1
|
Wylaź M, Kaczmarska A, Pajor D, Hryniewicki M, Gil D, Dulińska-Litewka J. Exploring the role of PI3K/AKT/mTOR inhibitors in hormone-related cancers: A focus on breast and prostate cancer. Biomed Pharmacother 2023; 168:115676. [PMID: 37832401 DOI: 10.1016/j.biopha.2023.115676] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 10/06/2023] [Accepted: 10/06/2023] [Indexed: 10/15/2023] Open
Abstract
Breast cancer (BC) and prostate cancer (PC) are at the top of the list when it comes to the most common types of cancers worldwide. The phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT)/mammalian target of rapamycin (mTOR) signaling pathway is important, in that it strongly influences the development and progression of these tumors. Previous studies have emphasized the key role of inhibitors of the PIK3/AKT/mTOR signaling pathway in the treatment of BC and PC, and it remains to be a crucial method of treatment. In this review, the inhibitors of these signaling pathways are compared, as well as their effectiveness in therapy and potential as therapeutic agents. The use of these inhibitors as polytherapy is evaluated, especially with the use of hormonal therapy, which has shown promising results.
Collapse
Affiliation(s)
- Mateusz Wylaź
- Student Scientific Group at Jagiellonian University Medical College, Faculty of Medicine, Medical Biochemistry, ul. Mikołaja Kopernika Street 7C, 31-034 Krakow, Poland
| | - Anna Kaczmarska
- Student Scientific Group at Jagiellonian University Medical College, Faculty of Medicine, Medical Biochemistry, ul. Mikołaja Kopernika Street 7C, 31-034 Krakow, Poland
| | - Dawid Pajor
- Student Scientific Group at Jagiellonian University Medical College, Faculty of Medicine, Medical Biochemistry, ul. Mikołaja Kopernika Street 7C, 31-034 Krakow, Poland
| | - Matthew Hryniewicki
- Student Scientific Group at Jagiellonian University Medical College, Faculty of Medicine, Medical Biochemistry, ul. Mikołaja Kopernika Street 7C, 31-034 Krakow, Poland
| | - Dorota Gil
- Chair of Medical Biochemistry, Jagiellonian University Medical College, ul. Mikołaja Kopernika Street 7C, 31-034 Krakow, Poland
| | - Joanna Dulińska-Litewka
- Chair of Medical Biochemistry, Jagiellonian University Medical College, ul. Mikołaja Kopernika Street 7C, 31-034 Krakow, Poland.
| |
Collapse
|
2
|
Zhou H, Chen B, Zhang L, Li C. Machine learning-based identification of lower grade glioma stemness subtypes discriminates patient prognosis and drug response. Comput Struct Biotechnol J 2023; 21:3827-3840. [PMID: 37560125 PMCID: PMC10407594 DOI: 10.1016/j.csbj.2023.07.029] [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: 03/06/2023] [Revised: 07/06/2023] [Accepted: 07/19/2023] [Indexed: 08/11/2023] Open
Abstract
Glioma stem cells (GSCs) remodel their tumor microenvironment to sustain a supportive niche. Identification and stratification of stemness related characteristics in patients with glioma might aid in the diagnosis and treatment of the disease. In this study, we calculated the mRNA stemness index in bulk and single-cell RNA-sequencing datasets using machine learning methods and investigated the correlation between stemness and clinicopathological characteristics. A glioma stemness-associated score (GSScore) was constructed using multivariate Cox regression analysis. We also generated a GSC cell line derived from a patient diagnosed with glioma and used glioma cell lines to validate the performance of the GSScore in predicting chemotherapeutic responses. Differentially expressed genes (DEGs) between GSCs with high and low GSScores were used to cluster lower-grade glioma (LGG) samples into three stemness subtypes. Differences in clinicopathological characteristics, including survival, copy number variations, mutations, tumor microenvironment, and immune and chemotherapeutic responses, among the three LGG stemness-associated subtypes were identified. Using machine learning methods, we further identified genes as subtype predictors and validated their performance using the CGGA datasets. In the current study, we identified a GSScore that correlated with LGG chemotherapeutic response. Through the score, we also identified a novel classification of the LGG subtype and associated subtype predictors, which might facilitate the development of precision therapy.
Collapse
Affiliation(s)
- Hongshu Zhou
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, PR China
- Hypothalamic-pituitary Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, PR China
| | - Bo Chen
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, PR China
- Hypothalamic-pituitary Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, PR China
- Department of Surgery, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong SAR, China
| | - Liyang Zhang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, PR China
- Hypothalamic-pituitary Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, PR China
- Clinical Diagnosis and Therapy Center for Glioma, Xiangya Hospital, Central South University, Changsha, Hunan, PR China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, PR China
| | - Chuntao Li
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, PR China
- Hypothalamic-pituitary Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, PR China
- Clinical Diagnosis and Therapy Center for Glioma, Xiangya Hospital, Central South University, Changsha, Hunan, PR China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, PR China
| |
Collapse
|
3
|
Schrottmaier WC, Kral-Pointner JB, Salzmann M, Mussbacher M, Schmuckenschlager A, Pirabe A, Brunnthaler L, Kuttke M, Maier B, Heber S, Datler H, Ekici Y, Niederreiter B, Heber U, Blomgren B, Gorki AD, Söderberg-Nauclér C, Payrastre B, Gratacap MP, Knapp S, Schabbauer G, Assinger A. Platelet p110β mediates platelet-leukocyte interaction and curtails bacterial dissemination in pneumococcal pneumonia. Cell Rep 2022; 41:111614. [PMID: 36351402 DOI: 10.1016/j.celrep.2022.111614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 07/15/2022] [Accepted: 10/14/2022] [Indexed: 11/09/2022] Open
Abstract
Phosphatidylinositol 3-kinase catalytic subunit p110β is involved in tumorigenesis and hemostasis. However, it remains unclear if p110β also regulates platelet-mediated immune responses, which could have important consequences for immune modulation during anti-cancer treatment with p110β inhibitors. Thus, we investigate how platelet p110β affects inflammation and infection. Using a mouse model of Streptococcus pneumoniae-induced pneumonia, we find that both platelet-specific p110β deficiency and pharmacologic inhibition of p110β with TGX-221 exacerbate disease pathogenesis by preventing platelet-monocyte and neutrophil interactions, diminishing their infiltration and enhancing bacterial dissemination. Platelet p110β mediates neutrophil phagocytosis of S. pneumoniae in vitro and curtails bacteremia in vivo. Genetic deficiency or inhibition of platelet p110β also impairs macrophage recruitment in an independent model of sterile peritonitis. Our results demonstrate that platelet p110β dysfunction exacerbates pulmonary infection by impeding leukocyte functions. Thereby, our findings provide important insights into the immunomodulatory potential of PI3K inhibitors in bacterial infection.
Collapse
Affiliation(s)
- Waltraud Cornelia Schrottmaier
- Department of Vascular Biology and Thrombosis Research, Centre for Physiology and Pharmacology, Medical University of Vienna, Schwarzspanierstraße 17, 1090 Vienna, Austria
| | - Julia Barbara Kral-Pointner
- Department of Vascular Biology and Thrombosis Research, Centre for Physiology and Pharmacology, Medical University of Vienna, Schwarzspanierstraße 17, 1090 Vienna, Austria
| | - Manuel Salzmann
- Department of Vascular Biology and Thrombosis Research, Centre for Physiology and Pharmacology, Medical University of Vienna, Schwarzspanierstraße 17, 1090 Vienna, Austria
| | - Marion Mussbacher
- Department of Vascular Biology and Thrombosis Research, Centre for Physiology and Pharmacology, Medical University of Vienna, Schwarzspanierstraße 17, 1090 Vienna, Austria; Department of Pharmacology and Toxicology, Institute of Pharmaceutical Sciences, University of Graz, 8010 Graz, Austria
| | - Anna Schmuckenschlager
- Department of Vascular Biology and Thrombosis Research, Centre for Physiology and Pharmacology, Medical University of Vienna, Schwarzspanierstraße 17, 1090 Vienna, Austria
| | - Anita Pirabe
- Department of Vascular Biology and Thrombosis Research, Centre for Physiology and Pharmacology, Medical University of Vienna, Schwarzspanierstraße 17, 1090 Vienna, Austria
| | - Laura Brunnthaler
- Department of Vascular Biology and Thrombosis Research, Centre for Physiology and Pharmacology, Medical University of Vienna, Schwarzspanierstraße 17, 1090 Vienna, Austria
| | - Mario Kuttke
- Department of Vascular Biology and Thrombosis Research, Centre for Physiology and Pharmacology, Medical University of Vienna, Schwarzspanierstraße 17, 1090 Vienna, Austria
| | - Barbara Maier
- Department of Medicine I, Research Division of Infection Biology, Medical University Vienna, 1090 Vienna, Austria
| | - Stefan Heber
- Institute of Physiology, Centre for Physiology and Pharmacology, Medical University of Vienna, 1090 Vienna, Austria
| | - Hannes Datler
- Department of Vascular Biology and Thrombosis Research, Centre for Physiology and Pharmacology, Medical University of Vienna, Schwarzspanierstraße 17, 1090 Vienna, Austria
| | - Yasemin Ekici
- Department of Vascular Biology and Thrombosis Research, Centre for Physiology and Pharmacology, Medical University of Vienna, Schwarzspanierstraße 17, 1090 Vienna, Austria
| | - Birgit Niederreiter
- Division of Rheumatology, Internal Medicine III, Medical University of Vienna, 1090 Vienna, Austria
| | - Ulrike Heber
- Department of Pathology and Comprehensive Cancer Centre, Medical University of Vienna, 1090 Vienna, Austria
| | - Bo Blomgren
- Department of Clinical Sciences, Danderyd Hospital, Department of Oncology-Pathology, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Anna-Dorothea Gorki
- Department of Medicine I, Research Division of Infection Biology, Medical University Vienna, 1090 Vienna, Austria
| | - Cecilia Söderberg-Nauclér
- Department of Medicine, Solna, Centre for Molecular Medicine, Microbial Pathogenesis Unit, Karolinska University Hospital, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Bernard Payrastre
- INSERM UMR1297, Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Université Toulouse III Paul Sabatier, 31024 Toulouse, France
| | - Marie-Pierre Gratacap
- INSERM UMR1297, Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Université Toulouse III Paul Sabatier, 31024 Toulouse, France
| | - Sylvia Knapp
- Department of Medicine I, Research Division of Infection Biology, Medical University Vienna, 1090 Vienna, Austria
| | - Gernot Schabbauer
- Department of Vascular Biology and Thrombosis Research, Centre for Physiology and Pharmacology, Medical University of Vienna, Schwarzspanierstraße 17, 1090 Vienna, Austria.
| | - Alice Assinger
- Department of Vascular Biology and Thrombosis Research, Centre for Physiology and Pharmacology, Medical University of Vienna, Schwarzspanierstraße 17, 1090 Vienna, Austria.
| |
Collapse
|
4
|
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: 0] [Impact Index Per Article: 0] [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
|
5
|
Cruz-Hernández CD, Rodríguez-Martínez G, Cortés-Ramírez SA, Morales-Pacheco M, Cruz-Burgos M, Losada-García A, Reyes-Grajeda JP, González-Ramírez I, González-Covarrubias V, Camacho-Arroyo I, Cerbón M, Rodríguez-Dorantes M. Aptamers as Theragnostic Tools in Prostate Cancer. Biomolecules 2022; 12:biom12081056. [PMID: 36008950 PMCID: PMC9406110 DOI: 10.3390/biom12081056] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 07/16/2022] [Accepted: 07/20/2022] [Indexed: 02/07/2023] Open
Abstract
Despite of the capacity that several drugs have for specific inhibition of the androgen receptor (AR), in most cases, PCa progresses to an androgen-independent stage. In this context, the development of new targeted therapies for prostate cancer (PCa) has remained as a challenge. To overcome this issue, new tools, based on nucleic acids technology, have been developed. Aptamers are small oligonucleotides with a three-dimensional structure capable of interacting with practically any desired target, even large targets such as mammalian cells or viruses. Recently, aptamers have been studied for treatment and detection of many diseases including cancer. In PCa, numerous works have reported their use in the development of new approaches in diagnostics and treatment strategies. Aptamers have been joined with drugs or other specific molecules such as silencing RNAs (aptamer–siRNA chimeras) to specifically reduce the expression of oncogenes in PCa cells. Even though these studies have shown good results in the early stages, more research is still needed to demonstrate the clinical value of aptamers in PCa. The aim of this review was to compile the existing scientific literature regarding the use of aptamers in PCa in both diagnosis and treatment studies. Since Prostate-Specific Membrane Antigen (PSMA) aptamers are the most studied type of aptamers in this field, special emphasis was given to these aptamers.
Collapse
Affiliation(s)
- Carlos David Cruz-Hernández
- Laboratorio de Oncogenómica, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City 14610, Mexico; (C.D.C.-H.); (G.R.-M.); (S.A.C.-R.); (M.M.-P.); (M.C.-B.); (A.L.-G.)
| | - Griselda Rodríguez-Martínez
- Laboratorio de Oncogenómica, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City 14610, Mexico; (C.D.C.-H.); (G.R.-M.); (S.A.C.-R.); (M.M.-P.); (M.C.-B.); (A.L.-G.)
| | - Sergio A. Cortés-Ramírez
- Laboratorio de Oncogenómica, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City 14610, Mexico; (C.D.C.-H.); (G.R.-M.); (S.A.C.-R.); (M.M.-P.); (M.C.-B.); (A.L.-G.)
| | - Miguel Morales-Pacheco
- Laboratorio de Oncogenómica, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City 14610, Mexico; (C.D.C.-H.); (G.R.-M.); (S.A.C.-R.); (M.M.-P.); (M.C.-B.); (A.L.-G.)
| | - Marian Cruz-Burgos
- Laboratorio de Oncogenómica, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City 14610, Mexico; (C.D.C.-H.); (G.R.-M.); (S.A.C.-R.); (M.M.-P.); (M.C.-B.); (A.L.-G.)
| | - Alberto Losada-García
- Laboratorio de Oncogenómica, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City 14610, Mexico; (C.D.C.-H.); (G.R.-M.); (S.A.C.-R.); (M.M.-P.); (M.C.-B.); (A.L.-G.)
| | - Juan Pablo Reyes-Grajeda
- Laboratorio de Estructura de Proteínas, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City 14610, Mexico;
| | - Imelda González-Ramírez
- Departamento de Atención a la Salud, Universidad Autónoma Metropolitana–Xochimilco, Mexico City 04960, Mexico;
| | | | - Ignacio Camacho-Arroyo
- Unidad de Investigación en Reproducción Humana, Instituto Nacional de Perinatología, Facultad de Química, Universidad Nacional Autónoma de México (UNAM), Mexico City 04510, Mexico; (I.C.-A.); (M.C.)
| | - Marco Cerbón
- Unidad de Investigación en Reproducción Humana, Instituto Nacional de Perinatología, Facultad de Química, Universidad Nacional Autónoma de México (UNAM), Mexico City 04510, Mexico; (I.C.-A.); (M.C.)
| | - Mauricio Rodríguez-Dorantes
- Laboratorio de Oncogenómica, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City 14610, Mexico; (C.D.C.-H.); (G.R.-M.); (S.A.C.-R.); (M.M.-P.); (M.C.-B.); (A.L.-G.)
- Correspondence:
| |
Collapse
|
6
|
Prakash P, Lee WH, Loo CY, Wong HSJ, Parumasivam T. Advances in Polyhydroxyalkanoate Nanocarriers for Effective Drug Delivery: An Overview and Challenges. NANOMATERIALS 2022; 12:nano12010175. [PMID: 35010124 PMCID: PMC8746483 DOI: 10.3390/nano12010175] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 12/06/2021] [Accepted: 12/07/2021] [Indexed: 01/10/2023]
Abstract
Polyhydroxyalkanoates (PHAs) are natural polymers produced under specific conditions by certain organisms, primarily bacteria, as a source of energy. These up-and-coming bioplastics are an undeniable asset in enhancing the effectiveness of drug delivery systems, which demand characteristics like non-immunogenicity, a sustained and controlled drug release, targeted delivery, as well as a high drug loading capacity. Given their biocompatibility, biodegradability, modifiability, and compatibility with hydrophobic drugs, PHAs often provide a superior alternative to free drug therapy or treatments using other polymeric nanocarriers. The many formulation methods of existing PHA nanocarriers, such as emulsion solvent evaporation, nanoprecipitation, dialysis, and in situ polymerization, are explained in this review. Due to their flexibility that allows for a vessel tailormade to its intended application, PHA nanocarriers have found their place in diverse therapy options like anticancer and anti-infective treatments, which are among the applications of PHA nanocarriers discussed in this article. Despite their many positive attributes, the advancement of PHA nanocarriers to clinical trials of drug delivery applications has been stunted due to the polymers’ natural hydrophobicity, controversial production materials, and high production costs, among others. These challenges are explored in this review, alongside their existing solutions and alternatives.
Collapse
Affiliation(s)
- Priyanka Prakash
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, Minden 11800, Penang, Malaysia;
| | - Wing-Hin Lee
- Faculty of Pharmacy and Health Sciences, Royal College of Medicine Perak, Universiti Kuala Lumpur (RCMP UniKL), Ipoh 30450, Perak, Malaysia; (W.-H.L.); (C.-Y.L.)
| | - Ching-Yee Loo
- Faculty of Pharmacy and Health Sciences, Royal College of Medicine Perak, Universiti Kuala Lumpur (RCMP UniKL), Ipoh 30450, Perak, Malaysia; (W.-H.L.); (C.-Y.L.)
| | - Hau Seung Jeremy Wong
- School of Biological Sciences, Universiti Sains Malaysia, Minden 11800, Penang, Malaysia;
| | - Thaigarajan Parumasivam
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, Minden 11800, Penang, Malaysia;
- Correspondence: ; Tel.: +60-4-6577888
| |
Collapse
|
7
|
The use of aptamers in prostate cancer: A systematic review of theranostic applications. Clin Biochem 2021; 93:9-25. [PMID: 33794195 DOI: 10.1016/j.clinbiochem.2021.03.014] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 03/22/2021] [Accepted: 03/24/2021] [Indexed: 02/07/2023]
Abstract
Since prostate cancer (PCa) relies on limited diagnosis and therapies, more effective alternatives are needed. Aptamers are versatile tools that may be applied for better clinical management of PCa patients. This review shows the trends on aptamer-based applications for PCa to understand their future development. We searched articles reporting aptamers applied in PCa on the Pubmed, Scopus and Web of Science databases over the last decade. Almost 80% of the articles used previously selected aptamers in novel approaches. However, cell-SELEX was the most applied technique for the selection of new aptamers allowing their binding to targets in their native configuration. ssDNA aptamers were 24% more common than RNA aptamers. The most studied PCa-specific aptamers were the DNA PSA-specific aptamer PSap4#5 and the PSMA-specific RNA aptamers A10 and A9, being PSA and PSMA the most reported targets. Thus, researchers still prefer the ease of use of DNA aptamers. Blood-based liquid biopsies represented 24% of all samples, being the most promising clinical samples. Especially noteworthy, electro-analytical methods accounted for more than 40% of the diagnostic techniques and treatment approaches with drug delivery systems or transcriptional modifiers were reported in 70% of the articles. Although all these articles showed clinically relevant aptamers for PCa and there are good prospects for their use, the development of all these strategies was in its early stages. Thus, the aptamers are not completely validated and we foresee that the completion of clinical studies will allow the implementation of these aptamer-based technologies in the clinical practice of PCa.
Collapse
|
8
|
Cohen L, Livney YD, Assaraf YG. Targeted nanomedicine modalities for prostate cancer treatment. Drug Resist Updat 2021; 56:100762. [PMID: 33857756 DOI: 10.1016/j.drup.2021.100762] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 03/16/2021] [Accepted: 03/18/2021] [Indexed: 12/17/2022]
Abstract
Prostate cancer (PC) is the second most common cause of death amongst men in the USA. Therapy of PC has been transformed in the past decade by introducing novel therapeutics, advanced functional imaging and diagnostic approaches, next generation sequencing, as well as improved application of existing therapies in localized PC. Treatment of PC at the different stages of the disease may include surgery, androgen deprivation therapy (ADT), chemotherapy and radiation therapy. However, although ADT has proven efficacious in PC treatment, its effectiveness may be temporary, as these tumors frequently develop molecular mechanisms of therapy resistance, which allow them to survive and proliferate even under conditions of testosterone deprivation, inhibition of androgen receptor signaling, or cytotoxic drug treatment. Importantly, ADT was found to induce key alterations which frequently result in the formation of metastatic tumors displaying a therapy refractory phenotype. Hence, to overcome these serious therapeutic impediments, novel PC cell-targeted therapeutic strategies are being developed. These include diverse platforms enabling specific enhanced antitumor drug uptake and increased intracellular accumulation. Studies have shown that these novel treatment modalities lead to enhanced antitumor activity and diminished systemic toxicity due to the use of selective targeting and decreased drug doses. The underlying mechanism of targeting and internalization is based upon the interaction between a selective ligand, conjugated to a drug-loaded nanoparticle or directly to an anti-cancer drug, and a specific plasma membrane biomarker, uniquely overexpressed on the surface of PC cells. Another targeted therapeutic approach is the delivery of unique anti-oncogenic signaling pathway-based therapeutic drugs, which are selectively cytotoxic to PC cells. The current paper reviews PC targeted modalities reported in the past 6 years, and discusses both the advantages and limitations of the various targeted treatment strategies.
Collapse
Affiliation(s)
- Lital Cohen
- The Laboratory of Biopolymers for Food and Health, Department of Biotechnology and Food Engineering, Technion - Israel Institute of Technology, Haifa, 3200003, Israel
| | - Yoav D Livney
- The Laboratory of Biopolymers for Food and Health, Department of Biotechnology and Food Engineering, Technion - Israel Institute of Technology, Haifa, 3200003, Israel.
| | - Yehuda G Assaraf
- The Fred Wyszkowski Cancer Research Laboratory, Department of Biology, Technion - Israel Institute of Technology, Haifa, 3200003, Israel.
| |
Collapse
|
9
|
Turnham DJ, Bullock N, Dass MS, Staffurth JN, Pearson HB. The PTEN Conundrum: How to Target PTEN-Deficient Prostate Cancer. Cells 2020; 9:E2342. [PMID: 33105713 PMCID: PMC7690430 DOI: 10.3390/cells9112342] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 10/17/2020] [Accepted: 10/20/2020] [Indexed: 12/17/2022] Open
Abstract
Loss of the tumor suppressor phosphatase and tensin homologue deleted on chromosome 10 (PTEN), which negatively regulates the PI3K-AKT-mTOR pathway, is strongly linked to advanced prostate cancer progression and poor clinical outcome. Accordingly, several therapeutic approaches are currently being explored to combat PTEN-deficient tumors. These include classical inhibition of the PI3K-AKT-mTOR signaling network, as well as new approaches that restore PTEN function, or target PTEN regulation of chromosome stability, DNA damage repair and the tumor microenvironment. While targeting PTEN-deficient prostate cancer remains a clinical challenge, new advances in the field of precision medicine indicate that PTEN loss provides a valuable biomarker to stratify prostate cancer patients for treatments, which may improve overall outcome. Here, we discuss the clinical implications of PTEN loss in the management of prostate cancer and review recent therapeutic advances in targeting PTEN-deficient prostate cancer. Deepening our understanding of how PTEN loss contributes to prostate cancer growth and therapeutic resistance will inform the design of future clinical studies and precision-medicine strategies that will ultimately improve patient care.
Collapse
Affiliation(s)
- Daniel J. Turnham
- The European Cancer Stem Cell Research Institute, School of Biosciences, Cardiff University, Hadyn Ellis Building, Cardiff CF24 4HQ, UK; (D.J.T.); (N.B.); (M.S.D.)
| | - Nicholas Bullock
- The European Cancer Stem Cell Research Institute, School of Biosciences, Cardiff University, Hadyn Ellis Building, Cardiff CF24 4HQ, UK; (D.J.T.); (N.B.); (M.S.D.)
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Heath Park, Cardiff CF14 4XN, UK;
| | - Manisha S. Dass
- The European Cancer Stem Cell Research Institute, School of Biosciences, Cardiff University, Hadyn Ellis Building, Cardiff CF24 4HQ, UK; (D.J.T.); (N.B.); (M.S.D.)
| | - John N. Staffurth
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Heath Park, Cardiff CF14 4XN, UK;
| | - Helen B. Pearson
- The European Cancer Stem Cell Research Institute, School of Biosciences, Cardiff University, Hadyn Ellis Building, Cardiff CF24 4HQ, UK; (D.J.T.); (N.B.); (M.S.D.)
| |
Collapse
|
10
|
Yari H, Gali H, Awasthi V. Nanoparticles for Targeting of Prostate Cancer. Curr Pharm Des 2020; 26:5393-5413. [PMID: 32693761 DOI: 10.2174/1381612826666200721001500] [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: 02/07/2020] [Accepted: 05/27/2020] [Indexed: 11/22/2022]
Abstract
Prostate cancer (PCa) is the leading cause of death by cancer in men. Because of the drastic decline in the survival rate of PCa patients with advanced/metastatic disease, early diagnosis of disease and therapy without toxic side effects is crucial. Chemotherapy is widely used to control the progression of PCa at the later stages; however, it is associated with off-target toxicities and severe adverse effects due to the lack of specificity. Delivery of therapeutic or diagnostic agents by using targeted nanoparticles is a promising strategy to enhance accuracy and sensitivity of diagnosis of PCa and to increase efficacy and specificity of therapeutic agents. Numerous efforts have been made in past decades to create nanoparticles with different architectural bases for specific delivery payloads to prostate tumors. Major PCa associated cell membrane protein markers identified as targets for such purposes include folate receptor, sigma receptors, transferrin receptor, gastrin-releasing peptide receptor, urokinase plasminogen activator receptor, and prostate specific membrane antigen. Among these markers, prostate specific membrane antigen has emerged as an extremely specific and sensitive targetable marker for designing targeted nanoparticle-based delivery systems for PCa. In this article, we review contemporary advances in design, specificity, and efficacy of nanoparticles functionalized against PCa. Whenever feasible, both diagnostic as well as therapeutic applications are discussed.
Collapse
Affiliation(s)
- Hooman Yari
- Department of Pharmaceutical Sciences, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States
| | - Hariprasad Gali
- Department of Pharmaceutical Sciences, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States
| | - Vibhudutta Awasthi
- Department of Pharmaceutical Sciences, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States
| |
Collapse
|
11
|
Enhancement of FAK alleviates ventilator-induced alveolar epithelial cell injury. Sci Rep 2020; 10:419. [PMID: 31942012 PMCID: PMC6962166 DOI: 10.1038/s41598-019-57350-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 12/16/2019] [Indexed: 01/19/2023] Open
Abstract
Mechanical ventilation induces lung injury by damaging alveolar epithelial cells (AECs), but the pathogenesis remains unknown. Focal adhesion kinase (FAK) is a cytoplasmic protein tyrosine kinase that is involved in cell growth and intracellular signal transduction pathways. This study explored the potential role of FAK in AECs during lung injury induced by mechanical ventilation. High-volume mechanical ventilation (HMV) was used to create a mouse lung injury model, which was validated by analysis of lung weight, bronchoalveolar lavage fluid and histological investigation. The expression of FAK and Akt in AECs were evaluated. In addition, recombinant FAK was administered to mice via the tail vein, and then the extent of lung injury was assessed. Mouse AECs were cultured in vitro, and FAK expression in cells under stretch was investigated. The effects of FAK on cell proliferation, migration and apoptosis were investigated. The results showed that HMV decreased FAK expression in AECs of mice, while FAK supplementation attenuated lung injury, reduced protein levels/cell counts in the bronchoalveolar lavage fluid and decreased histological lung injury and oedema. The protective effect of FAK promoted AEC proliferation and migration and prevented cells from undergoing apoptosis, which restored the integrity of the alveoli through Akt pathway. Therefore, the decrease in FAK expression by HMV is essential for injury to epithelial cells and the disruption of alveolar integrity. FAK supplementation can reduce AEC injury associated with HMV.
Collapse
|
12
|
Li C, He C, Xu Y, Xu H, Tang Y, Chavan H, Duan S, Artigues A, Laird Forrest M, Krishnamurthy P, Han S, Holzbeierlein JM, Li B. Alternol eliminates excessive ATP production by disturbing Krebs cycle in prostate cancer. Prostate 2019; 79:628-639. [PMID: 30663084 PMCID: PMC6644699 DOI: 10.1002/pros.23767] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 12/28/2018] [Indexed: 12/11/2022]
Abstract
BACKGROUND Alternol is a natural compound isolated from fermentation products of a mutant fungus. Our previous studies demonstrated that Alternol specifically kills cancer cells but spares benign cells. METHODS To investigate the mechanism underlying alternol-induced cancer cell-specific killing effect, we took a comprehensive strategy to identify Alternol's protein targets in prostate cancer cells, including PC-3, C4-2, and 22RV1, plus benign BPH1 cell lines. Major experimental techniques included biotin-streptavidin pulldown assay coupled with mass-spectrometry, in vitro enzyme activity assay for Krebs cycle enzymes and gas chromatography-mass spectrometry (GC-MS) for metabolomic analysis. RESULTS Among 14 verified protein targets, four were Krebs cycle enzymes, fumarate hydratase (FH), malate dehydrogenase-2 (MDH2), dihydrolipoamide acetyltransferase (DLAT) in pyruvate dehydrogenase complex (PDHC) and dihydrolipoamide S-succinyltransferase (DLST) in a-ketoglutarate dehydrogenase complex (KGDHC). Functional assays revealed that PDHC and KGDHC activities at the basal level were significantly higher in prostate cancer cells compared to benign prostate BPH1 cells, while alternol treatment reduced their activities in cancer cells close to the levels in BPH1 cells. Although FH and MDH2 activities were comparable among prostate cancer and benign cell lines at the basal level, Alternol treatment largely increased their activities in cancer cells. Metabolomic analysis revealed that Alternol treatment remarkably reduced the levels of malic acid, fumaric acid, and isocitric acid and mitochondrial respiration in prostate cancer cells. Alternol also drastically reduced mitochondrial respiration and ATP production in PC-3 cells in vitro or in xenograft tissues but not in BPH1 cells or host liver tissues. CONCLUSIONS Alternol interacts with multiple Krebs cycle enzymes, resulting in reduced mitochondrial respiration and ATP production in prostate cancer cells and xenograft tissues, providing a novel therapeutic strategy for prostate cancer treatment.
Collapse
Affiliation(s)
- Changlin Li
- Institute of Precision Medicine, Jining Medical University, Jining, China
- Department of Urology, The University of Kansas Medical Center, Kansas City, Kansas
| | - Chenchen He
- Department of Urology, The University of Kansas Medical Center, Kansas City, Kansas
- Department of Radiation Oncology, The First Affiliated Hospital, Xi’An Jiaotong University School of Medicine, Xi’An, China
| | - Ying Xu
- Department of Urology, The University of Kansas Medical Center, Kansas City, Kansas
| | - Haixia Xu
- Department of Urology, The University of Kansas Medical Center, Kansas City, Kansas
| | - Yuzhe Tang
- Department of Urology, The University of Kansas Medical Center, Kansas City, Kansas
| | - Hemantkumar Chavan
- Department of Pharmacology, Toxicology & Therapeutics, The University of Kansas Medical Center, Kansas City, Kansas
| | - Shaofeng Duan
- Department of Pharmaceutical Chemistry, The University of Kansas, Lawrence, Kansas
| | - Antonio Artigues
- Department of Biochemistry & Molecular Biology, The University of Kansas Medical Center, Kansas City, Kansas
| | - Marcus Laird Forrest
- Department of Pharmaceutical Chemistry, The University of Kansas, Lawrence, Kansas
| | - Partha Krishnamurthy
- Department of Pharmacology, Toxicology & Therapeutics, The University of Kansas Medical Center, Kansas City, Kansas
| | - Suxia Han
- Department of Radiation Oncology, The First Affiliated Hospital, Xi’An Jiaotong University School of Medicine, Xi’An, China
| | | | - Benyi Li
- Department of Urology, The University of Kansas Medical Center, Kansas City, Kansas
| |
Collapse
|
13
|
He L, Liu J, Li S, Feng X, Wang C, Zhuang X, Ding J, Chen X. Polymer Nanoplatforms at Work in Prostate Cancer Therapy. ADVANCED THERAPEUTICS 2019; 2. [DOI: 10.1002/adtp.201800122] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Indexed: 12/13/2022]
Abstract
AbstractProstate cancer (PCa) is the most common male urogenital malignancy worldwide. Surgery, endocrine therapy, radiotherapy, and chemotherapy are the main clinical management options for PCa. However, these three therapies each have limitations. For example, surgery is not suitable for the advanced PCa patients with extensive metastases, and radiotherapy causes serious side effects. Primary endocrine therapy promotes the progression of hormone‐sensitive PCa into the castration‐resistant prostate cancer. Therefore, considering these drawbacks, chemotherapy has become an effective and extensive treatment for PCa. Among the modern therapeutic strategies against advanced PCa, polymer‐nanocarrier‐incorporated formulations have gradually emerged due to their well‐controlled release profiles and improved tumor targeting abilities. The drug delivery systems based on polymer nanoplatforms passively target tumors via the enhanced permeability and retention effect. Simultaneously, stimuli‐responsive polymer nanoplatforms unload cargoes in response to certain stimuli in the tumor area. Furthermore, the active targeting ligand‐conjugated polymer nanoformulations against PCa‐specific markers have also achieved great success in PCa therapies. Herein, the advanced polymer nanoplatforms for PCa therapy are reviewed, while the future development of polymer nanoplatforms for PCa therapy is also predicted.
Collapse
Affiliation(s)
- Liang He
- Key Laboratory of Polymer Ecomaterials Changchun Institute of Applied Chemistry Chinese Academy of Sciences Changchun 130022 P. R. China
- Department of Urology The First Hospital of Jilin University Changchun 130021 P. R. China
| | - Jianhua Liu
- Key Laboratory of Polymer Ecomaterials Changchun Institute of Applied Chemistry Chinese Academy of Sciences Changchun 130022 P. R. China
- Department of Urology The First Hospital of Jilin University Changchun 130021 P. R. China
| | - Shengxian Li
- Key Laboratory of Polymer Ecomaterials Changchun Institute of Applied Chemistry Chinese Academy of Sciences Changchun 130022 P. R. China
- Department of Urology The First Hospital of Jilin University Changchun 130021 P. R. China
| | - Xiangru Feng
- Key Laboratory of Polymer Ecomaterials Changchun Institute of Applied Chemistry Chinese Academy of Sciences Changchun 130022 P. R. China
| | - Chunxi Wang
- Department of Urology The First Hospital of Jilin University Changchun 130021 P. R. China
| | - Xiuli Zhuang
- Key Laboratory of Polymer Ecomaterials Changchun Institute of Applied Chemistry Chinese Academy of Sciences Changchun 130022 P. R. China
| | - Jianxun Ding
- Key Laboratory of Polymer Ecomaterials Changchun Institute of Applied Chemistry Chinese Academy of Sciences Changchun 130022 P. R. China
| | - Xuesi Chen
- Key Laboratory of Polymer Ecomaterials Changchun Institute of Applied Chemistry Chinese Academy of Sciences Changchun 130022 P. R. China
| |
Collapse
|
14
|
Yang X, Yang JA, Liu BH, Liao JM, Yuan FE, Tan YQ, Chen QX. TGX-221 inhibits proliferation and induces apoptosis in human glioblastoma cells. Oncol Rep 2017; 38:2836-2842. [PMID: 29048665 PMCID: PMC5780035 DOI: 10.3892/or.2017.5991] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Accepted: 09/04/2017] [Indexed: 01/08/2023] Open
Abstract
Glioblastoma is the most common type of primary brain tumor in adults, with high mortality and morbidity rates. More effective therapeutic strategies are imperative. Previous studies have shown that the known p110-β-selective inhibitor TGX-221 blocks the activation of PKB/Akt in PTEN-deficient cells. We treated U87 and U251 glioblastoma cells with TGX-221 to determine the effect of TGX-221. We performed a Cell Counting Kit-8 (CCK-8) test, EDU staining and cell cycle distribution analysis and found that TGX-221 inhibited glioblastoma cell proliferation. Next, the effect of TGX-221 on cell apoptosis was investigated using flow cytometry. These results demonstrated that TGX-221 induced apoptosis in glioblastoma cells. Moreover, migration and invasion assays revealed that TGX-221 inhibited human glioblastoma cell migration and invasion. Collectively, our study revealed that TGX-221 could inhibit proliferation and induce apoptosis in glioblastoma cells.
Collapse
Affiliation(s)
- Xue Yang
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuchang, Wuhan, Hubei 430060, P.R. China
| | - Ji-An Yang
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuchang, Wuhan, Hubei 430060, P.R. China
| | - Bao-Hui Liu
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuchang, Wuhan, Hubei 430060, P.R. China
| | - Jian-Ming Liao
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuchang, Wuhan, Hubei 430060, P.R. China
| | - Fan-En Yuan
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuchang, Wuhan, Hubei 430060, P.R. China
| | - Yin-Qiu Tan
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuchang, Wuhan, Hubei 430060, P.R. China
| | - Qian-Xue Chen
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuchang, Wuhan, Hubei 430060, P.R. China
| |
Collapse
|
15
|
He C, Duan S, Dong L, Wang Y, Hu Q, Liu C, Forrest ML, Holzbeierlein JM, Han S, Li B. Characterization of a novel p110β-specific inhibitor BL140 that overcomes MDV3100-resistance in castration-resistant prostate cancer cells. Prostate 2017; 77:1187-1198. [PMID: 28631436 PMCID: PMC5527967 DOI: 10.1002/pros.23377] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 05/23/2017] [Indexed: 01/24/2023]
Abstract
BACKGROUND Our previous studies demonstrated that the class IA PI3K/p110β is critical in castration-resistant progression of prostate cancer (CRPC) and that targeting prostate cancer with nanomicelle-loaded p110β-specific inhibitor TGX221 blocked xenograft tumor growth in nude mice, confirming the feasibility of p110β-targeted therapy for CRPCs. To improve TGX221's aqueous solubility, in this study, we characterized four recently synthesized TGX221 analogs. METHODS TGX221 analog efficacy were examined in multiple prostate cancer cell lines with the SRB cell growth assay, Western blot assay for AKT phosphorylation and cell cycle protein levels. Target engagement with PI3K isoforms was evaluated with cellular thermal shift assay. PI3K activity was determined with the Kinase-Glo Plus luminescent kinase assay. Cell cycle distribution was evaluated with flow cytometry after propidium iodide staining. RESULTS As expected, replacing either one of two major functional groups in TGX221 by more hydrophilic groups dramatically improved the aqueous solubility (about 40-fold) compared to TGX221. In the CETSA assay, all the analogs dramatically shifted the melting curve of p110β protein while none of them largely affected the melting curves of p110α, p110γ, or Akt proteins, indicating target-specific engagement of these analogs with p110β protein. However, functional evaluation showed that only one of the analogs BL140 ubiquitously inhibited AKT phosphorylation in all CRPC cell lines tested with diverse genetic abnormalities including AR, PTEN, and p53 status. BL140 was superior than GSK2636771 (IC50 5.74 vs 20.49 nM), the only p110β-selective inhibitor currently in clinical trials, as revealed in an in vitro Kinase-Glo assay. Furthermore, BL140 exhibited a stronger inhibitory effect than GSK2636771 on multiple CRPC cell lines including a MDV3100-resistant C4-2B cell subline, indicating BL140 elimination of MDV3100 resistance. Mechanistic studies revealed that BL140 blocked G1 phase cell cycle entry by reducing cyclin D1 but increasing p27kip1 protein levels. CONCLUSION These studies suggested that BL140 is a promising p110β-specific inhibitor with multiple superb properties than GSK2636771 worthy for further clinical development.
Collapse
Affiliation(s)
- Chenchen He
- Department of Medical Oncology, The First Affiliated Hospital, Xi’An Jiaotong University School of Medicine, Xi’An 710061, China
- Department of Urology, The University of Kansas Medical Center, Kansas City, KS 66160
| | - Shaofeng Duan
- Pharmaceutical College, Henan University, Kaifeng 475004, China
| | - Liang Dong
- Department of Urology, The University of Kansas Medical Center, Kansas City, KS 66160
| | - Yifen Wang
- Department of Urology, The University of Kansas Medical Center, Kansas City, KS 66160
| | - Qingting Hu
- Department of Urology, The University of Kansas Medical Center, Kansas City, KS 66160
| | - Chunjing Liu
- Department of Pharmaceutical Chemistry, The University of Kansas School of Pharmacy, Lawrence, KS 66045
| | - M. Laird Forrest
- Department of Pharmaceutical Chemistry, The University of Kansas School of Pharmacy, Lawrence, KS 66045
| | | | - Suxia Han
- Department of Medical Oncology, The First Affiliated Hospital, Xi’An Jiaotong University School of Medicine, Xi’An 710061, China
| | - Benyi Li
- Department of Medical Oncology, The First Affiliated Hospital, Xi’An Jiaotong University School of Medicine, Xi’An 710061, China
- Department of Urology, The University of Kansas Medical Center, Kansas City, KS 66160
- Pharmaceutical College, Henan University, Kaifeng 475004, China
| |
Collapse
|
16
|
Cheng H, Chua V, Liao C, Purwin TJ, Terai M, Kageyama K, Davies MA, Sato T, Aplin AE. Co-targeting HGF/cMET Signaling with MEK Inhibitors in Metastatic Uveal Melanoma. Mol Cancer Ther 2017; 16:516-528. [PMID: 28138035 PMCID: PMC5337170 DOI: 10.1158/1535-7163.mct-16-0552] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Revised: 11/22/2016] [Accepted: 11/24/2016] [Indexed: 12/28/2022]
Abstract
Patients with metastatic uveal melanoma usually die within 1 year of diagnosis, emphasizing an urgent need to develop new treatment strategies. The liver is the most common site of metastasis. Mitogen-activated protein kinase kinase (MEK) inhibitors improve survival in V600 BRAF-mutated cutaneous melanoma patients but have limited efficacy in patients with uveal melanoma. Our previous work showed that hepatocyte growth factor (HGF) signaling elicits resistance to MEK inhibitors in metastatic uveal melanoma. In this study, we demonstrate that expression of two BH3-only family proteins, Bim-EL and Bmf, contributes to HGF-mediated resistance to MEK inhibitors. Targeting HGF/cMET signaling with LY2875358, a neutralizing and internalizing anti-cMET bivalent antibody, and LY2801653, a dual cMET/RON inhibitor, overcomes resistance to trametinib provided by exogenous HGF and by conditioned medium from primary hepatic stellate cells. We further determined that activation of PI3Kα/γ/δ isoforms mediates the resistance to MEK inhibitors by HGF. Combination of LY2801653 with trametinib decreases AKT phosphorylation and promotes proapoptotic PARP cleavage in metastatic uveal melanoma explants. Together, our data support the notion that selectively blocking cMET signaling or PI3K isoforms in metastatic uveal melanoma may break the intrinsic resistance to MEK inhibitors provided by factors from stromal cells in the liver. Mol Cancer Ther; 16(3); 516-28. ©2017 AACR.
Collapse
Affiliation(s)
- Hanyin Cheng
- Department of Cancer Biology and Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Vivian Chua
- Department of Cancer Biology and Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Connie Liao
- Department of Cancer Biology and Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Timothy J Purwin
- Department of Cancer Biology and Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Mizue Terai
- Department of Medical Oncology, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Ken Kageyama
- Department of Medical Oncology, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Michael A Davies
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Takami Sato
- Department of Medical Oncology, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Andrew E Aplin
- Department of Cancer Biology and Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania.
- Department of Dermatology and Cutaneous Biology, Thomas Jefferson University, Philadelphia, Pennsylvania
| |
Collapse
|
17
|
Pang J, Yang YW, Huang Y, Yang J, Zhang H, Chen R, Dong L, Huang Y, Wang D, Liu J, Li B. P110β Inhibition Reduces Histone H3K4 Di-Methylation in Prostate Cancer. Prostate 2017; 77:299-308. [PMID: 27800642 DOI: 10.1002/pros.23271] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Accepted: 10/05/2016] [Indexed: 12/27/2022]
Abstract
INTRODUCTION AND AIMS Epigenetic alteration plays a major role in the development and progression of human cancers, including prostate cancer. Histones are the key factors in modulating gene accessibility to transcription factors and post-translational modification of the histone N-terminal tail including methylation is associated with either transcriptional activation (H3K4me2) or repression (H3K9me3). Furthermore, phosphoinositide 3-kinase (PI3 K) signaling and the androgen receptor (AR) are the key determinants in prostate cancer development and progression. We recently showed that prostate-targeted nano-micelles loaded with PI3 K/p110beta specific inhibitor TGX221 blocked prostate cancer growth in vitro and in vivo. Our objective of this study was to determine the role of PI3 K signaling in histone methylation in prostate cancer, with emphasis on histone H3K4 methylation. METHODS PI3 K non-specific inhibitor LY294002 and p110beta-specific inhibitor TGX221 were used to block PI3 K/p110beta signaling. The global levels of H3K4 and H3K9 methylation in prostate cancer cells and tissue specimens were evaluated by Western blot assay and immunohistochemical staining. A synthetic androgen R1881 was used to stimulate AR activity in prostate cancer cells. A castration-resistant prostate cancer (CRPC) specific human tissue microarray (TMA) was used to assess the global levels of H3K4me2 methylation by immunostaining approach. RESULTS Our data revealed that H3K4me2 levels were significantly elevated after androgen stimulation. With RNA silencing and pharmacology approaches, we further defined that inhibition of PI3 K/p110beta activity through gene-specific knocking down and small chemical inhibitor TGX221 abolished androgen-stimulated H3K4me2 methylation. Consistently, prostate cancer-targeted delivery of TGX221 in vivo dramatically reduced the global levels of H3K4me2 as assessed by immunohistochemical staining on tissue section of mouse xenografts from CRPC cell lines 22RV1 and C4-2. Finally, immunostaining data revealed a strong H3K4me2 immunosignal in CRPC tissues compared to primary tumors and benign prostate tissues. CONCLUSIONS Taken together, our results suggest that PI3 K/p110beta-dependent signaling is involved in androgen-stimulated H3K4me2 methylation in prostate cancer, which might be used as a novel biomarker for disease prognosis and targeted therapy. Prostate 77:299-308, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Jun Pang
- Department of Urology, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Yue-Wu Yang
- Department of Traditional Chinese Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Yiling Huang
- Department of Urology, The University of Kansas Medical Center, Kansas City, Kansas
- Department of Pathology, China Three Gorges University School of Medicine, Yichang, China
| | - Jun Yang
- Department of Urology, The University of Kansas Medical Center, Kansas City, Kansas
- Department of Urology, Tongji Hospital, Huanzhong University of Science and Technology, Wuhan, China
| | - Hao Zhang
- Department of Urology, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Ruibao Chen
- Department of Urology, The University of Kansas Medical Center, Kansas City, Kansas
- Department of Urology, Tongji Hospital, Huanzhong University of Science and Technology, Wuhan, China
| | - Liang Dong
- Department of Urology, The University of Kansas Medical Center, Kansas City, Kansas
| | - Yan Huang
- Department of Urology, The University of Kansas Medical Center, Kansas City, Kansas
| | - Dongying Wang
- Department of Urology, The University of Kansas Medical Center, Kansas City, Kansas
| | - Jihong Liu
- Department of Urology, Tongji Hospital, Huanzhong University of Science and Technology, Wuhan, China
| | - Benyi Li
- Department of Urology, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- Department of Urology, The University of Kansas Medical Center, Kansas City, Kansas
- Department of Pathology, China Three Gorges University School of Medicine, Yichang, China
| |
Collapse
|
18
|
Li B, Li C. Suppression of Prostate Cancer Metastasis by DPYSL3-Targeted saRNA. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017. [DOI: 10.1007/978-981-10-4310-9_15] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
19
|
Evans JC, Malhotra M, Cryan JF, O'Driscoll CM. The therapeutic and diagnostic potential of the prostate specific membrane antigen/glutamate carboxypeptidase II (PSMA/GCPII) in cancer and neurological disease. Br J Pharmacol 2016; 173:3041-3079. [PMID: 27526115 PMCID: PMC5056232 DOI: 10.1111/bph.13576] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2016] [Revised: 07/08/2016] [Accepted: 07/25/2016] [Indexed: 12/11/2022] Open
Abstract
Prostate specific membrane antigen (PSMA) otherwise known as glutamate carboxypeptidase II (GCPII) is a membrane bound protein that is highly expressed in prostate cancer and in the neovasculature of a wide variety of tumours including glioblastomas, breast and bladder cancers. This protein is also involved in a variety of neurological diseases including schizophrenia and ALS. In recent years, there has been a surge in the development of both diagnostics and therapeutics that take advantage of the expression and activity of PSMA/GCPII. These include gene therapy, immunotherapy, chemotherapy and radiotherapy. In this review, we discuss the biological roles that PSMA/GCPII plays, both in normal and diseased tissues, and the current therapies exploiting its activity that are at the preclinical stage. We conclude by giving an expert opinion on the future direction of PSMA/GCPII based therapies and diagnostics and hurdles that need to be overcome to make them effective and viable.
Collapse
Affiliation(s)
- James C Evans
- Pharmacodelivery Group, School of Pharmacy, University College Cork, Cork, Ireland
| | - Meenakshi Malhotra
- Pharmacodelivery Group, School of Pharmacy, University College Cork, Cork, Ireland
| | - John F Cryan
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | | |
Collapse
|
20
|
Li C, Jiang W, Hu Q, Li LC, Dong L, Chen R, Zhang Y, Tang Y, Thrasher JB, Liu CB, Li B. Enhancing DPYSL3 gene expression via a promoter-targeted small activating RNA approach suppresses cancer cell motility and metastasis. Oncotarget 2016; 7:22893-910. [PMID: 27014974 PMCID: PMC5008410 DOI: 10.18632/oncotarget.8290] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Accepted: 02/23/2016] [Indexed: 12/11/2022] Open
Abstract
To explore a novel strategy in suppressing tumor metastasis, we took the advantage of a recent RNA activation (RNAa) theory and used small double-strand RNA molecules, termed as small activating RNAs (saRNA) that are complimentary to target gene promoter, to enhance transcription of metastasis suppressor gene. The target gene in this study is Dihydro-pyrimidinase-like 3 (DPYSL3, protein name CRMP4), which was identified as a metastatic suppressor in prostate cancers. There are two transcriptional variants of DPYSL3 gene in human genome, of which the variant 2 is the dominant transcript (DPYSL3v2, CRMP4a) but is also significantly down-regulated in primary prostate cancers. A total of 8 saRNAs for DPYSL3v1 and 14 saRNAs for DPYSL3v2 were tested in multiple prostate cancer cell lines. While none of the saRNAs significantly altered DPYSL3v1 expression, 4 saRNAs showed a strong enhancing effect on DPYSL3v2 expression, resulting in reduced cell mobility in vitro. To achieve a prostate cancer-specific delivery for in vivo testing, we conjugated the most potent saV2-9 RNA molecule with the prostate-specific membrane antigen (PSMA)-targeting aptamer A10-3.2. The conjugates successful increased DPYSL3v2 gene expression in PSMA-positive but not PSMA-negative prostate cancer cells. In nude mice bearing orthotopic xenograft of prostate cancer, a 10-day consecutive treatment with the saV2-9 conjugates significantly suppress distal metastasis compared to the control saRNAs. Analysis of xenograft tissues revealed that DPYSL3v2 expression was largely increased in saV2-9 conjugate-treated group compared to the control group. In conclusion, DPYSL3v2 promoter-targeted saRNA molecules might be used as an adjunctive therapy to suppress prostate cancer metastasis.
Collapse
Affiliation(s)
- Changlin Li
- Department of Urology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Wencong Jiang
- Department of Urology, University of Kansas Medical Center, Kansas City, KS 66160, USA
- Department of Urology, The Affiliated Hospital, Guangdong Medical University, Zhanjiang 524001, China
| | - Qingting Hu
- Department of Urology, University of Kansas Medical Center, Kansas City, KS 66160, USA
- Institute of Cell Therapy, China Three Gorges University, Yichang 443002, China
| | - Long-cheng Li
- Laboratory of Molecular Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing 100073, China
| | - Liang Dong
- Department of Urology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Ruibao Chen
- Department of Urology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Yinghong Zhang
- Department of Urology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Yuzhe Tang
- Department of Urology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - J. Brantley Thrasher
- Department of Urology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Chang-Bai Liu
- Institute of Cell Therapy, China Three Gorges University, Yichang 443002, China
| | - Benyi Li
- Department of Urology, University of Kansas Medical Center, Kansas City, KS 66160, USA
- Department of Urology, The Affiliated Hospital, Guangdong Medical University, Zhanjiang 524001, China
- Institute of Cell Therapy, China Three Gorges University, Yichang 443002, China
| |
Collapse
|
21
|
Novel roles for class II Phosphoinositide 3-Kinase C2β in signalling pathways involved in prostate cancer cell invasion. Sci Rep 2016; 6:23277. [PMID: 26983806 PMCID: PMC4794650 DOI: 10.1038/srep23277] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Accepted: 03/03/2016] [Indexed: 12/14/2022] Open
Abstract
Phosphoinositide 3-kinases (PI3Ks) regulate several cellular functions such as proliferation, growth, survival and migration. The eight PI3K isoforms are grouped into three classes and the three enzymes belonging to the class II subfamily (PI3K-C2α, β and γ) are the least investigated amongst all PI3Ks. Interest on these isoforms has been recently fuelled by the identification of specific physiological roles for class II PI3Ks and by accumulating evidence indicating their involvement in human diseases. While it is now established that these isoforms can regulate distinct cellular functions compared to other PI3Ks, there is still a limited understanding of the signalling pathways that can be specifically regulated by class II PI3Ks. Here we show that PI3K-C2β regulates mitogen-activated protein kinase kinase (MEK1/2) and extracellular signal-regulated kinase (ERK1/2) activation in prostate cancer (PCa) cells. We further demonstrate that MEK/ERK and PI3K-C2β are required for PCa cell invasion but not proliferation. In addition we show that PI3K-C2β but not MEK/ERK regulates PCa cell migration as well as expression of the transcription factor Slug. These data identify novel signalling pathways specifically regulated by PI3K-C2β and they further identify this enzyme as a key regulator of PCa cell migration and invasion.
Collapse
|