1
|
Blanchard Z, Brown EA, Ghazaryan A, Welm AL. PDX models for functional precision oncology and discovery science. Nat Rev Cancer 2024:10.1038/s41568-024-00779-3. [PMID: 39681638 DOI: 10.1038/s41568-024-00779-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/19/2024] [Indexed: 12/18/2024]
Abstract
Precision oncology relies on detailed molecular analysis of how diverse tumours respond to various therapies, with the aim to optimize treatment outcomes for individual patients. Patient-derived xenograft (PDX) models have been key to preclinical validation of precision oncology approaches, enabling the analysis of each tumour's unique genomic landscape and testing therapies that are predicted to be effective based on specific mutations, gene expression patterns or signalling abnormalities. To extend these standard precision oncology approaches, the field has strived to complement the otherwise static and often descriptive measurements with functional assays, termed functional precision oncology (FPO). By utilizing diverse PDX and PDX-derived models, FPO has gained traction as an effective preclinical and clinical tool to more precisely recapitulate patient biology using in vivo and ex vivo functional assays. Here, we explore advances and limitations of PDX and PDX-derived models for precision oncology and FPO. We also examine the future of PDX models for precision oncology in the age of artificial intelligence. Integrating these two disciplines could be the key to fast, accurate and cost-effective treatment prediction, revolutionizing oncology and providing patients with cancer with the most effective, personalized treatments.
Collapse
Affiliation(s)
- Zannel Blanchard
- Department of Oncological Sciences, University of Utah, Huntsman Cancer Institute, Salt Lake City, UT, USA
| | - Elisabeth A Brown
- Department of Oncological Sciences, University of Utah, Huntsman Cancer Institute, Salt Lake City, UT, USA
| | - Arevik Ghazaryan
- Department of Oncological Sciences, University of Utah, Huntsman Cancer Institute, Salt Lake City, UT, USA
| | - Alana L Welm
- Department of Oncological Sciences, University of Utah, Huntsman Cancer Institute, Salt Lake City, UT, USA.
| |
Collapse
|
2
|
Pallavi R, Gatti E, Durfort T, Stendardo M, Ravasio R, Leonardi T, Falvo P, Duso BA, Punzi S, Xieraili A, Polazzi A, Verrelli D, Trastulli D, Ronzoni S, Frascolla S, Perticari G, Elgendy M, Varasi M, Colombo E, Giorgio M, Lanfrancone L, Minucci S, Mazzarella L, Pelicci PG. Caloric restriction leads to druggable LSD1-dependent cancer stem cells expansion. Nat Commun 2024; 15:828. [PMID: 38280853 PMCID: PMC10821871 DOI: 10.1038/s41467-023-44348-y] [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: 08/07/2023] [Accepted: 12/10/2023] [Indexed: 01/29/2024] Open
Abstract
Caloric Restriction (CR) has established anti-cancer effects, but its clinical relevance and molecular mechanism remain largely undefined. Here, we investigate CR's impact on several mouse models of Acute Myeloid Leukemias, including Acute Promyelocytic Leukemia, a subtype strongly affected by obesity. After an initial marked anti-tumor effect, lethal disease invariably re-emerges. Initially, CR leads to cell-cycle restriction, apoptosis, and inhibition of TOR and insulin/IGF1 signaling. The relapse, instead, is associated with the non-genetic selection of Leukemia Initiating Cells and the downregulation of double-stranded RNA (dsRNA) sensing and Interferon (IFN) signaling genes. The CR-induced adaptive phenotype is highly sensitive to pharmacological or genetic ablation of LSD1, a lysine demethylase regulating both stem cells and dsRNA/ IFN signaling. CR + LSD1 inhibition leads to the re-activation of dsRNA/IFN signaling, massive RNASEL-dependent apoptosis, and complete leukemia eradication in ~90% of mice. Importantly, CR-LSD1 interaction can be modeled in vivo and in vitro by combining LSD1 ablation with pharmacological inhibitors of insulin/IGF1 or dual PI3K/MEK blockade. Mechanistically, insulin/IGF1 inhibition sensitizes blasts to LSD1-induced death by inhibiting the anti-apoptotic factor CFLAR. CR and LSD1 inhibition also synergize in patient-derived AML and triple-negative breast cancer xenografts. Our data provide a rationale for epi-metabolic pharmacologic combinations across multiple tumors.
Collapse
Affiliation(s)
- Rani Pallavi
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Elena Gatti
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Tiphanie Durfort
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Massimo Stendardo
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Roberto Ravasio
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Tommaso Leonardi
- Center for Genomic Science of IIT@SEMM, Fondazione Istituto Italiano di Tecnologia, Milan, Italy
| | - Paolo Falvo
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Bruno Achutti Duso
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Simona Punzi
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Aobuli Xieraili
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Andrea Polazzi
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Doriana Verrelli
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Deborah Trastulli
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Simona Ronzoni
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Simone Frascolla
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Giulia Perticari
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Mohamed Elgendy
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Milan, Italy
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital and Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
- Medical Clinic I, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Mildred-Scheel Early Career Center, National Center for Tumor Diseases Dresden (NCT/UCC) University Hospital and Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
- Cancer Cell Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, CZ-14220, Czech Republic
| | - Mario Varasi
- IFOM ETS - The AIRC Institute of Molecular Oncology, Milan, Italy
| | - Emanuela Colombo
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Milan, Italy
- Department of Hemato-Oncology, Universita' Statale di Milano, Milan, Italy
| | - Marco Giorgio
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Milan, Italy
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Luisa Lanfrancone
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Saverio Minucci
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Milan, Italy
- Department of Hemato-Oncology, Universita' Statale di Milano, Milan, Italy
| | - Luca Mazzarella
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Milan, Italy.
| | - Pier Giuseppe Pelicci
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Milan, Italy.
- Department of Hemato-Oncology, Universita' Statale di Milano, Milan, Italy.
| |
Collapse
|
3
|
Marcolin JC, Lichtenfels M, da Silva CA, de Farias CB. Gynecologic and Breast Cancers: What's New in Chemoresistance and Chemosensitivity Tests? Curr Probl Cancer 2023; 47:100996. [PMID: 37467541 DOI: 10.1016/j.currproblcancer.2023.100996] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 06/14/2023] [Accepted: 07/03/2023] [Indexed: 07/21/2023]
Abstract
Gynecological and breast cancers affect women's health worldwide. Although chemotherapy is one of the principal treatments for cancer, it also has limitations owing to toxicity and tumor resistance to the drugs used. Thus, individualized treatment based on personal tumor characteristics is essential for improving therapeutic outcomes and patient survival. Chemoresistance and chemosensitivity tests can be useful for predicting tumor response and guiding chemotherapy choices. This methodology has already been applied to breast, ovarian, cervical, and endometrial cancers, identifying successfully which drugs cause resistance and sensitivity responses for each individual person, influencing their progression-free survival and overall response. In addition, more recent techniques, such as organoids and patient-derived xenografts, can also recapitulate patients' tumor characteristics and contribute to chemo response evaluation. Therefore, this review compiles information on chemoresistance and chemosensitivity tests performed in gynecologic and breast cancers and their main results for women's health improvement.
Collapse
Affiliation(s)
- Júlia Caroline Marcolin
- Ziel Biosciences, Department of Translational Research, Porto Alegre, Rio Grande do Sul, Brazil; Programa de Pós-Graduação em Farmacologia e Terapêutica, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil.
| | - Martina Lichtenfels
- Ziel Biosciences, Department of Translational Research, Porto Alegre, Rio Grande do Sul, Brazil
| | - Camila Alves da Silva
- Ziel Biosciences, Department of Translational Research, Porto Alegre, Rio Grande do Sul, Brazil
| | | |
Collapse
|
4
|
Nili Ahmadabadi M, Rezaee E, Nematpour M, Karami L, Mokhtari S, Kobarfard F, Tabatabai SA. Synthesis, Molecular Dynamics Simulation, and In-vitro Antitumor Activity of Quinazoline-2,4,6-triamine Derivatives as Novel EGFR Tyrosine Kinase Inhibitors. IRANIAN JOURNAL OF PHARMACEUTICAL RESEARCH : IJPR 2022; 21:e133840. [PMID: 36915409 PMCID: PMC10008000 DOI: 10.5812/ijpr-133840] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 12/24/2022] [Accepted: 01/02/2023] [Indexed: 01/31/2023]
Abstract
Background Developing a potent and safe scaffold is challenging in anti-cancer drug discovery. Objectives The study focused on developing novel series of compounds based on the inhibition of epidermal growth factor receptor tyrosine kinase (EGFR-TK) as one of the most promising compounds in cancer therapy. Methods In this study, a novel series of quinazoline-2,4,6-triamine derivatives were designed and synthesized through intramolecular C-H activation reaction of para-nitro aniline, trichloroacetonitrile, and isocyanides employing a one-pot reaction. Results The in-vitro antitumor activities of the compounds which showed acceptable inhibitory effects were investigated against breast (MCF-7), lung (A-549), and colon (HT-29) cancer cell lines by employing MTT assay. All compounds had the most negligible cytotoxicity toward normal fibroblast human cell lines. Based on structural and thermodynamics analysis results, it was found that Met 769 is a key residue in interaction with all inhibitors through the formation of hydrogen bonds with high occupancies with the amine group on the quinazoline ring of inhibitors. Also, there was a good consistency between calculated ΔG binding and experimental IC50 values of compounds 10d, 10e, and erlotinib. Conclusions Compound 10e had an extensive range of antitumor activity on three diverse cell lines comparable with erlotinib and doxorubicin reference drugs. Also, compound 10d showed selective cytotoxicity against cancerous lung cells (A-549). On the other side, computational studies confirmed that Met 769 is a crucial residue in interaction with all inhibitors.
Collapse
Affiliation(s)
- Maryam Nili Ahmadabadi
- Department of Pharmaceutical Chemistry, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Elham Rezaee
- Department of Pharmaceutical Chemistry, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Manijeh Nematpour
- Department of Pharmaceutical Chemistry, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Leila Karami
- Department of Cell and Molecular Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Shaya Mokhtari
- Central Research Laboratories, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Phytochemistry Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Farzad Kobarfard
- Department of Pharmaceutical Chemistry, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sayyed Abbas Tabatabai
- Department of Pharmaceutical Chemistry, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Corresponding Author: Department of Pharmaceutical Chemistry, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
5
|
Single cell RNA-seq reveals the CCL5/SDC1 receptor-ligand interaction between T cells and tumor cells in pancreatic cancer. Cancer Lett 2022; 545:215834. [DOI: 10.1016/j.canlet.2022.215834] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 07/04/2022] [Accepted: 07/17/2022] [Indexed: 12/15/2022]
|
6
|
Lee WJ, Tu SH, Cheng TC, Lin JH, Sheu MT, Kuo CC, Changou CA, Wu CH, Chang HW, Chang HL, Chen LC, Ho YS. Type-3 Hyaluronan Synthase Attenuates Tumor Cells Invasion in Human Mammary Parenchymal Tissues. Molecules 2021; 26:molecules26216548. [PMID: 34770956 PMCID: PMC8587416 DOI: 10.3390/molecules26216548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 10/22/2021] [Accepted: 10/26/2021] [Indexed: 12/24/2022] Open
Abstract
The microenvironment for tumor growth and developing metastasis should be essential. This study demonstrated that the hyaluronic acid synthase 3 (HAS3) protein and its enzymatic product hyaluronic acid (HA) encompassed in the subcutaneous extracellular matrix can attenuate the invasion of human breast tumor cells. Decreased HA levels in subcutaneous Has3-KO mouse tissues promoted orthotopic breast cancer (E0771) cell-derived allograft tumor growth. MDA-MB-231 cells premixed with higher concentration HA attenuate tumor growth in xenografted nude mice. Human patient-derived xenotransplantation (PDX) experiments found that HA selected the highly migratory breast cancer cells with CD44 expression accumulated in the tumor/stroma junction. In conclusion, HAS3 and HA were detected in the stroma breast tissues at a high level attenuates effects for induced breast cancer cell death, and inhibit the cancer cells invasion at the initial stage. However, the highly migratory cancer cells were resistant to the HA-mediated effects with unknown mechanisms.
Collapse
Affiliation(s)
- Wen-Jui Lee
- Ph.D. Program for Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan;
- Ph.D. Program for Neural Regenerative Medicine, College of Medical Science and Technology, National Health Research Institutes, Miaoli County 350, Taiwan
| | - Shih-Hsin Tu
- Breast Medical Center, Taipei Medical University Hospital, Taipei 110, Taiwan;
- Taipei Cancer Center, Taipei Medical University, Taipei 110, Taiwan
- Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Tzu-Chun Cheng
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 110, Taiwan;
| | - Juo-Han Lin
- Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University and Academia Sinica, Taipei 110, Taiwan;
| | - Ming-Thau Sheu
- Department of Pharmaceutical Sciences, Taipei Medical University, Taipei 110, Taiwan;
| | - Ching-Chuan Kuo
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan 350, Taiwan;
| | - Chun A. Changou
- The PhD Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan;
- The PhD Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University and Academia Sinica, Taipei 110, Taiwan
- The Core Facility Center, Office of Research and Development, Taipei Medical University, Taipei 110, Taiwan
| | - Chih-Hsiung Wu
- Department of General Surgery, En Chu Kong Hospital, New Taipei City 110, Taiwan;
| | - Hui-Wen Chang
- Department of Laboratory Medicine, Taipei Medical University Hospital, Taipei 110, Taiwan;
| | - Hang-Lung Chang
- Department of General Surgery, En Chu Kong Hospital, New Taipei City 237, Taiwan;
| | - Li-Ching Chen
- Breast Medical Center, Taipei Medical University Hospital, Taipei 110, Taiwan;
- Taipei Cancer Center, Taipei Medical University, Taipei 110, Taiwan
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei 110, Taiwan
- Correspondence: (L.-C.C.); (Y.-S.H.)
| | - Yuan-Soon Ho
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 110, Taiwan;
- Department of Laboratory Medicine, Taipei Medical University Hospital, Taipei 110, Taiwan;
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei 110, Taiwan
- Graduate Institute of Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan
- Correspondence: (L.-C.C.); (Y.-S.H.)
| |
Collapse
|
7
|
Honkala A, Malhotra SV, Kummar S, Junttila MR. Harnessing the predictive power of preclinical models for oncology drug development. Nat Rev Drug Discov 2021; 21:99-114. [PMID: 34702990 DOI: 10.1038/s41573-021-00301-6] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/27/2021] [Indexed: 12/21/2022]
Abstract
Recent progress in understanding the molecular basis of cellular processes, identification of promising therapeutic targets and evolution of the regulatory landscape makes this an exciting and unprecedented time to be in the field of oncology drug development. However, high costs, long development timelines and steep rates of attrition continue to afflict the drug development process. Lack of predictive preclinical models is considered one of the key reasons for the high rate of attrition in oncology. Generating meaningful and predictive results preclinically requires a firm grasp of the relevant biological questions and alignment of the model systems that mirror the patient context. In doing so, the ability to conduct both forward translation, the process of implementing basic research discoveries into practice, as well as reverse translation, the process of elucidating the mechanistic basis of clinical observations, greatly enhances our ability to develop effective anticancer treatments. In this Review, we outline issues in preclinical-to-clinical translatability of molecularly targeted cancer therapies, present concepts and examples of successful reverse translation, and highlight the need to better align tumour biology in patients with preclinical model systems including tracking of strengths and weaknesses of preclinical models throughout programme development.
Collapse
Affiliation(s)
- Alexander Honkala
- Department of Cell Development & Cancer Biology, Oregon Health & Science University, Portland, OR, USA
| | - Sanjay V Malhotra
- Department of Cell Development & Cancer Biology, Oregon Health & Science University, Portland, OR, USA.,Center for Experimental Therapeutics, Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| | - Shivaani Kummar
- Center for Experimental Therapeutics, Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA. .,Division of Hematology & Medical Oncology, Oregon Health & Science University, Portland, OR, USA.
| | | |
Collapse
|
8
|
John A, Qin B, Kalari KR, Wang L, Yu J. Patient-specific multi-omics models and the application in personalized combination therapy. Future Oncol 2020; 16:1737-1750. [PMID: 32462937 DOI: 10.2217/fon-2020-0119] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The rapid advancement of high-throughput technologies and sharp decrease in cost have opened up the possibility to generate large amount of multi-omics data on an individual basis. The development of high-throughput -omics, including genomics, epigenomics, transcriptomics, proteomics, metabolomics and microbiomics, enables the application of multi-omics technologies in the clinical settings. Combination therapy, defined as disease treatment with two or more drugs to achieve efficacy with lower doses or lower drug toxicity, is the basis for the care of diseases like cancer. Patient-specific multi-omics data integration can help the identification and development of combination therapies. In this review, we provide an overview of different -omics platforms, and discuss the methods for multi-omics, high-throughput, data integration, personalized combination therapy.
Collapse
Affiliation(s)
- August John
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN 55905, USA
| | - Bo Qin
- Department of Molecular Pharmacology & Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA.,Gastroenterology Research Unit, Mayo Clinic, Rochester, MN 55905, USA.,Department of Oncology, Mayo Clinic, Rochester, MN 55905, USA
| | - Krishna R Kalari
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN 55905, USA
| | - Liewei Wang
- Department of Molecular Pharmacology & Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA
| | - Jia Yu
- Department of Molecular Pharmacology & Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
9
|
Zhou X, Li R, Chen R, Liu J. Altered Mitochondrial Dynamics, Biogenesis, and Functions in the Paclitaxel-Resistant Lung Adenocarcinoma Cell Line A549/Taxol. Med Sci Monit 2020; 26:e918216. [PMID: 32129321 PMCID: PMC7071736 DOI: 10.12659/msm.918216] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Chemoresistance is a primary hindrance for current cancer treatments. The influence of abnormal mitochondria in chemotherapy resistance is not well known. To explore the correlation between mitochondria and acquired chemoresistance, this work studied alterations in mitochondrial dynamics, biogenesis, and functions for paclitaxel-resistant cancer cell line A549/Taxol and its parental line A549. MATERIAL AND METHODS Mitochondrial morphology was observed by transmission electron microscopy and confocal microscopy. We measured the mitochondrial mass and mitochondrial membrane potential using fluorescent dyes. The glucose metabolic profile and ATP (adenosine triphosphate) content were determined by bioluminescent cell assays. Seahorse bio-energy analyzer XF24 was used to detect the mitochondrial respiratory function. The expressions of mitochondrial dynamics and biogenesis related genes were quantified using real-time polymerase chain reaction. RESULTS We observed fusion morphology of the mitochondrial network in A549/Taxol cells, with upregulation of fusion genes (Mfn1 and Mfn2) and downregulation of fission gene Fis1. In A549/Taxol cells, mitochondrial mass showed a significant decrease, while the mitochondrial biogenesis pathway was strongly activated. Despite the decreased mitochondrial membrane potential, the capability for mitochondrial respiration was not impaired in A549/Taxol cells. CONCLUSIONS Our study revealed a series changes of mitochondrial characteristics in paclitaxel-resistant cells. Mfn1 and Mfn2 and PGC-1alpha increased, while Fis1 expression and mitochondrial oxidative phosphorylation decreased in A549/Taxol cell lines. These changes to mitochondrial fusion, fission, and biological function contributed to the occurrence of paclitaxel resistance in tumor cells which induced paclitaxel resistance.
Collapse
Affiliation(s)
- Xiang Zhou
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China (mainland)
| | - Rui Li
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China (mainland)
| | - Ruohua Chen
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China (mainland)
| | - Jianjun Liu
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China (mainland)
| |
Collapse
|
10
|
Invrea F, Rovito R, Torchiaro E, Petti C, Isella C, Medico E. Patient-derived xenografts (PDXs) as model systems for human cancer. Curr Opin Biotechnol 2020; 63:151-156. [PMID: 32070860 DOI: 10.1016/j.copbio.2020.01.003] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2020] [Accepted: 01/08/2020] [Indexed: 12/13/2022]
Abstract
Patient-derived xenografts (PDXs) are obtained by transplanting fragments of a patient's tumour into immunodeficient mice. Growth and propagation of PDXs allows correlating therapeutic response in vivo with extensive, multi-dimensional molecular annotation, leading to identification of predictive biomarkers. PDXs are increasingly recognised as clinically relevant models of cancer for several reasons, of which the main is the possibility of studying the behaviour of cancer cells in a natural microenvironment, where they interact with stromal components accrued from the mouse host. PDXs maintain close similarities with the tumour of origin, in terms of tissue architecture, molecular features and response to treatments. Indeed, preclinical trials in PDXs have been shown to match and also anticipate data obtained in patients. Exploration of more complex processes like metastatic evolution and antitumour immune responses is actively pursued with PDXs, as new generations of host models emerge on the horizon.
Collapse
Affiliation(s)
- Federica Invrea
- Candiolo Cancer Institute, FPO-IRCCS, strada Prov. 142, km 3,95, 10060 Candiolo (TO), Italy
| | - Roberta Rovito
- Candiolo Cancer Institute, FPO-IRCCS, strada Prov. 142, km 3,95, 10060 Candiolo (TO), Italy
| | - Erica Torchiaro
- Candiolo Cancer Institute, FPO-IRCCS, strada Prov. 142, km 3,95, 10060 Candiolo (TO), Italy
| | - Consalvo Petti
- Candiolo Cancer Institute, FPO-IRCCS, strada Prov. 142, km 3,95, 10060 Candiolo (TO), Italy
| | - Claudio Isella
- Candiolo Cancer Institute, FPO-IRCCS, strada Prov. 142, km 3,95, 10060 Candiolo (TO), Italy; University of Torino, Department of Oncology, strada Prov. 142, km 3,95, 10060 Candiolo (TO), Italy
| | - Enzo Medico
- Candiolo Cancer Institute, FPO-IRCCS, strada Prov. 142, km 3,95, 10060 Candiolo (TO), Italy; University of Torino, Department of Oncology, strada Prov. 142, km 3,95, 10060 Candiolo (TO), Italy.
| |
Collapse
|
11
|
Liu J, Li Y, Zhao M, Lei Z, Guo H, Tang Y, Yan H. Redox-responsive hollow mesoporous silica nanoparticles constructed via host-guest interactions for controllable drug release. JOURNAL OF BIOMATERIALS SCIENCE-POLYMER EDITION 2019; 31:472-490. [PMID: 31791208 DOI: 10.1080/09205063.2019.1700601] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
A novel redox-responsive hollow mesoporous silica (HMS) was constructed by host-guest interaction between β-cyclodextrin modified hollow mesoporus silica nanoparticles (HMS@β-CD) and the ferrocene-containing amphiphilic block copolymer PEG-b-PMAFc (PPFc), the prepared HMS@β-CD@PPFc system was used to control drug delivery in targeted cancer therapy through redox stimulus. The self-assembled morphology was investigated by transmission electron microscopy (TEM) and dynamic light scattering (DLS). Intracellular localization of DOX-loaded HMS@β-CD@PPFc in A549 cells was further investigated by confocal laser scanning microscopy (CLSM), and the results indicated that DOX-loaded HMS@β-CD@PPFc was ingested by A549 cells effectively. Furthermore, the redox agent H2O2 was used to trigger the release of DOX. The cytotoxicity evaluated by MTT method indicated that HMS@β-CD@PPFc had good biocompatibility and was promising as the drug carrier.
Collapse
Affiliation(s)
- Jiangtao Liu
- College of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Yan Li
- Key Laboratory of Applied Surface and Colloid Chemistry, School of Chemistry & Chemical Engineering, Shaanxi Normal University, Xi'an, China
| | - Min Zhao
- Key Laboratory of Applied Surface and Colloid Chemistry, School of Chemistry & Chemical Engineering, Shaanxi Normal University, Xi'an, China
| | - Zhongli Lei
- Key Laboratory of Applied Surface and Colloid Chemistry, School of Chemistry & Chemical Engineering, Shaanxi Normal University, Xi'an, China
| | - Hui Guo
- College of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Yuping Tang
- College of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Hao Yan
- College of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang, China
| |
Collapse
|
12
|
Punzi S, Balestrieri C, D'Alesio C, Bossi D, Dellino GI, Gatti E, Pruneri G, Criscitiello C, Lovati G, Meliksetyan M, Carugo A, Curigliano G, Natoli G, Pelicci PG, Lanfrancone L. WDR5 inhibition halts metastasis dissemination by repressing the mesenchymal phenotype of breast cancer cells. Breast Cancer Res 2019; 21:123. [PMID: 31752957 PMCID: PMC6873410 DOI: 10.1186/s13058-019-1216-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Accepted: 10/16/2019] [Indexed: 02/07/2023] Open
Abstract
Background Development of metastases and drug resistance are still a challenge for a successful systemic treatment in breast cancer (BC) patients. One of the mechanisms that confer metastatic properties to the cell relies in the epithelial-to-mesenchymal transition (EMT). Moreover, both EMT and metastasis are partly modulated through epigenetic mechanisms, by repression or induction of specific related genes. Methods We applied shRNAs and drug targeting approaches in BC cell lines and metastatic patient-derived xenograft (PDX) models to inhibit WDR5, the core subunit of histone H3 K4 methyltransferase complexes, and evaluate its role in metastasis regulation. Result We report that WDR5 is crucial in regulating tumorigenesis and metastasis spreading during BC progression. In particular, WDR5 loss reduces the metastatic properties of the cells by reverting the mesenchymal phenotype of triple negative- and luminal B-derived cells, thus inducing an epithelial trait. We also suggest that this regulation is mediated by TGFβ1, implying a prominent role of WDR5 in driving EMT through TGFβ1 activation. Moreover, such EMT reversion can be induced by drug targeting of WDR5 as well, leading to BC cell sensitization to chemotherapy and enhancement of paclitaxel-dependent effects. Conclusions We suggest that WDR5 inhibition could be a promising pharmacologic approach to reduce cell migration, revert EMT, and block metastasis formation in BC, thus overcoming resistance to standard treatments.
Collapse
Affiliation(s)
- Simona Punzi
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy
| | - Chiara Balestrieri
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy.,Humanitas University, Pieve Emanuele (MI), 20090, Italy.,Humanitas Clinical and Research Institute, Rozzano (MI), 20089, Italy
| | - Carolina D'Alesio
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy.,Present address: Department of Internal Medicine and Medical Specialties (Di.M.I), University of Genova, Genoa, Italy
| | - Daniela Bossi
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy.,Present address: Institute of Oncology Research (IOR), Bellinzona, Switzerland
| | - Gaetano Ivan Dellino
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy.,Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Elena Gatti
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy
| | - Giancarlo Pruneri
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy.,Department of Pathology, Biobank for Translational Medicine Unit, European Institute of Oncology, IRCCS, Milan, Italy.,Present address: Istituto Nazionale dei Tumori - Fondazione IRCCS, Milan, Italy
| | - Carmen Criscitiello
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy.,Division of Early Drug Development for Innovative Therapy, European Institute of Oncology IRCCS, Milan, Italy
| | - Giulia Lovati
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy
| | - Marine Meliksetyan
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy
| | - Alessandro Carugo
- Institute for Applied Cancer Science, UT MD Anderson Cancer Cente, Houston, TX, 77030, USA
| | - Giuseppe Curigliano
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy.,Division of Early Drug Development for Innovative Therapy, European Institute of Oncology IRCCS, Milan, Italy
| | - Gioacchino Natoli
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy.,Humanitas University, Pieve Emanuele (MI), 20090, Italy.,Humanitas Clinical and Research Institute, Rozzano (MI), 20089, Italy
| | - Pier Giuseppe Pelicci
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy.,Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Luisa Lanfrancone
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy.
| |
Collapse
|