1
|
De Robertis M, Bozic T, Santek I, Marzano F, Markelc B, Silvestris DA, Tullo A, Pesole G, Cemazar M, Signori E. Transcriptomic analysis of the immune response to in vivo gene electrotransfer in colorectal cancer. MOLECULAR THERAPY. NUCLEIC ACIDS 2025; 36:102448. [PMID: 39967849 PMCID: PMC11834060 DOI: 10.1016/j.omtn.2025.102448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 01/10/2025] [Indexed: 02/20/2025]
Abstract
Gene electrotransfer (GET) has recently emerged as an effective nonviral approach for plasmid DNA (pDNA) delivery in gene therapy for several pathologies, including cancer. Multiple mechanisms have been identified that influence cell biology after GET, as electroporation significantly increases pDNA uptake and immunogenicity, which may directly influence target cell death. However, the molecular effects of in vivo electroporation-mediated DNA delivery have yet to be fully elucidated. In this study, we evaluated the transcriptomes of murine colorectal tumors treated with two protocols, short- and high-voltage (SHV) electric pulses or an adapted high-voltage-low-voltage (HV-LV) pulse protocol, both of which are used for reversible electroporation. Although no significant differences in clinical outcomes were observed, variations in intratumoral macrophage infiltration were reported between the two treatment methods. Transcriptomic analysis revealed that apoptosis is a predominant mode of cell death after GET by SHV pulses, whereas GET by HV-LV pulses is associated with immunogenic necrotic pathways as well as the activation of both the innate and adaptive immune response. We demonstrated that specific pulse parameters can induce distinct immunomodulatory profiles in the tumor microenvironment; therefore, these aspects should be considered carefully when selecting the most suitable GET-based approach for antitumor immunization.
Collapse
Affiliation(s)
- Mariangela De Robertis
- Department of Biosciences, Biotechnology, and Environment, University of Bari “Aldo Moro”, 70126 Bari, Italy
- Institute of Biomembranes, Bioenergetics, and Molecular Biotechnologies, Consiglio Nazionale delle Ricerche, 70126 Bari, Italy
| | - Tim Bozic
- Department of Experimental Oncology, Institute of Oncology Ljubljana, Zaloska cesta 2, 1000 Ljubljana, Slovenia
| | - Iva Santek
- Department of Experimental Oncology, Institute of Oncology Ljubljana, Zaloska cesta 2, 1000 Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, Vrazov trg 2, 1000 Ljubljana, Slovenia
| | - Flaviana Marzano
- Institute of Biomembranes, Bioenergetics, and Molecular Biotechnologies, Consiglio Nazionale delle Ricerche, 70126 Bari, Italy
| | - Bostjan Markelc
- Department of Experimental Oncology, Institute of Oncology Ljubljana, Zaloska cesta 2, 1000 Ljubljana, Slovenia
- Biotechnical Faculty, University of Ljubljana, Jamnikarjeva ulica 101, 1000 Ljubljana, Slovenia
| | | | - Apollonia Tullo
- Institute of Biomembranes, Bioenergetics, and Molecular Biotechnologies, Consiglio Nazionale delle Ricerche, 70126 Bari, Italy
| | - Graziano Pesole
- Department of Biosciences, Biotechnology, and Environment, University of Bari “Aldo Moro”, 70126 Bari, Italy
- Institute of Biomembranes, Bioenergetics, and Molecular Biotechnologies, Consiglio Nazionale delle Ricerche, 70126 Bari, Italy
| | - Maja Cemazar
- Department of Experimental Oncology, Institute of Oncology Ljubljana, Zaloska cesta 2, 1000 Ljubljana, Slovenia
- Faculty of Health Sciences, University of Primorska, Polje 42, 6310 Izola, Slovenia
| | - Emanuela Signori
- Laboratory of Molecular Pathology and Experimental Oncology, Institute of Translational Pharmacology, Consiglio Nazionale delle Ricerche, 0133 Rome, Italy
| |
Collapse
|
2
|
Xia Y, Li X, Liu F. Targeted redox-responsive peptide for arterial chemoembolization therapy of orthotropic hepatocellular carcinoma. Abdom Radiol (NY) 2024; 49:3925-3934. [PMID: 38990300 PMCID: PMC11519146 DOI: 10.1007/s00261-024-04481-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Revised: 06/27/2024] [Accepted: 06/30/2024] [Indexed: 07/12/2024]
Abstract
OBJECTIVE Transcatheter Arterial Chemoembolization (TACE) is the first choice for the treatment of advanced-stage hepatocellular carcinoma (HCC). However, TACE suffers from a lack of specificity and rapid drug release. Herein, a targeted redox-responsive peptide (TRRP) was synthesized and used as a carrier of doxorubicin (DOX) to enhance the efficacy of TACE through tumor cells targeting and controlled drug release. METHODS TRRP has a high loading capacity of DOX and a sensitive drug release behavior at high glutathione (GSH) concentration. Moreover, TRRP could bind to the transferrin receptor on the surface of tumor cells, which enhanced the efficacy of TACE and reduced side effects of TACE. TACE with TRRP@DOX dispersed in lipiodol shows an enhanced therapeutic outcome compared to the treatment with DOX + lipiodol emulsion in orthotopic rat HCC models. RESULTS TRRP has a high loading capacity of DOX and a sensitive drug release behavior at GSH concentration. Moreover, TRRP could bind to the transferrin receptor on the surface of tumor cells, which enhanced the efficacy of TACE and reduced side effects of TACE. TACE with TRRP@DOX dispersed in lipiodol shows an enhanced therapeutic outcome compared to the treatment with DOX + lipiodol emulsion in orthotopic rat HCC models. CONCLUSIONS This study demonstrated that TRRP was a promising therapeutic agent for enhancing TACE therapy for HCC treatment.
Collapse
Affiliation(s)
- Yimao Xia
- Chinese PLA Medical School, Beijing, 100853, China
| | - Xin Li
- Department of Interventional Radiology, The Fifth Medical Center of PLA General Hospital, Beijing, 100039, China
| | - Fengyong Liu
- Chinese PLA Medical School, Beijing, 100853, China.
- Department of Interventional Radiology, The Fifth Medical Center of PLA General Hospital, Beijing, 100039, China.
| |
Collapse
|
3
|
Meric-Bernstam F, Lloyd MW, Koc S, Evrard YA, McShane LM, Lewis MT, Evans KW, Li D, Rubinstein L, Welm A, Dean DA, Srivastava A, Grover JW, Ha MJ, Chen H, Huang X, Varadarajan K, Wang J, Roth JA, Welm B, Govinden R, Ding L, Kaochar S, Mitsiades N, Carvajal-Carmona L, Herylyn M, Davies MA, Shapiro GI, Fields R, Trevino JG, Harrell JC, Doroshow JH, Chuang JH, Moscow JA. Assessment of Patient-Derived Xenograft Growth and Antitumor Activity: The NCI PDXNet Consensus Recommendations. Mol Cancer Ther 2024; 23:924-938. [PMID: 38641411 PMCID: PMC11217730 DOI: 10.1158/1535-7163.mct-23-0471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 12/08/2023] [Accepted: 03/29/2024] [Indexed: 04/21/2024]
Abstract
Although patient-derived xenografts (PDX) are commonly used for preclinical modeling in cancer research, a standard approach to in vivo tumor growth analysis and assessment of antitumor activity is lacking, complicating the comparison of different studies and determination of whether a PDX experiment has produced evidence needed to consider a new therapy promising. We present consensus recommendations for assessment of PDX growth and antitumor activity, providing public access to a suite of tools for in vivo growth analyses. We expect that harmonizing PDX study design and analysis and assessing a suite of analytical tools will enhance information exchange and facilitate identification of promising novel therapies and biomarkers for guiding cancer therapy.
Collapse
Affiliation(s)
- Funda Meric-Bernstam
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | | | - Soner Koc
- Seven Bridges Genomics, Charlestown, Massachusetts.
| | - Yvonne A. Evrard
- Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland.
| | - Lisa M. McShane
- Biometric Research Program, DCTD, National Cancer Institute, National Institutes of Health, Bethesda, Maryland.
| | - Michael T. Lewis
- Departments of Molecular and Cellular Biology and Radiology, Lester and Sue Smith Breast Center, Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas.
| | - Kurt W. Evans
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Dali Li
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Lawrence Rubinstein
- Biometric Research Program, DCTD, National Cancer Institute, National Institutes of Health, Bethesda, Maryland.
| | - Alana Welm
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah.
| | | | - Anuj Srivastava
- The Jackson Laboratory for Genomic Medicine, Farmington, Connecticut.
| | | | - Min J. Ha
- Department of Biostatistics, Graduate School of Public Health, Yonsei University, Seoul, Republic of Korea.
| | - Huiqin Chen
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Xuelin Huang
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Kaushik Varadarajan
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Jing Wang
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Jack A. Roth
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Bryan Welm
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah.
| | - Ramaswamy Govinden
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri.
| | - Li Ding
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri.
| | - Salma Kaochar
- Department of Medicine, Baylor College of Medicine, Houston, Texas.
| | - Nicholas Mitsiades
- Department of Molecular Cellular Biology, Baylor College of Medicine, Houston, Texas.
| | - Luis Carvajal-Carmona
- Department of Biochemistry and Molecular Medicine, University of California, Davis, California.
| | | | - Michael A. Davies
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Geoffrey I. Shapiro
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.
| | - Ryan Fields
- Department of Surgery, Washington University School of Medicine, St. Louis, Missouri.
| | - Jose G. Trevino
- Department of Surgery, Virginia Commonwealth University, Richmond, Virginia.
| | - Joshua C. Harrell
- Department of Pathology, Virginia Commonwealth University, Richmond, Virginia.
| | | | - James H. Doroshow
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Bethesda, Maryland.
| | - Jeffrey H. Chuang
- The Jackson Laboratory for Genomic Medicine, Farmington, Connecticut.
| | - Jeffrey A. Moscow
- Investigational Drug Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland.
| |
Collapse
|
4
|
Godoy PM, Oyedeji A, Mudd JL, Morikis VA, Zarov AP, Longmore GD, Fields RC, Kaufman CK. Functional analysis of recurrent CDC20 promoter variants in human melanoma. Commun Biol 2023; 6:1216. [PMID: 38030698 PMCID: PMC10686982 DOI: 10.1038/s42003-023-05526-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 10/30/2023] [Indexed: 12/01/2023] Open
Abstract
Small nucleotide variants in non-coding regions of the genome can alter transcriptional regulation, leading to changes in gene expression which can activate oncogenic gene regulatory networks. Melanoma is heavily burdened by non-coding variants, representing over 99% of total genetic variation, including the well-characterized TERT promoter mutation. However, the compendium of regulatory non-coding variants is likely still functionally under-characterized. We developed a pipeline to identify hotspots, i.e. recurrently mutated regions, in melanoma containing putatively functional non-coding somatic variants that are located within predicted melanoma-specific regulatory regions. We identified hundreds of statistically significant hotspots, including the hotspot containing the TERT promoter variants, and focused on a hotspot in the promoter of CDC20. We found that variants in the promoter of CDC20, which putatively disrupt an ETS motif, lead to lower transcriptional activity in reporter assays. Using CRISPR/Cas9, we generated an indel in the CDC20 promoter in human A375 melanoma cell lines and observed decreased expression of CDC20, changes in migration capabilities, increased growth of xenografts, and an altered transcriptional state previously associated with a more proliferative and less migratory state. Overall, our analysis prioritized several recurrent functional non-coding variants that, through downregulation of CDC20, led to perturbation of key melanoma phenotypes.
Collapse
Affiliation(s)
- Paula M Godoy
- Division of Medical Oncology, Department of Medicine and Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Abimbola Oyedeji
- Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
- Siteman Cancer Center, Washington University in Saint Louis, St. Louis, MO, USA
| | - Jacqueline L Mudd
- Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
- Siteman Cancer Center, Washington University in Saint Louis, St. Louis, MO, USA
| | - Vasilios A Morikis
- Departments of Medicine (Oncology) and Cell Biology and Physiology and the ICCE Institute, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Anna P Zarov
- Division of Medical Oncology, Department of Medicine and Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Gregory D Longmore
- Siteman Cancer Center, Washington University in Saint Louis, St. Louis, MO, USA
- Departments of Medicine (Oncology) and Cell Biology and Physiology and the ICCE Institute, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Ryan C Fields
- Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
- Siteman Cancer Center, Washington University in Saint Louis, St. Louis, MO, USA
| | - Charles K Kaufman
- Division of Medical Oncology, Department of Medicine and Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA.
- Siteman Cancer Center, Washington University in Saint Louis, St. Louis, MO, USA.
| |
Collapse
|
5
|
Jang H, Ojha U, Jeong JH, Park KG, Lee SY, Lee YM. Myriocin suppresses tumor growth by modulating macrophage polarization and function through the PI3K/Akt/mTOR pathway. Arch Pharm Res 2023; 46:629-645. [PMID: 37468765 DOI: 10.1007/s12272-023-01454-1] [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: 02/21/2023] [Accepted: 07/02/2023] [Indexed: 07/21/2023]
Abstract
Macrophages within the tumor microenvironment (TME), referred to as tumor-associated macrophages (TAMs), are involved in various aspects of tumor progression including initiation, angiogenesis, metastasis, immunosuppression, and resistance to therapy. Myriocin, a natural compound isolated from Mycelia sterilia, is an immunosuppressant that inhibits tumor growth and angiogenesis. However, the mechanisms underlying the effects of myriocin on TAMs and TAM-mediated tumor growth have not yet been elucidated. In this study, we determined the effects of myriocin on TAMs and the underlying mechanism in vitro and in vivo. Myriocin significantly suppressed monocyte-macrophage differentiation and M2 polarization of macrophages but not M1 polarization. In addition, myriocin inhibited the expression of anti-inflammatory cytokines and secretion of proangiogenic factors, such as vascular endothelial growth factor, in M2 macrophages as well as M2-induced endothelial cell permeability. Myriocin also inhibited the PI3K/Akt/mTOR signaling pathway in M2 macrophages. Myriocin reduced the population of M2-like TAMs within the tumor tissue of a mouse allograft tumor model but not that of M1-like TAMs. Moreover, combined treatment with myriocin and cisplatin synergistically suppressed tumor growth and enhanced survival rate in mice by reducing the population of M2-like TAMs. Overall, these results suggest that myriocin inhibits tumor growth by remodeling the TME through suppression of differentiation and polarization of M2-like TAMs via the PI3K/Akt/mTOR signaling pathway.
Collapse
Affiliation(s)
- Hyeonha Jang
- Vessel-Organ Interaction Research Center (VOICE, MRC), College of Pharmacy, Kyungpook National University, Daegu, 41566, Republic of Korea
- College of Pharmacy, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Uttam Ojha
- Vessel-Organ Interaction Research Center (VOICE, MRC), College of Pharmacy, Kyungpook National University, Daegu, 41566, Republic of Korea
- College of Pharmacy, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Ji-Hak Jeong
- Vessel-Organ Interaction Research Center (VOICE, MRC), College of Pharmacy, Kyungpook National University, Daegu, 41566, Republic of Korea
- College of Pharmacy, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Keun-Gyu Park
- Vessel-Organ Interaction Research Center (VOICE, MRC), College of Pharmacy, Kyungpook National University, Daegu, 41566, Republic of Korea
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, 41944, Republic of Korea
| | - Shin Yup Lee
- Vessel-Organ Interaction Research Center (VOICE, MRC), College of Pharmacy, Kyungpook National University, Daegu, 41566, Republic of Korea
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, 41944, Republic of Korea
| | - You Mie Lee
- Vessel-Organ Interaction Research Center (VOICE, MRC), College of Pharmacy, Kyungpook National University, Daegu, 41566, Republic of Korea.
- Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, 41566, Republic of Korea.
- College of Pharmacy, Kyungpook National University, Daegu, 41566, Republic of Korea.
| |
Collapse
|
6
|
Chen K, Li Y, Wang B, Yan X, Tao Y, Song W, Xi Z, He K, Xia Q. Patient-derived models facilitate precision medicine in liver cancer by remodeling cell-matrix interaction. Front Immunol 2023; 14:1101324. [PMID: 37215109 PMCID: PMC10192760 DOI: 10.3389/fimmu.2023.1101324] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 04/24/2023] [Indexed: 05/24/2023] Open
Abstract
Liver cancer is an aggressive tumor originating in the liver with a dismal prognosis. Current evidence suggests that liver cancer is the fifth most prevalent cancer worldwide and the second most deadly type of malignancy. Tumor heterogeneity accounts for the differences in drug responses among patients, emphasizing the importance of precision medicine. Patient-derived models of cancer are widely used preclinical models to study precision medicine since they preserve tumor heterogeneity ex vivo in the study of many cancers. Patient-derived models preserving cell-cell and cell-matrix interactions better recapitulate in vivo conditions, including patient-derived xenografts (PDXs), induced pluripotent stem cells (iPSCs), precision-cut liver slices (PCLSs), patient-derived organoids (PDOs), and patient-derived tumor spheroids (PDTSs). In this review, we provide a comprehensive overview of the different modalities used to establish preclinical models for precision medicine in liver cancer.
Collapse
Affiliation(s)
- Kaiwen Chen
- Department of Liver Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Engineering Research Center of Transplantation and Immunology, Shanghai, China
- Shanghai Institute of Transplantation, Shanghai, China
| | - Yanran Li
- Department of Liver Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Engineering Research Center of Transplantation and Immunology, Shanghai, China
- Shanghai Institute of Transplantation, Shanghai, China
| | - Bingran Wang
- Department of Liver Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Engineering Research Center of Transplantation and Immunology, Shanghai, China
- Shanghai Institute of Transplantation, Shanghai, China
| | - Xuehan Yan
- Department of Gastrointestinal Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yiying Tao
- Department of Anesthesiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weizhou Song
- Ottawa-Shanghai Joint School of Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhifeng Xi
- Department of Liver Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Engineering Research Center of Transplantation and Immunology, Shanghai, China
- Shanghai Institute of Transplantation, Shanghai, China
| | - Kang He
- Department of Liver Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Engineering Research Center of Transplantation and Immunology, Shanghai, China
- Shanghai Institute of Transplantation, Shanghai, China
| | - Qiang Xia
- Department of Liver Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Engineering Research Center of Transplantation and Immunology, Shanghai, China
- Shanghai Institute of Transplantation, Shanghai, China
| |
Collapse
|
7
|
Liu Y, Wu W, Cai C, Zhang H, Shen H, Han Y. Patient-derived xenograft models in cancer therapy: technologies and applications. Signal Transduct Target Ther 2023; 8:160. [PMID: 37045827 PMCID: PMC10097874 DOI: 10.1038/s41392-023-01419-2] [Citation(s) in RCA: 134] [Impact Index Per Article: 67.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 03/21/2023] [Indexed: 04/14/2023] Open
Abstract
Patient-derived xenograft (PDX) models, in which tumor tissues from patients are implanted into immunocompromised or humanized mice, have shown superiority in recapitulating the characteristics of cancer, such as the spatial structure of cancer and the intratumor heterogeneity of cancer. Moreover, PDX models retain the genomic features of patients across different stages, subtypes, and diversified treatment backgrounds. Optimized PDX engraftment procedures and modern technologies such as multi-omics and deep learning have enabled a more comprehensive depiction of the PDX molecular landscape and boosted the utilization of PDX models. These irreplaceable advantages make PDX models an ideal choice in cancer treatment studies, such as preclinical trials of novel drugs, validating novel drug combinations, screening drug-sensitive patients, and exploring drug resistance mechanisms. In this review, we gave an overview of the history of PDX models and the process of PDX model establishment. Subsequently, the review presents the strengths and weaknesses of PDX models and highlights the integration of novel technologies in PDX model research. Finally, we delineated the broad application of PDX models in chemotherapy, targeted therapy, immunotherapy, and other novel therapies.
Collapse
Affiliation(s)
- Yihan Liu
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China
| | - Wantao Wu
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China
| | - Changjing Cai
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China
| | - Hao Zhang
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Hong Shen
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China.
| | - Ying Han
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China.
| |
Collapse
|
8
|
Weiss J, Pham NA, Pintilie M, Li M, Liu G, Shepherd FA, Tsao MS, Xu W. Optimizing Drug Response Study Design in Patient-Derived Tumor Xenografts. Cancer Inform 2022; 21:11769351221136056. [PMID: 36439025 PMCID: PMC9685207 DOI: 10.1177/11769351221136056] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 10/14/2022] [Indexed: 05/26/2024] Open
Abstract
Patient-derived tumor xenograft (PDX) models were used to evaluate the effectiveness of preclinical anticancer agents. A design using 1 mouse per patient per drug (1 × 1 × 1) was considered practical for large-scale drug efficacy studies. We evaluated modifiable parameters that could increase the statistical power of this design based on our consolidated PDX experiments. Real studies were used as a reference to investigate the relationship between statistical power with treatment effect size, inter-mouse variation, and tumor measurement frequencies. Our results showed that large effect sizes could be detected at a significance level of .2 or .05 under a 1 × 1 × 1 design. We found that the minimum number of mice required to achieve 80% power at an alpha level of .05 under all situations explored was 21 mice per group for a small effect size and 5 mice per group for a medium effect size.
Collapse
Affiliation(s)
- Jessica Weiss
- Department of Biostatistics, Princess
Margaret Cancer Centre, University Health Network, University of Toronto, Toronto,
ON, Canada
| | - Nhu-An Pham
- Princess Margaret Cancer Centre,
University Health Network, Toronto, ON, Canada
| | - Melania Pintilie
- Department of Biostatistics, Princess
Margaret Cancer Centre, University Health Network, University of Toronto, Toronto,
ON, Canada
| | - Ming Li
- Princess Margaret Cancer Centre,
University Health Network, Toronto, ON, Canada
| | - Geoffrey Liu
- Princess Margaret Cancer Centre,
University Health Network, Toronto, ON, Canada
- Department of Medicine, Division of
Medical Oncology, University of Toronto, Toronto, ON, Canada
- Department of Medical Biophysics,
University of Toronto, Toronto, ON, Canada
| | - Frances A Shepherd
- Princess Margaret Cancer Centre,
University Health Network, Toronto, ON, Canada
- Department of Medicine, Division of
Medical Oncology, University of Toronto, Toronto, ON, Canada
| | - Ming-Sound Tsao
- Princess Margaret Cancer Centre,
University Health Network, Toronto, ON, Canada
- Department of Medical Biophysics,
University of Toronto, Toronto, ON, Canada
- Department of Laboratory Medicine and
Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Wei Xu
- Department of Biostatistics, Princess
Margaret Cancer Centre, University Health Network, University of Toronto, Toronto,
ON, Canada
- Department of Biostatistics, Dalla Lana
School of Public Health, Toronto, ON, Canada
| |
Collapse
|
9
|
Shebl B, Ng D, Lalazar G, Rosemore C, Finkelstein TM, Migler RD, Zheng G, Zhang P, Jiang CS, Qureshi A, Vaughan R, Yarchoan M, de Jong YP, Rice CM, Coffino P, Ortiz MV, Zhou D, Simon SM. Targeting BCL-XL in fibrolamellar hepatocellular carcinoma. JCI Insight 2022; 7:e161820. [PMID: 36073545 PMCID: PMC9536265 DOI: 10.1172/jci.insight.161820] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 08/03/2022] [Indexed: 11/17/2022] Open
Abstract
Fibrolamellar hepatocellular carcinoma (FLC) is a rare and often lethal liver cancer with no proven effective systemic therapy. Inhibition of the antiapoptotic protein BCL-XL was found to synergize with a variety of systemic therapies in vitro using cells dissociated from patient-derived xenografts (PDX) of FLC or cells dissociated directly from surgical patient resections. As BCL-XL is physiologically expressed in platelets, prior efforts to leverage this vulnerability in other cancers have been hampered by severe thrombocytopenia. To overcome this toxicity, we treated FLC models with DT2216, a proteolysis targeting chimera (PROTAC) that directs BCL-XL for degradation via the von Hippel-Lindau (VHL) E3 ligase, which is minimally expressed in platelets. The combination of irinotecan and DT2216 in vitro on cells directly acquired from patients or in vivo using several xenografts derived from patients with FLC demonstrated remarkable synergy and at clinically achievable doses not associated with significant thrombocytopenia.
Collapse
Affiliation(s)
- Bassem Shebl
- Laboratory of Cellular Biophysics, The Rockefeller University, New York, New York, USA
| | - Denise Ng
- Laboratory of Cellular Biophysics, The Rockefeller University, New York, New York, USA
| | - Gadi Lalazar
- Laboratory of Cellular Biophysics, The Rockefeller University, New York, New York, USA
| | - Carly Rosemore
- Laboratory of Cellular Biophysics, The Rockefeller University, New York, New York, USA
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Tova M. Finkelstein
- Laboratory of Cellular Biophysics, The Rockefeller University, New York, New York, USA
| | | | - Guangrong Zheng
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, Florida, USA
| | - Peiyi Zhang
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, Florida, USA
| | - Caroline S. Jiang
- Hospital Biostatistics, The Rockefeller University, New York, New York, USA
| | - Adam Qureshi
- Hospital Biostatistics, The Rockefeller University, New York, New York, USA
| | - Roger Vaughan
- Hospital Biostatistics, The Rockefeller University, New York, New York, USA
| | - Mark Yarchoan
- The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Ype P. de Jong
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, New York, USA
- Division of Gastroenterology and Hepatology, Weill Cornell Medicine, New York, New York, USA
| | - Charles M. Rice
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, New York, USA
| | - Philip Coffino
- Laboratory of Cellular Biophysics, The Rockefeller University, New York, New York, USA
| | - Michael V. Ortiz
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Daohong Zhou
- Department of Biochemistry and Structural Biology and Center for Innovative Drug Discovery, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Sanford M. Simon
- Laboratory of Cellular Biophysics, The Rockefeller University, New York, New York, USA
| |
Collapse
|
10
|
Sahib AS, Wennas ON, Mahdi BW, Al abood RM. In vivo antitumor activity study of targeted chlorambucil-loaded nanolipid carrier for breast cancer. PHARMACIA 2022. [DOI: 10.3897/pharmacia.69.e85390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Chlorambucil (CBL) is an efficient anticancer drug. It is a lipophilic agent with serious adverse effects. The objective of this study was to formulate a CBL-loaded nanolipid carrier and target breast cancer using folic acid as a targeting probe. Characterizations of the optimum formulation were 79.9±3% EE after the addition of 4mg CBL, 119±6nm particle size which is considered appropriate for parenteral use, 0.3±0.02 PDI, -42±1mV ZP that stabilized the formulation. Tumor volume, body weight, and tumor mass weight were recorded to evaluate tumor volume doubling time, tumor growth inhibition rate, and systemic toxicity. It appeared there was a significant antitumor activity of targeted formulation compared with non-targeted one and free CBL. Moreover, the systemic toxicity was less after body weight evaluation concerning the targeted formulation when compared with other formulations.
Collapse
|
11
|
Yang S, Yao Y, Dong Y, Liu J, Li Y, Yi L, Huang Y, Gao Y, Yin J, Li Q, Ye D, Gong H, Xu B, Li J, Song Q. Prediction of Radiation Pneumonitis Using Genome-Scale Flux Analysis of RNA-Seq Derived From Peripheral Blood. Front Med (Lausanne) 2021; 8:715961. [PMID: 34532331 PMCID: PMC8438228 DOI: 10.3389/fmed.2021.715961] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 07/30/2021] [Indexed: 01/09/2023] Open
Abstract
Purpose: Radiation pneumonitis (RP) frequently occurs during a treatment course of chest radiotherapy, which significantly reduces the clinical outcome and efficacy of radiotherapy. The ability to easily predict RP before radiotherapy would allow this disease to be avoided. Methods and Materials: This study recruited 48 lung cancer patients requiring chest radiotherapy. For each participant, RNA sequencing (RNA-Seq) was performed on a peripheral blood sample before radiotherapy. The RNA-Seq data was then integrated into a genome-scale flux analysis to develop an RP scoring system for predicting the probability of occurrence of RP. Meanwhile, the clinical information and radiation dosimetric parameters of this cohort were collected for analysis of any statistical associations between these parameters and RP. A non-parametric rank sum test showed no significant difference between the predicted results from the RP score system and the clinically observed occurrence of RP in this cohort. Results: The results of the univariant analysis suggested that the tumor stage, exposure dose, and bilateral lung dose of V5 and V20 were significantly associated with the occurrence of RP. The results of the multivariant analysis suggested that the exposure doses of V5 and V20 were independent risk factors associated with RP and a level of RP ≥ 2, respectively. Thus, our results indicate that our RP scoring system could be applied to accurately predict the risk of RP before radiotherapy because the scores were highly consistent with the clinically observed occurrence of RP. Conclusion: Compared with the standard statistical methods, this genome-scale flux-based scoring system is more accurate, straightforward, and economical, and could therefore be of great significance when making clinical decisions for chest radiotherapy.
Collapse
Affiliation(s)
- Siqi Yang
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yi Yao
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China.,Hubei Provincial Research Center for Precision Medicine of Cancer, Wuhan, China
| | - Yi Dong
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Junqi Liu
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yingge Li
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Lina Yi
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yani Huang
- Oncology Department, Zhongxiang Hospital, Renmin Hospital of Wuhan University, Zhongxiang, China
| | - Yanjun Gao
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Junping Yin
- Institute of Experimental Immunology, University Clinic of Rheinische Friedrich-Wilhelms-University, Bonn, Germany
| | - Qingqing Li
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Dafu Ye
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Hongyun Gong
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Bin Xu
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jian Li
- Institute of Experimental Immunology, University Clinic of Rheinische Friedrich-Wilhelms-University, Bonn, Germany
| | - Qibin Song
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China.,Hubei Provincial Research Center for Precision Medicine of Cancer, Wuhan, China
| |
Collapse
|
12
|
A Novel Monoclonal Antibody Targeting Cancer-Specific Plectin Has Potent Antitumor Activity in Ovarian Cancer. Cells 2021; 10:cells10092218. [PMID: 34571866 PMCID: PMC8466582 DOI: 10.3390/cells10092218] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 08/19/2021] [Accepted: 08/24/2021] [Indexed: 01/25/2023] Open
Abstract
Cancer-specific plectin (CSP) is a pro-tumorigenic protein selectively expressed on the cell surface of major cancers, including ovarian cancer (OC). Despite its assessable localization, abundance, and functional significance, the therapeutic efficacy of targeting CSP remains unexplored. Here, we generated and investigated the anticancer effects of a novel CSP-targeting monoclonal antibody, 1H11, in OC models. Its therapeutic efficacy as a monotherapy and in combination with chemotherapy was evaluated in vitro using two OC cell lines and in vivo by a subcutaneous ovarian cancer model. 1H11 demonstrated rapid internalization and high affinity and specificity for both human and murine CSP. Moreover, 1H11 induced significant and selective cytotoxicity (EC50 = 260 nM), G0/G1 arrest, and decreased OC cell migration. Mechanistically, these results are associated with increased ROS levels and reduced activation of the JAK2-STAT3 pathway. In vivo, 1H11 decreased Ki67 expression, induced 65% tumor growth inhibition, and resulted in 30% tumor necrosis. Moreover, 1H11 increased chemosensitivity to cisplatin resulting in 60% greater tumor growth inhibition compared to cisplatin alone. Taken together, CSP-targeting with 1H11 exhibits potent anticancer activity against ovarian cancer and is deserving of future clinical development.
Collapse
|
13
|
A reliable method to determine which candidate chemotherapeutic drugs effectively inhibit tumor growth in patient-derived xenografts (PDX) in single mouse trials. Cancer Chemother Pharmacol 2019; 84:1167-1178. [PMID: 31512030 DOI: 10.1007/s00280-019-03942-y] [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: 05/02/2019] [Accepted: 08/24/2019] [Indexed: 10/26/2022]
Abstract
PURPOSE We report on a statistical method for grouping anti-cancer drugs (GRAD) in single mouse trials (SMT). The method assigns candidate drugs into groups that inhibit or do not inhibit tumor growth in patient-derived xenografts (PDX). It determines the statistical significance of the group assignments without replicate trials of each drug. METHODS The GRAD method applies a longitudinal finite mixture model, implemented in the statistical package PROC TRAJ, to analyze a mixture of tumor growth curves for portions of the same tumor in different mice, each single mouse exposed to a different drug. Each drug is classified into an inhibitory or non-inhibitory group. There are several advantages to the GRAD method for SMT. It determines that probability that the grouping is correct, uses the entire longitudinal tumor growth curve data for each drug treatment, can fit different shape growth curves, accounts for missing growth curve data, and accommodates growth curves of different time periods. RESULTS We analyzed data for 22 drugs for 18 human colorectal tumors provided by researchers in a previous publication. The GRAD method identified 18 drugs that were inhibitory against at least one tumor, and 10 tumors for which there was at least one inhibitory drug. Analysis of simulated data indicated that the GRAD method has a sensitivity of 84% and a specificity of 98%. CONCLUSION A statistical method, GRAD, can group anti-cancer drugs into those that are inhibitory and those that are non-inhibitory in single mouse trials and provide probabilities that the grouping is correct.
Collapse
|