inserm, CRCINA, F-44000, Nantes, France
| | | | | | | |
Collapse
|
22
|
Martinelli C. Smart Nanocarriers for Targeted Cancer Therapy. Anticancer Agents Med Chem 2021; 21:546-557. [PMID: 32560615 DOI: 10.2174/1871520620666200619181425] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 02/27/2020] [Accepted: 04/24/2020] [Indexed: 11/22/2022]
Abstract
Cancer is considered one of the most threatening diseases worldwide. Although many therapeutic approaches have been developed and optimized for ameliorating patient's conditions and life expectancy, however, it frequently remains an incurable pathology. Notably, conventional treatments may reveal inefficient in the presence of metastasis development, multidrug resistance and inability to achieve targeted drug delivery. In the last decades, nanomedicine has gained a prominent role, due to many properties ascribable to nanomaterials. It is worth mentioning their small size, their ability to be loaded with small drugs and bioactive molecules and the possibility to be functionalized for tumor targeting. Natural vehicles have been exploited, such as exosomes, and designed, such as liposomes. Biomimetic nanomaterials have been engineered, by modification with biological membrane coating. Several nanoparticles have already entered clinical trials and some liposomal formulations have been approved for therapeutic applications. In this review, natural and synthetic nanocarriers functionalized for actively targeting cancer cells will be described, focusing on their advantages with respect to conventional treatments. Recent innovations related to biomimetic nanoparticles camouflaged with membranes isolated from different types of cells will be reported, together with their promising applications. Finally, a short overview on the latest advances in carrier-free nanomaterials will be provided.
Collapse
Affiliation(s)
- Chiara Martinelli
- Istituto Italiano di Tecnologia, Smart Bio-Interfaces, Viale Rinaldo Piaggio 34, 56025 Pontedera, Pisa, Italy
| |
Collapse
|
23
|
Yang Y, Yang Y, Yang J, Zhao X, Wei X. Tumor Microenvironment in Ovarian Cancer: Function and Therapeutic Strategy. Front Cell Dev Biol 2020; 8:758. [PMID: 32850861 PMCID: PMC7431690 DOI: 10.3389/fcell.2020.00758] [Citation(s) in RCA: 122] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 07/20/2020] [Indexed: 02/05/2023] Open
Abstract
Ovarian cancer is one of the leading causes of death in patients with gynecological malignancy. Despite optimal cytoreductive surgery and platinum-based chemotherapy, ovarian cancer disseminates and relapses frequently, with poor prognosis. Hence, it is urgent to find new targeted therapies for ovarian cancer. Recently, the tumor microenvironment has been reported to play a vital role in the tumorigenesis of ovarian cancer, especially with discoveries from genome-, transcriptome- and proteome-wide studies; thus tumor microenvironment may present potential therapeutic target for ovarian cancer. Here, we review the interactions between the tumor microenvironment and ovarian cancer and various therapies targeting the tumor environment.
Collapse
Affiliation(s)
- Yanfei Yang
- Department of Gynecology and Obstetrics, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan University, Chengdu, China
| | - Yang Yang
- Department of Gynecology and Obstetrics, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan University, Chengdu, China
| | - Jing Yang
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Xia Zhao
- Department of Gynecology and Obstetrics, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan University, Chengdu, China
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
24
|
Bhatt H, Ghosh B, Biswas S. Cell-Penetrating Peptide and α-Tocopherol-Conjugated Poly(amidoamine) Dendrimers for Improved Delivery and Anticancer Activity of Loaded Paclitaxel. ACS APPLIED BIO MATERIALS 2020; 3:3157-3169. [DOI: 10.1021/acsabm.0c00179] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Himanshu Bhatt
- Department of Pharmacy, Birla Institute of Technology & Science-Pilani, Hyderabad Campus, Medchal, Hyderabad, Telangana 500078, India
| | - Balaram Ghosh
- Department of Pharmacy, Birla Institute of Technology & Science-Pilani, Hyderabad Campus, Medchal, Hyderabad, Telangana 500078, India
| | - Swati Biswas
- Department of Pharmacy, Birla Institute of Technology & Science-Pilani, Hyderabad Campus, Medchal, Hyderabad, Telangana 500078, India
| |
Collapse
|
25
|
Tumor microenvironment targeted nanotherapeutics for cancer therapy and diagnosis: A review. Acta Biomater 2020; 101:43-68. [PMID: 31518706 DOI: 10.1016/j.actbio.2019.09.009] [Citation(s) in RCA: 199] [Impact Index Per Article: 39.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2019] [Revised: 08/09/2019] [Accepted: 09/09/2019] [Indexed: 02/07/2023]
Abstract
Recent findings suggest that the cellular and extracellular materials surrounding the cancerous cells from an atypical tumor microenvironment (TM) play a pivotal role in the process of tumor initiation and progression. TM comprises an intricate system involving diverse cell types including endothelial cells, pericytes, smooth muscle cells, fibroblasts, various inflammatory cells, dendritic cells, and cancer stem cells (CSCs). The TM-forming cells dynamically interact with the cancerous cells through various signaling mechanisms and pathways. The existence of this dynamic cellular communication is responsible for creating an environment suitable for sustaining a reasonably high cellular proliferation. Presently, researchers are showing interest to use these TM conditions to mediate effective targeting measures for cancer therapy. The use of nanotherapeutics-based combination therapy; stimuli-responsive nanotherapeutics targeting acidic pH, hypoxic environment; and nanoparticle-induced hyperthermia are some of the approaches that are under intense investigation for cancer therapy. This review discusses TM and its role in cancer progression and crosstalk understanding, opportunities, and epigenetic modifications involved therein to materialize the capability of nanotherapeutics to target cancer by availing TM. STATEMENT OF SIGNIFICANCE: This article presents various recent reports, proof-of-concept studies, patents, and clinical trials on the concept of tumor microenvironment for mediating the cancer-specific delivery of nanotechnology-based systems bearing anticancer drug and diagnostics. We highlight the potential of tumor microenvironment; its role in disease progression, opportunities, challenges, and allied treatment strategies for effective cancer therapy by conceptual understanding of tumor microenvironment and epigenetic modifications involved. Specifically, nanoparticle-based approaches to target various processes related to tumor microenvironment (pH responsive, hypoxic environment responsive, targeting of specific cells involved in tumor microenvironment, etc.) are dealt in detail.
Collapse
|
26
|
Huai Y, Hossen MN, Wilhelm S, Bhattacharya R, Mukherjee P. Nanoparticle Interactions with the Tumor Microenvironment. Bioconjug Chem 2019; 30:2247-2263. [PMID: 31408324 PMCID: PMC6892461 DOI: 10.1021/acs.bioconjchem.9b00448] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Compared to normal tissues, the tumor microenvironment (TME) has a number of aberrant characteristics including hypoxia, acidosis, and vascular abnormalities. Many researchers have sought to exploit these anomalous features of the TME to develop anticancer therapies, and several nanoparticle-based cancer therapeutics have resulted. In this Review, we discuss the composition and pathophysiology of the TME, introduce nanoparticles (NPs) used in cancer therapy, and address the interaction between the TME and NPs. Finally, we outline both the potential problems that affect TME-based nanotherapy and potential strategies to overcome these challenges.
Collapse
Affiliation(s)
- Yanyan Huai
- peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, United States
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, United States
| | - Md Nazir Hossen
- peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, United States
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, United States
| | - Stefan Wilhelm
- peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, United States
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma 73072, United States
| | - Resham Bhattacharya
- peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, United States
- Department of Obstetrics and Gynecology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, United States
| | - Priyabrata Mukherjee
- peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, United States
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, United States
| |
Collapse
|
27
|
Gong BS, Wang R, Xu HX, Miao MY, Yao ZZ. Nanotherapy Targeting the Tumor Microenvironment. Curr Cancer Drug Targets 2019; 19:525-533. [DOI: 10.2174/1568009619666181220103714] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Revised: 11/11/2018] [Accepted: 11/13/2018] [Indexed: 01/24/2023]
Abstract
Cancer is characterized by high mortality and low curability. Recent studies have shown
that the mechanism of tumor resistance involves not only endogenous changes to tumor cells, but
also to the tumor microenvironment (TME), which provides the necessary conditions for the growth,
invasion, and metastasis of cancer cells, akin to Stephen Paget’s hypothesis of “seed and soil.”
Hence, the TME is a significant target for cancer therapy via nanoparticles, which can carry different
kinds of drugs targeting different types or stages of tumors. The key step of nanotherapy is the
achievement of accurate active or passive targeting to trigger drugs precisely at tumor cells, with less
toxicity and fewer side effects. With deepened understanding of the tumor microenvironment and
rapid development of the nanomaterial industry, the mechanisms of nanotherapy could be individualized
according to the specific TME characteristics, including low pH, cancer-associated fibroblasts
(CAFs), and increased expression of metalloproteinase. However, some abnormal features of the
TME limit drugs from reaching all tumor cells in lethal concentrations, and the characteristics of tumors
vary in numerous ways, resulting in great challenges for the clinical application of nanotherapy.
In this review, we discuss the essential role of the tumor microenvironment in the genesis and
development of tumors, as well as the measures required to improve the therapeutic effects of tumor
microenvironment-targeting nanoparticles and ways to reduce damage to normal tissue.
Collapse
Affiliation(s)
- Bo-Shen Gong
- Administration Office for Undergraduates, Second Military Medical University, Shanghai, 200433, China
| | - Rui Wang
- Department of Biochemistry and Molecular Biology, Second Military Medical University, Shanghai, 200433, China
| | - Hong-Xia Xu
- Department of Nutrition, Daping Hospital and Research Institute of Surgery, Third Military Medical University, Chongqing, 400042, China
| | - Ming-Yong Miao
- Department of Biochemistry and Molecular Biology, Second Military Medical University, Shanghai, 200433, China
| | - Zhen-Zhen Yao
- Department of Biochemistry and Molecular Biology, Second Military Medical University, Shanghai, 200433, China
| |
Collapse
|
28
|
Pasto A, Giordano F, Evangelopoulos M, Amadori A, Tasciotti E. Cell membrane protein functionalization of nanoparticles as a new tumor-targeting strategy. Clin Transl Med 2019; 8:8. [PMID: 30877412 PMCID: PMC6420595 DOI: 10.1186/s40169-019-0224-y] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Accepted: 03/08/2019] [Indexed: 02/06/2023] Open
Abstract
Nanoparticles have seen considerable popularity as effective tools for drug delivery. However, non-specific targeting continues to remain a challenge. Recently, biomimetic nanoparticles have emerged as an innovative solution that exploits biologically-derived components to improve therapeutic potential. Specifically, cell membrane proteins extracted from various cells (i.e., leukocytes, erythrocytes, platelets, mesenchymal stem cells, cancer) have shown considerable promise in bestowing nanoparticles with increased circulation and targeting efficacy. Traditional nanoparticles can be detected and removed by the immune system which significantly hinders their clinical success. Biomimicry has been proposed as a promising approach to overcome these limitations. In this review, we highlight the current trends in biomimetic nanoparticles and describe how they are being used to increase their chemotherapeutic effect in cancer treatment.
Collapse
Affiliation(s)
- Anna Pasto
- Veneto Institute of Oncology-IRCCS, Padua, Italy.,Center for Biomimetic Medicine, Houston Methodist Research Institute, 6670 Bertner Ave, Houston, TX, 77030, USA
| | - Federica Giordano
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy.,Center for Biomimetic Medicine, Houston Methodist Research Institute, 6670 Bertner Ave, Houston, TX, 77030, USA
| | - Michael Evangelopoulos
- Center for Biomimetic Medicine, Houston Methodist Research Institute, 6670 Bertner Ave, Houston, TX, 77030, USA
| | - Alberto Amadori
- Veneto Institute of Oncology-IRCCS, Padua, Italy.,Department of Surgery, Oncology and Gastroenterology, University of Padova, Padua, Italy
| | - Ennio Tasciotti
- Center for Biomimetic Medicine, Houston Methodist Research Institute, 6670 Bertner Ave, Houston, TX, 77030, USA. .,Houston Methodist Orthopedics and Sports Medicine, Houston Methodist Hospital, Houston, TX, USA.
| |
Collapse
|
29
|
Pasto A, Giordano F, Evangelopoulos M, Amadori A, Tasciotti E. Cell membrane protein functionalization of nanoparticles as a new tumor-targeting strategy. Clin Transl Med 2019. [PMID: 30877412 DOI: 10.1186/s40169019-0224-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/30/2023] Open
Abstract
Nanoparticles have seen considerable popularity as effective tools for drug delivery. However, non-specific targeting continues to remain a challenge. Recently, biomimetic nanoparticles have emerged as an innovative solution that exploits biologically-derived components to improve therapeutic potential. Specifically, cell membrane proteins extracted from various cells (i.e., leukocytes, erythrocytes, platelets, mesenchymal stem cells, cancer) have shown considerable promise in bestowing nanoparticles with increased circulation and targeting efficacy. Traditional nanoparticles can be detected and removed by the immune system which significantly hinders their clinical success. Biomimicry has been proposed as a promising approach to overcome these limitations. In this review, we highlight the current trends in biomimetic nanoparticles and describe how they are being used to increase their chemotherapeutic effect in cancer treatment.
Collapse
Affiliation(s)
- Anna Pasto
- Veneto Institute of Oncology-IRCCS, Padua, Italy
- Center for Biomimetic Medicine, Houston Methodist Research Institute, 6670 Bertner Ave, Houston, TX, 77030, USA
| | - Federica Giordano
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
- Center for Biomimetic Medicine, Houston Methodist Research Institute, 6670 Bertner Ave, Houston, TX, 77030, USA
| | - Michael Evangelopoulos
- Center for Biomimetic Medicine, Houston Methodist Research Institute, 6670 Bertner Ave, Houston, TX, 77030, USA
| | - Alberto Amadori
- Veneto Institute of Oncology-IRCCS, Padua, Italy
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padua, Italy
| | - Ennio Tasciotti
- Center for Biomimetic Medicine, Houston Methodist Research Institute, 6670 Bertner Ave, Houston, TX, 77030, USA.
- Houston Methodist Orthopedics and Sports Medicine, Houston Methodist Hospital, Houston, TX, USA.
| |
Collapse
|
30
|
Tumor heterogeneity and nanoparticle-mediated tumor targeting: the importance of delivery system personalization. Drug Deliv Transl Res 2018; 8:1508-1526. [PMID: 30128797 DOI: 10.1007/s13346-018-0578-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
After the discovery of the enhanced permeability and retention effect in 1986, it was envisioned that nanoparticle-mediated tumor-targeted delivery of chemotherapeutics would make a radical change in cancer therapy. However, after three decades of extensive research, only a few nanotherapeutics have been approved for clinical use. Although significant advantages of nanomedicines have been demonstrated in pre-clinical studies, clinical outcome was found to be variable. Advanced research has revealed that significant biochemical and structural variations exist between (and among) different tumors. These variations can considerably affect the tumor delivery and efficacy of nanomedicines. Tumor penetration is an important determining factor for positive therapeutic outcome and same nanomedicine can show diverse efficacy against different tumors depending on the extent of tumor accumulation and penetration. Recent research has started shading light on how the tumor variations can influence nanoparticle tumor delivery. These findings indicate that there is no "ideal" design of nanoparticles for exhibiting equally high efficacy against a broad spectrum of tumors. For achieving maximum benefit of the nanotherapeutics, it is necessary to analyze the tumor microenvironment for understanding the biological and structural characteristics of the tumor. Designing of the nanomedicine should be done according to the tumor characteristics. In this comprehensive review, we have first given a brief overview of the design characteristics of nanomedicine which impact their tumor delivery. Then we discussed about the variability in the tumor architecture and how it influences nanomedicine delivery. Finally, we have discussed the possibility of delivery system personalization based on the tumor characteristics.
Collapse
|
31
|
Autonomous self-navigating drug-delivery vehicles: from science fiction to reality. Ther Deliv 2017; 8:1063-1075. [DOI: 10.4155/tde-2017-0086] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Low efficacy of targeted nanomedicines in biological experiments enforced us to challenge the traditional concept of drug targeting and suggest a paradigm of ‘addressed self-navigating drug-delivery vehicles,’ in which affinity selection of targeting peptides and vasculature-directed in vivo phage screening is replaced by the migration selection, which explores ability of ‘promiscuous’ phages and their proteins to migrate through the tumor-surrounding cellular barriers, using a ‘hub and spoke’ delivery strategy, and penetrate into the tumor affecting the diverse tumor cell population. The ‘self-navigating’ drug-delivery paradigm can be used as a theoretical and technical platform in design of a novel generation of molecular medications and imaging probes for precise and personal medicine. [Formula: see text]
Collapse
|