1
|
Lee HJ, Choi HJ, Jeong YJ, Na YH, Hong JT, Han JM, Hoe HS, Lim KH. Developing theragnostics for Alzheimer's disease: Insights from cancer treatment. Int J Biol Macromol 2024; 269:131925. [PMID: 38685540 DOI: 10.1016/j.ijbiomac.2024.131925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Revised: 04/24/2024] [Accepted: 04/25/2024] [Indexed: 05/02/2024]
Abstract
The prevalence of Alzheimer's disease (AD) and its associated economic and societal burdens are on the rise, but there are no curative treatments for AD. Interestingly, this neurodegenerative disease shares several biological and pathophysiological features with cancer, including cell-cycle dysregulation, angiogenesis, mitochondrial dysfunction, protein misfolding, and DNA damage. However, the genetic factors contributing to the overlap in biological processes between cancer and AD have not been actively studied. In this review, we discuss the shared biological features of cancer and AD, the molecular targets of anticancer drugs, and therapeutic approaches. First, we outline the common biological features of cancer and AD. Second, we describe several anticancer drugs, their molecular targets, and their effects on AD pathology. Finally, we discuss how protein-protein interactions (PPIs), receptor inhibition, immunotherapy, and gene therapy can be exploited for the cure and management of both cancer and AD. Collectively, this review provides insights for the development of AD theragnostics based on cancer drugs and molecular targets.
Collapse
Affiliation(s)
- Hyun-Ju Lee
- Korea Brain Research Institute (KBRI), 61, Cheomdan-ro, Dong-gu, Daegu 41062, Republic of Korea
| | - Hee-Jeong Choi
- Korea Brain Research Institute (KBRI), 61, Cheomdan-ro, Dong-gu, Daegu 41062, Republic of Korea
| | - Yoo Joo Jeong
- Korea Brain Research Institute (KBRI), 61, Cheomdan-ro, Dong-gu, Daegu 41062, Republic of Korea; Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science & Technology (DGIST), 333, Techno jungang-daero, Hyeonpung-eup, Dalseong-gun, Daegu 42988, Republic of Korea
| | - Yoon-Hee Na
- College of Pharmacy, Chungbuk National University, Cheongju-si 28160, Republic of Korea
| | - Jin Tae Hong
- College of Pharmacy, Chungbuk National University, Cheongju-si 28160, Republic of Korea
| | - Ji Min Han
- College of Pharmacy, Chungbuk National University, Cheongju-si 28160, Republic of Korea.
| | - Hyang-Sook Hoe
- Korea Brain Research Institute (KBRI), 61, Cheomdan-ro, Dong-gu, Daegu 41062, Republic of Korea; Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science & Technology (DGIST), 333, Techno jungang-daero, Hyeonpung-eup, Dalseong-gun, Daegu 42988, Republic of Korea.
| | - Key-Hwan Lim
- College of Pharmacy, Chungbuk National University, Cheongju-si 28160, Republic of Korea.
| |
Collapse
|
2
|
Chou CC, Tseng CE, Lin YS, Wang M, Chen PL, Chang D, Shen CH, Fang CY. Inhibition of orthotopic castration-resistant prostate cancer growth and metastasis in mice by JC VLPs carrying a suicide gene driven by the PSA promoter. Cancer Gene Ther 2024; 31:250-258. [PMID: 38072969 PMCID: PMC10874888 DOI: 10.1038/s41417-023-00699-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Revised: 10/30/2023] [Accepted: 11/13/2023] [Indexed: 02/20/2024]
Abstract
Metastatic castration-resistant prostate cancer (mCRPC) is challenging to treat. Virus-like particles (VLPs), originating from JC polyomavirus (JCPyV) and carrying a suicide gene driven by the PSA promoter (PSAtk-VLPs), can inhibit tumor growth in animal models of human prostate cancer. However, the efficacy of suppression of orthotopic PCa growth and metastasis by PSAtk-VLPs remains undetermined. Here, we established an iRFP stable expression CRPC cell line suitable for deep-tissue observation using fluorescence molecular tomography (FMT). These cells were implanted into murine prostate tissue, and PSAtk-VLPs were systemically administered via the tail vein along with the prodrug ganciclovir (GCV), allowing for the real-time observation of orthotopic prostate tumor growth and CRPC tumor metastasis. Our findings demonstrated that systemic PSAtk-VLPs administration with GCV and subsequent FMT scanning facilitated real-time observation of the suppressed growth in mouse iRFP CRPC orthotopic tumors, which further revealed a notable metastasis rate reduction. Systemic PSAtk-VLPs and GCV administration effectively inhibited orthotopic prostate cancer growth and metastasis. These findings suggest the potential of JCPyV VLPs as a promising vector for mCRPC gene therapy. Conclusively, systemically administered JCPyV VLPs carrying a tissue-specific promoter, JCPyV VLPs can protect genes within the bloodstream to be specifically expressed in specific organs.
Collapse
Affiliation(s)
- Chih-Chieh Chou
- Institute of Molecular Biology, National Chung Cheng University, Chiayi, Taiwan
| | - Chih-En Tseng
- Department of Anatomic Pathology, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Chia-Yi, Taiwan
| | - Yu-Shih Lin
- Department of Pharmacy, Chiayi Chang Gung Memorial hospital, Chiayi Branch, Puzi, Taiwan
| | - Meilin Wang
- Department of Microbiology and Immunology, School of Medicine, Chung-Shan. Medical University and Clinical Laboratory, Chung-Shan Medical University Hospital, Taichung, Taiwan
| | - Pei-Lain Chen
- Department of Medical Laboratory Science and Biotechnology, Central Taiwan University of Science and Technology, Taichung, Taiwan
| | - Deching Chang
- Institute of Molecular Biology, National Chung Cheng University, Chiayi, Taiwan
| | - Cheng-Huang Shen
- Institute of Molecular Biology, National Chung Cheng University, Chiayi, Taiwan.
- Department of Urology, Ditmanson Medical Foundation Chiayi Christian Hospital, Chiayi, Taiwan.
| | - Chiung-Yao Fang
- Institute of Molecular Biology, National Chung Cheng University, Chiayi, Taiwan.
- Department of Medical Research, Ditmanson Medical Foundation Chiayi Christian Hospital, Chiayi, Taiwan.
| |
Collapse
|
3
|
Chen YL, Bao CJ, Duan JL, Xie Y, Lu WL. Overcoming biological barriers by virus-like drug particles for drug delivery. Adv Drug Deliv Rev 2023; 203:115134. [PMID: 37926218 DOI: 10.1016/j.addr.2023.115134] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Revised: 10/27/2023] [Accepted: 10/30/2023] [Indexed: 11/07/2023]
Abstract
Virus-like particles (VLPs) have natural structural antigens similar to those found in viruses, making them valuable in vaccine immunization. Furthermore, VLPs have demonstrated significant potential in drug delivery, and emerged as promising vectors for transporting chemical drug, genetic drug, peptide/protein, and even nanoparticle drug. With virus-like permeability and strong retention, they can effectively target specific organs, tissues or cells, facilitating efficient intracellular drug release. Further modifications allow VLPs to transfer across various physiological barriers, thus acting the purpose of efficient drug delivery and accurate therapy. This article provides an overview of VLPs, covering their structural classifications, deliverable drugs, potential physiological barriers in drug delivery, strategies for overcoming these barriers, and future prospects.
Collapse
Affiliation(s)
- Yu-Ling Chen
- State Key Laboratory of Natural and Biomimetic Drugs, Beijing Key Laboratory of Molecular Pharmaceutics and Drug Delivery Systems, and School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Chun-Jie Bao
- State Key Laboratory of Natural and Biomimetic Drugs, Beijing Key Laboratory of Molecular Pharmaceutics and Drug Delivery Systems, and School of Pharmaceutical Sciences, Peking University, Beijing 100191, China; School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China; Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Jia-Lun Duan
- State Key Laboratory of Natural and Biomimetic Drugs, Beijing Key Laboratory of Molecular Pharmaceutics and Drug Delivery Systems, and School of Pharmaceutical Sciences, Peking University, Beijing 100191, China; Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Ying Xie
- State Key Laboratory of Natural and Biomimetic Drugs, Beijing Key Laboratory of Molecular Pharmaceutics and Drug Delivery Systems, and School of Pharmaceutical Sciences, Peking University, Beijing 100191, China.
| | - Wan-Liang Lu
- State Key Laboratory of Natural and Biomimetic Drugs, Beijing Key Laboratory of Molecular Pharmaceutics and Drug Delivery Systems, and School of Pharmaceutical Sciences, Peking University, Beijing 100191, China.
| |
Collapse
|
4
|
Butic AB, Spencer SA, Shaheen SK, Lukacher AE. Polyomavirus Wakes Up and Chooses Neurovirulence. Viruses 2023; 15:2112. [PMID: 37896889 PMCID: PMC10612099 DOI: 10.3390/v15102112] [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: 09/29/2023] [Revised: 10/13/2023] [Accepted: 10/16/2023] [Indexed: 10/29/2023] Open
Abstract
JC polyomavirus (JCPyV) is a human-specific polyomavirus that establishes a silent lifelong infection in multiple peripheral organs, predominantly those of the urinary tract, of immunocompetent individuals. In immunocompromised settings, however, JCPyV can infiltrate the central nervous system (CNS), where it causes several encephalopathies of high morbidity and mortality. JCPyV-induced progressive multifocal leukoencephalopathy (PML), a devastating demyelinating brain disease, was an AIDS-defining illness before antiretroviral therapy that has "reemerged" as a complication of immunomodulating and chemotherapeutic agents. No effective anti-polyomavirus therapeutics are currently available. How depressed immune status sets the stage for JCPyV resurgence in the urinary tract, how the virus evades pre-existing antiviral antibodies to become viremic, and where/how it enters the CNS are incompletely understood. Addressing these questions requires a tractable animal model of JCPyV CNS infection. Although no animal model can replicate all aspects of any human disease, mouse polyomavirus (MuPyV) in mice and JCPyV in humans share key features of peripheral and CNS infection and antiviral immunity. In this review, we discuss the evidence suggesting how JCPyV migrates from the periphery to the CNS, innate and adaptive immune responses to polyomavirus infection, and how the MuPyV-mouse model provides insights into the pathogenesis of JCPyV CNS disease.
Collapse
Affiliation(s)
| | | | | | - Aron E. Lukacher
- Department of Microbiology and Immunology, Penn State College of Medicine, Hershey, PA 17033, USA; (A.B.B.); (S.A.S.); (S.K.S.)
| |
Collapse
|
5
|
Kumar A, Das SK, Emdad L, Fisher PB. Applications of tissue-specific and cancer-selective gene promoters for cancer diagnosis and therapy. Adv Cancer Res 2023; 160:253-315. [PMID: 37704290 DOI: 10.1016/bs.acr.2023.03.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/15/2023]
Abstract
Current treatment of solid tumors with standard of care chemotherapies, radiation therapy and/or immunotherapies are often limited by severe adverse toxic effects, resulting in a narrow therapeutic index. Cancer gene therapy represents a targeted approach that in principle could significantly reduce undesirable side effects in normal tissues while significantly inhibiting tumor growth and progression. To be effective, this strategy requires a clear understanding of the molecular biology of cancer development and evolution and developing biological vectors that can serve as vehicles to target cancer cells. The advent and fine tuning of omics technologies that permit the collective and spatial recognition of genes (genomics), mRNAs (transcriptomics), proteins (proteomics), metabolites (metabolomics), epiomics (epigenomics, epitranscriptomics, and epiproteomics), and their interactomics in defined complex biological samples provide a roadmap for identifying crucial targets of relevance to the cancer paradigm. Combining these strategies with identified genetic elements that control target gene expression uncovers significant opportunities for developing guided gene-based therapeutics for cancer. The purpose of this review is to overview the current state and potential limitations in developing gene promoter-directed targeted expression of key genes and highlights their potential applications in cancer gene therapy.
Collapse
Affiliation(s)
- Amit Kumar
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States
| | - Swadesh K Das
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Massey Comprehensive Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States
| | - Luni Emdad
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Massey Comprehensive Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States
| | - Paul B Fisher
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Massey Comprehensive Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States.
| |
Collapse
|
6
|
Dianat-Moghadam H, Sharifi M, Salehi R, Keshavarz M, Shahgolzari M, Amoozgar Z. Engaging stemness improves cancer immunotherapy. Cancer Lett 2023; 554:216007. [PMID: 36396102 DOI: 10.1016/j.canlet.2022.216007] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 11/06/2022] [Accepted: 11/09/2022] [Indexed: 11/16/2022]
Abstract
Intra-tumoral immune cells promote the stemness of cancer stem cells (CSCs) in the tumor microenvironment (TME). CSCs promote tumor progression, relapse, and resistance to immunotherapy. Cancer stemness induces the expression of neoantigens and neo-properties in CSCs, creating an opportunity for targeted immunotherapies. Isolation of stem-like T cells or retaining stemness in T clonotypes strategies produces exhaustion-resistance T cells with superior re-expansion capacity and long-lasting responses after adoptive cell therapies. Stem cells-derived NK cells may be the next generation of NK cell products for immunotherapy. Here, we have reviewed mechanisms by which stemness factors modulated the immunoediting of the TME and summarized the potentials of CSCs in the development of immunotherapy regimens, including CAR-T cells, CAR-NK cells, cancer vaccines, and monoclonal antibodies. We have discussed the natural or genetically engineered stem-like T cells and stem cell-derived NK cells with increased cytotoxicity to tumor cells. Finally, we have provided a perspective on approaches that may improve the therapeutic efficacy of these novel adoptive cell-based products in targeting immunosuppressive TME.
Collapse
Affiliation(s)
- Hassan Dianat-Moghadam
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran; Pediatric Inherited Diseases Research Center, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Mohammadreza Sharifi
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Rasoul Salehi
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran; Pediatric Inherited Diseases Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mohsen Keshavarz
- The Persian Gulf Tropical Medicine Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Mehdi Shahgolzari
- Dental Implants Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Zohreh Amoozgar
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
7
|
Johnson RP, Ratnacaram CK, Kumar L, Jose J. Combinatorial approaches of nanotherapeutics for inflammatory pathway targeted therapy of prostate cancer. Drug Resist Updat 2022; 64:100865. [PMID: 36099796 DOI: 10.1016/j.drup.2022.100865] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 08/27/2022] [Accepted: 08/30/2022] [Indexed: 12/24/2022]
Abstract
Prostate cancer (PC) is the most prevalent male urogenital cancer worldwide. PC patients presenting an advanced or metastatic cancer succumb to the disease, even after therapeutic interventions including radiotherapy, surgery, androgen deprivation therapy (ADT), and chemotherapy. One of the hallmarks of PC is evading immune surveillance and chronic inflammation, which is a major challenge towards designing effective therapeutic formulations against PC. Chronic inflammation in PC is often characterized by tumor microenvironment alterations, epithelial-mesenchymal transition and extracellular matrix modifications. The inflammatory events are modulated by reactive nitrogen and oxygen species, inflammatory cytokines and chemokines. Major signaling pathways in PC includes androgen receptor, PI3K and NF-κB pathways and targeting these inter-linked pathways poses a major therapeutic challenge. Notably, many conventional treatments are clinically unsuccessful, due to lack of targetability and poor bioavailability of the therapeutics, untoward toxicity and multidrug resistance. The past decade witnessed an advancement of nanotechnology as an excellent therapeutic paradigm for PC therapy. Modern nanovectorization strategies such as stimuli-responsive and active PC targeting carriers offer controlled release patterns and superior anti-cancer effects. The current review initially describes the classification, inflammatory triggers and major inflammatory pathways of PC, various PC treatment strategies and their limitations. Subsequently, recent advancement in combinatorial nanotherapeutic approaches, which target PC inflammatory pathways, and the mechanism of action are discussed. Besides, the current clinical status and prospects of PC homing nanovectorization, and major challenges to be addressed towards the advancement PC therapy are also addressed.
Collapse
Affiliation(s)
- Renjith P Johnson
- Polymer Nanobiomaterial Research Laboratory, Nanoscience and Microfluidics Division, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, Karnataka 575018, India
| | - Chandrahas Koumar Ratnacaram
- Cell Signaling and Cancer Biology Division, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, Karnataka 575018, India
| | - Lalit Kumar
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Udupi, Karnataka 576 104, India
| | - Jobin Jose
- NITTE Deemed-to-be University, NGSM Institute of Pharmaceutical Sciences, Department of Pharmaceutics, Mangalore 575018, India.
| |
Collapse
|
8
|
Chen Y, Wu FH, Wu PQ, Xing HY, Ma T. The Role of The Tumor Microbiome in Tumor Development and Its Treatment. Front Immunol 2022; 13:935846. [PMID: 35911695 PMCID: PMC9334697 DOI: 10.3389/fimmu.2022.935846] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 06/21/2022] [Indexed: 01/05/2023] Open
Abstract
Commensal bacteria and other microorganisms that reside in the human body are closely associated with the development and treatment of cancers. Recently, tumor microbiome (TM) has been identified in a variety of cancers such as pancreatic, lung, and breast cancers. TM has different compositions in different tumors and has different effects on tumors. TM plays an important role in the formation of the tumor microenvironment, regulation of local immunity, and modification of tumor cell biology, and directly affects the efficacy of drug treatment for tumors. TM is expected to be a biomarker for tumors, and engineered tumor-targeting bacteria and anti-cancer microbial agents (GEN-001) have an important role in the treatment of tumors. This paper reviews the relevant studies on TM in recent years and describes its distribution in different tumors, its correlation with clinical features, its effect on local immunity, and the research directions of TM in tumor treatment.
Collapse
Affiliation(s)
- Yan Chen
- Department of Hematology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Fa-Hong Wu
- Department of General Surgery, Hepatic-Biliary-Pancreatic Institute, Lanzhou University Second Hospital, Lanzhou, China
| | - Peng-Qiang Wu
- Department of Hematology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Hong-Yun Xing
- Department of Hematology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- *Correspondence: Hong-Yun Xing, ; Tao Ma,
| | - Tao Ma
- Department of Hematology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- *Correspondence: Hong-Yun Xing, ; Tao Ma,
| |
Collapse
|
9
|
Singh R, Kumar S. Cancer Targeting and Diagnosis: Recent Trends with Carbon Nanotubes. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 12:2283. [PMID: 35808119 PMCID: PMC9268713 DOI: 10.3390/nano12132283] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 06/30/2022] [Accepted: 07/01/2022] [Indexed: 02/06/2023]
Abstract
Cancer belongs to a category of disorders characterized by uncontrolled cell development with the potential to invade other bodily organs, resulting in an estimated 10 million deaths globally in 2020. With advancements in nanotechnology-based systems, biomedical applications of nanomaterials are attracting increasing interest as prospective vehicles for targeted cancer therapy and enhancing treatment results. In this context, carbon nanotubes (CNTs) have recently garnered a great deal of interest in the field of cancer diagnosis and treatment due to various factors such as biocompatibility, thermodynamic properties, and varied functionalization. In the present review, we will discuss recent advancements regarding CNT contributions to cancer diagnosis and therapy. Various sensing strategies like electrochemical, colorimetric, plasmonic, and immunosensing are discussed in detail. In the next section, therapy techniques like photothermal therapy, photodynamic therapy, drug targeting, gene therapy, and immunotherapy are also explained in-depth. The toxicological aspect of CNTs for biomedical application will also be discussed in order to ensure the safe real-life and clinical use of CNTs.
Collapse
Affiliation(s)
- Ragini Singh
- College of Agronomy, Liaocheng University, Liaocheng 252059, China;
| | - Santosh Kumar
- Shandong Key Laboratory of Optical Communication Science and Technology, School of Physics Science and Information Technology, Liaocheng University, Liaocheng 252059, China
| |
Collapse
|
10
|
Che B, Zhang W, Xu S, Yin J, He J, Huang T, Li W, Yu Y, Tang K. Prostate Microbiota and Prostate Cancer: A New Trend in Treatment. Front Oncol 2021; 11:805459. [PMID: 34956913 PMCID: PMC8702560 DOI: 10.3389/fonc.2021.805459] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Accepted: 11/22/2021] [Indexed: 01/01/2023] Open
Abstract
Although the incidence and mortality of prostate cancer have gradually begun to decline in the past few years, it is still one of the leading causes of death from malignant tumors in the world. The occurrence and development of prostate cancer are affected by race, family history, microenvironment, and other factors. In recent decades, more and more studies have confirmed that prostate microflora in the tumor microenvironment may play an important role in the occurrence, development, and prognosis of prostate cancer. Microorganisms or their metabolites may affect the occurrence and metastasis of cancer cells or regulate anti-cancer immune surveillance. In addition, the use of tumor microenvironment bacteria in interventional targeting therapy of tumors also shows a unique advantage. In this review, we introduce the pathway of microbiota into prostate cancer, focusing on the mechanism of microorganisms in tumorigenesis and development, as well as the prospect and significance of microorganisms as tumor biomarkers and tumor prevention and treatment.
Collapse
Affiliation(s)
- Bangwei Che
- Department of Urology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Wenjun Zhang
- Department of Urology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Shenghan Xu
- Department of Urology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Jingju Yin
- Department of Stomatology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Jun He
- Department of Urology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Tao Huang
- Department of Urology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Wei Li
- Department of Urology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Ying Yu
- Department of Urology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Kaifa Tang
- Department of Urology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
- Institute of Medical Science of Guizhou Medical University, Guiyang, China
| |
Collapse
|
11
|
Suppression of bone metastatic castration-resistant prostate cancer cell growth by a suicide gene delivered by JC polyomavirus-like particles. Gene Ther 2021:10.1038/s41434-021-00280-8. [PMID: 34285388 DOI: 10.1038/s41434-021-00280-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 07/01/2021] [Accepted: 07/09/2021] [Indexed: 11/08/2022]
Abstract
Prostate cancer is one of the most common cancers in men. The heterogeneity and mutations exhibited by prostate cancer cells often results in the progression to incurable metastatic castration-resistant prostate cancer (mCRPC). Our previous investigations demonstrated that the virus-like particles (VLPs) of JC polyomavirus (JCPyV) can deliver exogenous genes to prostate cancer cells for expression. JCPyV VLPs packaging pPSAtk (PSAtk-VLPs) possess the ability to transcriptionally target and selectively induce cytotoxicity in prostate cancer cells in vitro and in vivo, as pPSAtk can only express the thymidine kinase gene, a suicide gene, in androgen receptor-positive cells. To further investigate whether PSAtk-VLPs inhibit the growth of metastasized prostate cancer cells, we established an animal model of bone-metastatic prostate cancer to compare PSAtk-VLPs with leuprorelin acetate and enzalutamide, hormonal agents commonly used in clinical settings, and investigated the effectiveness of PSAtk-VLPs. In the present study, we observed that PSAtk-VLPs effectively inhibited the growth of prostate cancer cells that had metastasized to the bone in the metastatic animal model. In addition, PSAtk-VLPs showed a higher effectiveness than hormone therapy in this animal model study. These results suggest that PSAtk-VLPs may serve as a treatment option for mCRPC therapy in the future.
Collapse
|
12
|
Lai WH, Fang CY, Chou MC, Lin MC, Shen CH, Chao CN, Jou YC, Chang D, Wang M. Peptide-guided JC polyomavirus-like particles specifically target bladder cancer cells for gene therapy. Sci Rep 2021; 11:11889. [PMID: 34088940 PMCID: PMC8178405 DOI: 10.1038/s41598-021-91328-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 05/25/2021] [Indexed: 12/03/2022] Open
Abstract
The ultimate goal of gene delivery vectors is to establish specific and effective treatments for human diseases. We previously demonstrated that human JC polyomavirus (JCPyV) virus-like particles (VLPs) can package and deliver exogenous DNA into susceptible cells for gene expression. For tissue-specific targeting in this study, JCPyV VLPs were conjugated with a specific peptide for bladder cancer (SPB) that specifically binds to bladder cancer cells. The suicide gene thymidine kinase was packaged and delivered by SPB-conjugated VLPs (VLP-SPBs). Expression of the suicide gene was detected only in human bladder cancer cells and not in lung cancer or neuroblastoma cells susceptible to JCPyV VLP infection in vitro and in vivo, demonstrating the target specificity of VLP-SPBs. The gene transduction efficiency of VLP-SPBs was approximately 100 times greater than that of VLPs without the conjugated peptide. JCPyV VLPs can be specifically guided to target particular cell types when tagged with a ligand molecule that binds to a cell surface marker, thereby improving gene therapy.
Collapse
Affiliation(s)
- Wei-Hong Lai
- Department of Urology, Ditmanson Medical Foundation, Chiayi Christian Hospital, Chiayi, Taiwan
| | - Chiung-Yao Fang
- Department of Medical Research, Ditmanson Medical Foundation, Chiayi Christian Hospital, Chiayi, Taiwan
| | - Ming-Chieh Chou
- Institute of Molecular Biology, National Chung Cheng University, 168, University Rd., Min-Hsiung, Chiayi, 621, Taiwan
| | - Mien-Chun Lin
- Department of Urology, Ditmanson Medical Foundation, Chiayi Christian Hospital, Chiayi, Taiwan
| | - Cheng-Huang Shen
- Department of Urology, Ditmanson Medical Foundation, Chiayi Christian Hospital, Chiayi, Taiwan
| | - Chun-Nun Chao
- Department of Pediatrics, Ditmanson Medical Foundation, Chiayi Christian Hospital, Chiayi, Taiwan
| | - Yeong-Chin Jou
- Department of Urology, Ditmanson Medical Foundation, Chiayi Christian Hospital, Chiayi, Taiwan
| | - Deching Chang
- Institute of Molecular Biology, National Chung Cheng University, 168, University Rd., Min-Hsiung, Chiayi, 621, Taiwan.
| | - Meilin Wang
- Department of Microbiology and Immunology, School of Medicine, Chung-Shan Medical University and Clinical Laboratory, Chung-Shan Medical University Hospital, No. 110, Sec. 1, Jianguo N. Rd., Taichung City, 40201, Taiwan.
| |
Collapse
|
13
|
Xue J, Chen K, Hu H, Gopinath SCB. Progress in gene therapy treatments for prostate cancer. Biotechnol Appl Biochem 2021; 69:1166-1175. [PMID: 33988271 DOI: 10.1002/bab.2193] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Accepted: 05/12/2021] [Indexed: 01/17/2023]
Abstract
Prostate cancer is one of the predominant cancers affecting men and has been widely reported. In the past, various therapies and drugs have been proposed to treat prostate cancer. Among these treatments, gene therapy has been considered to be an optimal and widely applicable treatment. Furthermore, due to the increased specificity of gene sequence complementation, the targeted delivery of complementary gene sequences may represent a useful treatment in certain instances. Various gene therapies, including tumor-suppressor gene therapy, suicide gene therapy, immunomodulation gene therapy and anti-oncogene therapies, have been established to treat a wide range of diseases, such as cardiac disease, cystic fibrosis, HIV/AIDS, diabetes, hemophilia, and cancers. To this end, several gene therapy clinical trials at various phases are underway. This overview describes the developments and progress in gene therapy, with a special focus being placed on prostate cancer.
Collapse
Affiliation(s)
- Jingxin Xue
- Department of Urology, Affiliated Jinan Third Hospital of Jining Medical University, Jining Medical University, Jinan, Shandong, China
| | - Keming Chen
- Department of Urology, Affiliated Jinan Third Hospital of Jining Medical University, Jining Medical University, Jinan, Shandong, China
| | - Heyi Hu
- Department of Urology, Affiliated Jinan Third Hospital of Jining Medical University, Jining Medical University, Jinan, Shandong, China
| | - Subash C B Gopinath
- Institute of Nano Electronic Engineering, Universiti Malaysia Perlis (UniMAP), Kangar, Perlis, 01000, Malaysia.,Faculty of Chemical Engineering Technology, Universiti Malaysia Perlis (UniMAP), Arau, Perlis, 02600, Malaysia
| |
Collapse
|
14
|
Omabe K, Paris C, Lannes F, Taïeb D, Rocchi P. Nanovectorization of Prostate Cancer Treatment Strategies: A New Approach to Improved Outcomes. Pharmaceutics 2021; 13:591. [PMID: 33919150 PMCID: PMC8143094 DOI: 10.3390/pharmaceutics13050591] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 04/06/2021] [Accepted: 04/08/2021] [Indexed: 12/21/2022] Open
Abstract
Prostate cancer (PC) is the most frequent male cancer in the Western world. Progression to Castration Resistant Prostate Cancer (CRPC) is a known consequence of androgen withdrawal therapy, making CRPC an end-stage disease. Combination of cytotoxic drugs and hormonal therapy/or genotherapy is a recognized modality for the treatment of advanced PC. However, this strategy is limited by poor bio-accessibility of the chemotherapy to tumor sites, resulting in an increased rate of collateral toxicity and incidence of multidrug resistance (MDR). Nanovectorization of these strategies has evolved to an effective approach to efficacious therapeutic outcomes. It offers the possibility to consolidate their antitumor activity through enhanced specific and less toxic active or passive targeting mechanisms, as well as enabling diagnostic imaging through theranostics. While studies on nanomedicine are common in other cancer types, only a few have focused on prostate cancer. This review provides an in-depth knowledge of the principles of nanotherapeutics and nanotheranostics, and how the application of this rapidly evolving technology can clinically impact CRPC treatment. With particular reference to respective nanovectors, we draw clinical and preclinical evidence, demonstrating the potentials and prospects of homing nanovectorization into CRPC treatment strategies.
Collapse
Affiliation(s)
- Kenneth Omabe
- Centre de Recherche en Cancérologie de Marseille, CRCM, Inserm UMR1068, CNRS UMR7258, Aix-Marseille University U105, Institut Paoli-Calmettes, 13273 Marseille, France; (K.O.); (C.P.); (F.L.); (D.T.)
- Department of Biochemistry & Molecular Biology, Alex Ekwueme Federal University, Ndufu-Alike Ikwo, PMB 1010, Abakaliki 84001, Nigeria
| | - Clément Paris
- Centre de Recherche en Cancérologie de Marseille, CRCM, Inserm UMR1068, CNRS UMR7258, Aix-Marseille University U105, Institut Paoli-Calmettes, 13273 Marseille, France; (K.O.); (C.P.); (F.L.); (D.T.)
| | - François Lannes
- Centre de Recherche en Cancérologie de Marseille, CRCM, Inserm UMR1068, CNRS UMR7258, Aix-Marseille University U105, Institut Paoli-Calmettes, 13273 Marseille, France; (K.O.); (C.P.); (F.L.); (D.T.)
| | - David Taïeb
- Centre de Recherche en Cancérologie de Marseille, CRCM, Inserm UMR1068, CNRS UMR7258, Aix-Marseille University U105, Institut Paoli-Calmettes, 13273 Marseille, France; (K.O.); (C.P.); (F.L.); (D.T.)
- Biophysics and Nuclear Medicine, La Timone University Hospital, European Center for Research in Medical Imaging, Aix-Marseille University, 13005 Marseille, France
| | - Palma Rocchi
- Centre de Recherche en Cancérologie de Marseille, CRCM, Inserm UMR1068, CNRS UMR7258, Aix-Marseille University U105, Institut Paoli-Calmettes, 13273 Marseille, France; (K.O.); (C.P.); (F.L.); (D.T.)
| |
Collapse
|
15
|
Montaño-Samaniego M, Bravo-Estupiñan DM, Méndez-Guerrero O, Alarcón-Hernández E, Ibáñez-Hernández M. Strategies for Targeting Gene Therapy in Cancer Cells With Tumor-Specific Promoters. Front Oncol 2020; 10:605380. [PMID: 33381459 PMCID: PMC7768042 DOI: 10.3389/fonc.2020.605380] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Accepted: 10/30/2020] [Indexed: 12/16/2022] Open
Abstract
Cancer is the second cause of death worldwide, surpassed only by cardiovascular diseases, due to the lack of early diagnosis, and high relapse rate after conventional therapies. Chemotherapy inhibits the rapid growth of cancer cells, but it also affects normal cells with fast proliferation rate. Therefore, it is imperative to develop other safe and more effective treatment strategies, such as gene therapy, in order to significantly improve the survival rate and life expectancy of patients with cancer. The aim of gene therapy is to transfect a therapeutic gene into the host cells to express itself and cause a beneficial biological effect. However, the efficacy of the proposed strategies has been insufficient for delivering the full potential of gene therapy in the clinic. The type of delivery vehicle (viral or non viral) chosen depends on the desired specificity of the gene therapy. The first gene therapy trials were performed with therapeutic genes driven by viral promoters such as the CMV promoter, which induces non-specific toxicity in normal cells and tissues, in addition to cancer cells. The use of tumor-specific promoters over-expressed in the tumor, induces specific expression of therapeutic genes in a given tumor, increasing their localized activity. Several cancer- and/or tumor-specific promoters systems have been developed to target cancer cells. This review aims to provide up-to-date information concerning targeting gene therapy with cancer- and/or tumor-specific promoters including cancer suppressor genes, suicide genes, anti-tumor angiogenesis, gene silencing, and gene-editing technology, as well as the type of delivery vehicle employed. Gene therapy can be used to complement traditional therapies to provide more effective treatments.
Collapse
Affiliation(s)
- Mariela Montaño-Samaniego
- Laboratorio de Terapia Génica, Departamento de Bioquímica, Escuela Nacional de Ciencias Biológicas del Instituto Politécnico Nacional, Ciudad de México, México
| | - Diana M. Bravo-Estupiñan
- Laboratorio de Terapia Génica, Departamento de Bioquímica, Escuela Nacional de Ciencias Biológicas del Instituto Politécnico Nacional, Ciudad de México, México
| | - Oscar Méndez-Guerrero
- Laboratorio de Terapia Génica, Departamento de Bioquímica, Escuela Nacional de Ciencias Biológicas del Instituto Politécnico Nacional, Ciudad de México, México
| | - Ernesto Alarcón-Hernández
- Laboratorio de Genética Molecular, Departamento de Bioquímica, Escuela Nacional de Ciencias Biológicas del Instituto Politécnico Nacional, Ciudad de México, México
| | - Miguel Ibáñez-Hernández
- Laboratorio de Terapia Génica, Departamento de Bioquímica, Escuela Nacional de Ciencias Biológicas del Instituto Politécnico Nacional, Ciudad de México, México
| |
Collapse
|
16
|
Comas-Garcia M, Colunga-Saucedo M, Rosales-Mendoza S. The Role of Virus-Like Particles in Medical Biotechnology. Mol Pharm 2020; 17:4407-4420. [PMID: 33147978 DOI: 10.1021/acs.molpharmaceut.0c00828] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Virus-like particles (VLPs) are protein-based, nanoscale, self-assembling, cage architectures, which have relevant applications in biomedicine. They can be used for the development of vaccines, imaging approaches, drug and gene therapy delivery systems, and in vitro diagnostic methods. Today, three relevant viruses are targeted using VLP-based recombinant vaccines. VLP-based drug delivery, nanoreactors for therapy, and imaging systems are approaches under development with promising outcomes. Several VLP-based vaccines are under clinical evaluation. Herein, an updated view on the VLP-based biomedical applications is provided; advanced methods for the production, functionalization, and drug loading of VLPs are described, and perspectives for the field are identified.
Collapse
Affiliation(s)
- Mauricio Comas-Garcia
- Department of Sciences, Autonomous University of San Luis Potosi, San Luis Potosi 78295, México.,Genomic Medicine Section, Research Center for Health Sciences and Biomedicine, Autonomous University of San Luis Potosi, San Luis Potosi 78210, México.,High-Resolution Microscopy Section, Research Center for Health Sciences and Biomedicine, Autonomous University of San Luis Potosi, San Luis Potosi 78210, México
| | - Mayra Colunga-Saucedo
- Genomic Medicine Section, Research Center for Health Sciences and Biomedicine, Autonomous University of San Luis Potosi, San Luis Potosi 78210, México
| | - Sergio Rosales-Mendoza
- Departament of Chemical Sciences, Autonomous University of San Luis Potosi, San Luis Potosi 78210, México.,Biotechnology Section, Research Center for Health Sciences and Biomedicine, Autonomous University of San Luis Potosi, San Luis Potosi 78210, México
| |
Collapse
|
17
|
CircPDZD8 promotes gastric cancer progression by regulating CHD9 via sponging miR-197-5p. Aging (Albany NY) 2020; 12:19352-19364. [PMID: 33049714 PMCID: PMC7732272 DOI: 10.18632/aging.103805] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Accepted: 07/09/2020] [Indexed: 01/24/2023]
Abstract
CircRNAs have been shown to be associated with gastric cancer tumorigenesis. But little was known about the role of circPDZD8 in gastric cancer. CircPDZD8 was up-regulated in gastric cancer tissues and cells, Kaplan-Meier survival analysis indicated that gastric patients had a poor overall survival when circPDZD8 levels were high. CircPDZD8 knockdown could hinder proliferation and migration of gastric cancer cells. MiR-197-5p, which was down-regulated in gastric cancer, was shown to be a target of circPDZD8 and was inversely correlated with circPDZD8 expression. CHD9, as a target gene of miR-197-5p, was negatively regulated by miR-197-5p and positively correlated with circPDZD8 expression. Importantly, circPDZD8 could up-regulate CHD9 expression by sponging miR-197-5p, and modulate cell progression by regulation of the miR-197-5p/CHD9 axis in gastric cancer. CircPDZD8 knockdown repressed the progression of gastric cancer cells by sponging miR-197-5p and down-regulating CHD9.
Collapse
|
18
|
Fifty Years of JC Polyomavirus: A Brief Overview and Remaining Questions. Viruses 2020; 12:v12090969. [PMID: 32882975 PMCID: PMC7552028 DOI: 10.3390/v12090969] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 08/28/2020] [Accepted: 08/30/2020] [Indexed: 12/11/2022] Open
Abstract
In the fifty years since the discovery of JC polyomavirus (JCPyV), the body of research representing our collective knowledge on this virus has grown substantially. As the causative agent of progressive multifocal leukoencephalopathy (PML), an often fatal central nervous system disease, JCPyV remains enigmatic in its ability to live a dual lifestyle. In most individuals, JCPyV reproduces benignly in renal tissues, but in a subset of immunocompromised individuals, JCPyV undergoes rearrangement and begins lytic infection of the central nervous system, subsequently becoming highly debilitating-and in many cases, deadly. Understanding the mechanisms allowing this process to occur is vital to the development of new and more effective diagnosis and treatment options for those at risk of developing PML. Here, we discuss the current state of affairs with regards to JCPyV and PML; first summarizing the history of PML as a disease and then discussing current treatment options and the viral biology of JCPyV as we understand it. We highlight the foundational research published in recent years on PML and JCPyV and attempt to outline which next steps are most necessary to reduce the disease burden of PML in populations at risk.
Collapse
|
19
|
|
20
|
Jiménez-Chávez ÁDJ, Moreno-Fierros L, Bustos-Jaimes I. Therapy with multi-epitope virus-like particles of B19 parvovirus reduce tumor growth and lung metastasis in an aggressive breast cancer mouse model. Vaccine 2019; 37:7256-7268. [PMID: 31570181 DOI: 10.1016/j.vaccine.2019.09.068] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Revised: 08/26/2019] [Accepted: 09/20/2019] [Indexed: 12/30/2022]
Abstract
Triple-negative breast cancer is a major health problem that lacks molecular targets for therapy. Neoepitopes represent a viable option to induce antitumor immune responses, but they have limitations, such as low immunogenicity and tolerance induction. Parvovirus B19 virus-like particles may be used to deliver neoepitopes to prime cellular immunity. We designed and evaluated the therapeutic effect of VP2 B19-virus-like particles, with multi-neoepitopes, in a 4T1 breast cancer model. Balb/c mice received four therapeutic immunizations with multi-neoepitopes-virus-like, wild type-virus-like, vehicle, or virus-like plus Cry1Ac adjuvant particles, intraperitoneally and peritumorally. Tumor growth, lung macro-metastasis, and specific immune responses were evaluated. Therapeutic administration of multi-epitopes virus-like particles significantly delayed tumor growth and decreased the lung macro-metastasis number, in comparison to treatment with wild type-virus-like particles, which surprisingly also elicited antitumoral effects that were improved with the adjuvant. Only treatments with multi-epitope virus-like particles induced specific proliferative responses of CD8 and CD4 T lymphocytes and Granzyme-B production in lymphatic nodes local to the tumor. Treatment with recombinant multiple neoepitopes-virus-like particles induced specific cellular responses, inhibited tumor growth and macro-metastasis, thus B19-virus-like particles may function as an effective delivery system for neoepitopes for personalized immunotherapy.
Collapse
Affiliation(s)
- Ángel de Jesús Jiménez-Chávez
- Biomedicine Unit, Faculty of Higher Studies Iztacala, National Autonomous University of Mexico, Avenida de los Barrios 1, Los Reyes Iztacala, Tlalnepantla 54090, Estado de México, Mexico
| | - Leticia Moreno-Fierros
- Biomedicine Unit, Faculty of Higher Studies Iztacala, National Autonomous University of Mexico, Avenida de los Barrios 1, Los Reyes Iztacala, Tlalnepantla 54090, Estado de México, Mexico.
| | - Ismael Bustos-Jaimes
- Department of Biochemistry, Faculty of Medicine, National Autonomous University of Mexico (UNAM), Mexico City 04510, Mexico
| |
Collapse
|