1
|
Villette CC, Dupuy N, Brightman FA, Zimmermann A, Lignet F, Zenke FT, Terranova N, Bolleddula J, El Bawab S, Chassagnole C. Semi-mechanistic efficacy model for PARP + ATR inhibitors-application to rucaparib and talazoparib in combination with gartisertib in breast cancer PDXs. Br J Cancer 2025:10.1038/s41416-024-02935-w. [PMID: 39875558 DOI: 10.1038/s41416-024-02935-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 12/04/2024] [Accepted: 12/17/2024] [Indexed: 01/30/2025] Open
Abstract
BACKGROUND Promising cancer treatments, such as DDR inhibitors, are often challenged by the heterogeneity of responses in clinical trials. The present work aimed to build a computational framework to address those challenges. METHODS A semi-mechanistic pharmacokinetic-pharmacodynamic model of tumour growth inhibition was developed to investigate the efficacy of PARP and ATR inhibitors as monotherapies, and in combination. Key features of the DNA damage response were incorporated into the model to allow the emergence of synthetic lethality, including redundant DNA repair pathways that may be impaired due to genetic mutations, and due to PARP and ATR inhibition. Model parameters were calibrated using preclinical in vivo data for PARP inhibitors rucaparib and talazoparib and the ATR inhibitor gartisertib. RESULTS The model successfully captured the monotherapy efficacies of rucaparib and talazoparib, as well as the combination efficacy with gartisertib. The model was evaluated against multiple tumour xenografts with diverse genetic backgrounds and was able to capture the observed heterogeneity of response profiles. CONCLUSIONS By enabling simulation of in vivo tumour growth inhibition with PARP and ATR inhibitors for specific tumour types, the model provides a rational approach to support the optimisation of dosing regimens to stratified populations.
Collapse
Affiliation(s)
| | | | | | | | | | - Frank T Zenke
- EMD Serono Research & Development Institute, Inc., Billerica, MA, USA
| | - Nadia Terranova
- Quantitative Pharmacology, Ares Trading S.A. (An Affiliate of Merck KGaA, Darmstadt, Germany), Lausanne, Switzerland
| | | | | | | |
Collapse
|
2
|
Lin X, Soni A, Hessenow R, Sun Y, Mladenov E, Guberina M, Stuschke M, Iliakis G. Talazoparib enhances resection at DSBs and renders HR-proficient cancer cells susceptible to Polθ inhibition. Radiother Oncol 2024; 200:110475. [PMID: 39147034 DOI: 10.1016/j.radonc.2024.110475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 07/02/2024] [Accepted: 08/12/2024] [Indexed: 08/17/2024]
Abstract
BACKGROUND AND PURPOSE The PARP inhibitor (PARPi), Talazoparib (BMN673), effectively and specifically radiosensitizes cancer cells. Radiosensitization is mediated by a shift in the repair of ionizing radiation (IR)-induced DNA double-strand breaks (DSBs) toward PARP1-independent, alternative end-joining (alt-EJ). DNA polymerase theta (Polθ) is a key component of this PARP1-independent alt-EJ pathway and we show here that its inhibition can further radiosensitize talazoparib-treated cells. The purpose of the present work is to explore mechanisms and dynamics underpinning enhanced talazoparib radiosensitization by Polθ inhibitors in HR-proficient cancer cells. METHODS AND MATERIALS Radiosensitization to PARPis, talazoparib, olaparib, rucaparib and veliparib was assessed by clonogenic survival. Polθ-proficient and -deficient cells were treated with PARPis and/or with the Polθ inhibitors ART558 or novobiocin. The role of DNA end-resection was studied by down-regulating CtIP and MRE11 expression using siRNAs. DSB repair was assessed by scoring γH2AX foci. The formation of chromosomal abnormalities was assessed as evidence of alt-EJ function using G2-specific cytogenetic analysis. RESULTS Talazoparib exerted pronounced radiosensitization that varied among the tested cancer cell lines; however, radiosensitization was undetectable in normal cells. Other commonly used PARPis, olaparib, veliparib, or rucaparib were ineffective radiosensitizers under our experimental conditions. Although genetic ablation or pharmacological inhibition of Polθ only mildly radiosensitized cancer cells, talazoparib-treated cells were markedly further radiosensitized. Mechanistically, talazoparib shunted DSBs to Polθ-dependent alt-EJ by enhancing DNA end-resection in a CtIP- and MRE11-dependent manner - an effect detectable at low, but not high IR doses. Chromosomal translocation analysis in talazoparib-treated cells exposed to Polθ inhibitors suggested that PARP1- and Polθ-dependent alt-EJ pathways may complement, but also back up each other. CONCLUSION We propose that talazoparib promotes low-dose, CtIP/MRE11-dependent resection and increases the reliance of irradiated HR-proficient cancer cells, on Polθ-mediated alt-EJ. The combination of Polθ inhibitors with talazoparib suppresses this option and causes further radiosensitization. The results suggest that Polθ inhibition may be exploited to maximize talazoparib radiosensitization of HR-proficient tumors in the clinic.
Collapse
Affiliation(s)
- Xixi Lin
- Division of Experimental Radiation Biology, Department of Radiation Therapy, University Hospital Essen, University of Duisburg-Essen, 45147, Essen, Germany; Institute of Medical Radiation Biology, University Hospital Essen, University of Duisburg-Essen, 45147, Essen, Germany
| | - Aashish Soni
- Division of Experimental Radiation Biology, Department of Radiation Therapy, University Hospital Essen, University of Duisburg-Essen, 45147, Essen, Germany; Institute of Medical Radiation Biology, University Hospital Essen, University of Duisburg-Essen, 45147, Essen, Germany.
| | - Razan Hessenow
- West German Proton Therapy Center Essen (WPE), University of Duisburg-Essen, 45147, Essen, Germany
| | - Yanjie Sun
- Institute of Medical Radiation Biology, University Hospital Essen, University of Duisburg-Essen, 45147, Essen, Germany; West German Proton Therapy Center Essen (WPE), University of Duisburg-Essen, 45147, Essen, Germany
| | - Emil Mladenov
- Division of Experimental Radiation Biology, Department of Radiation Therapy, University Hospital Essen, University of Duisburg-Essen, 45147, Essen, Germany; Institute of Medical Radiation Biology, University Hospital Essen, University of Duisburg-Essen, 45147, Essen, Germany
| | - Maja Guberina
- Department of Radiation Therapy, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, 45147, Essen, Germany; German Cancer Consortium (DKTK), Partner Site University Hospital Essen, German Cancer Research Center (DKFZ), 45147, Essen, Germany
| | - Martin Stuschke
- Division of Experimental Radiation Biology, Department of Radiation Therapy, University Hospital Essen, University of Duisburg-Essen, 45147, Essen, Germany; Department of Radiation Therapy, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, 45147, Essen, Germany; German Cancer Consortium (DKTK), Partner Site University Hospital Essen, German Cancer Research Center (DKFZ), 45147, Essen, Germany
| | - George Iliakis
- Division of Experimental Radiation Biology, Department of Radiation Therapy, University Hospital Essen, University of Duisburg-Essen, 45147, Essen, Germany; Institute of Medical Radiation Biology, University Hospital Essen, University of Duisburg-Essen, 45147, Essen, Germany.
| |
Collapse
|
3
|
Schreiber T, Koopmann I, Brandstetter J, Talbot SR, Goldstein L, Hoffmann L, Schildt A, Joksch M, Krause B, Jaster R, Palme R, Zechner D, Vollmar B, Kumstel S. Evidence-Based Severity Assessment of Animal Models for Pancreatic Cancer. Biomedicines 2024; 12:1494. [PMID: 39062067 PMCID: PMC11275077 DOI: 10.3390/biomedicines12071494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 06/24/2024] [Accepted: 06/27/2024] [Indexed: 07/28/2024] Open
Abstract
Animal models are crucial to preclinical oncological research and drug development. Animal experiments must be performed in accordance with the 3R principles of replacement and reduction, if possible, and refinement where these procedures remain crucial. In addition, European Union legislations demand a continuous refinement approach, as well as pro- and retrospective severity assessment. In this study, an objective databased severity assessment was performed in murine models for pancreatic cancer induced by orthotopic, subcutaneous, or intravenous injection of Panc02 cells. Parameters such as body weight change, distress score, perianal temperature, mouse grimace scale, burrowing, nesting behavior, and the concentration of corticosterone in plasma and its metabolites in feces were monitored during tumor progression. The most important parameters were combined into a score and mapped against a reference data set by the Relative Severity Assessment procedure (RELSA) to obtain the maximum achieved severity for each animal (RELSAmax). This scoring revealed a significantly higher RELSAmax for the orthotopic model than for the subcutaneous and intravenous models. However, compared to animal models such as pancreatitis and bile duct ligation, the pancreatic cancer models are shown to be less severe. Data-based animal welfare assessment proved to be a valuable tool for comparing the severity of differently induced cancer models.
Collapse
Affiliation(s)
- Tim Schreiber
- Rudolf-Zenker-Institute of Experimental Surgery, Rostock University Medical Center, 18057 Rostock, Germany; (T.S.); (I.K.); (J.B.); (L.G.); (L.H.); (D.Z.); (B.V.)
| | - Ingo Koopmann
- Rudolf-Zenker-Institute of Experimental Surgery, Rostock University Medical Center, 18057 Rostock, Germany; (T.S.); (I.K.); (J.B.); (L.G.); (L.H.); (D.Z.); (B.V.)
| | - Jakob Brandstetter
- Rudolf-Zenker-Institute of Experimental Surgery, Rostock University Medical Center, 18057 Rostock, Germany; (T.S.); (I.K.); (J.B.); (L.G.); (L.H.); (D.Z.); (B.V.)
| | - Steven R. Talbot
- Institute for Laboratory Animal Science, Preclinical Data Science, Hannover Medical School, 30625 Hannover, Germany;
| | - Lea Goldstein
- Rudolf-Zenker-Institute of Experimental Surgery, Rostock University Medical Center, 18057 Rostock, Germany; (T.S.); (I.K.); (J.B.); (L.G.); (L.H.); (D.Z.); (B.V.)
| | - Lisa Hoffmann
- Rudolf-Zenker-Institute of Experimental Surgery, Rostock University Medical Center, 18057 Rostock, Germany; (T.S.); (I.K.); (J.B.); (L.G.); (L.H.); (D.Z.); (B.V.)
| | - Anna Schildt
- Core Facility Multimodal Small Animal Imaging, Rostock University Medical Center, 18057 Rostock, Germany;
| | - Markus Joksch
- Department of Nuclear Medicine, Rostock University Medical Center, 18057 Rostock, Germany; (M.J.); (B.K.)
| | - Bernd Krause
- Department of Nuclear Medicine, Rostock University Medical Center, 18057 Rostock, Germany; (M.J.); (B.K.)
| | - Robert Jaster
- Division of Gastroenterology, Department of Medicine II, Rostock University Medical Center, 18057 Rostock, Germany;
| | - Rupert Palme
- Unit of Experimental Endocrinology, Department of Biological Sciences and Pathobiology, University of Veterinary Medicine, 1210 Vienna, Austria;
| | - Dietmar Zechner
- Rudolf-Zenker-Institute of Experimental Surgery, Rostock University Medical Center, 18057 Rostock, Germany; (T.S.); (I.K.); (J.B.); (L.G.); (L.H.); (D.Z.); (B.V.)
| | - Brigitte Vollmar
- Rudolf-Zenker-Institute of Experimental Surgery, Rostock University Medical Center, 18057 Rostock, Germany; (T.S.); (I.K.); (J.B.); (L.G.); (L.H.); (D.Z.); (B.V.)
| | - Simone Kumstel
- Rudolf-Zenker-Institute of Experimental Surgery, Rostock University Medical Center, 18057 Rostock, Germany; (T.S.); (I.K.); (J.B.); (L.G.); (L.H.); (D.Z.); (B.V.)
| |
Collapse
|
4
|
Watterson A, Coelho MA. Cancer immune evasion through KRAS and PD-L1 and potential therapeutic interventions. Cell Commun Signal 2023; 21:45. [PMID: 36864508 PMCID: PMC9979509 DOI: 10.1186/s12964-023-01063-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 01/31/2023] [Indexed: 03/04/2023] Open
Abstract
Oncogenic driver mutations have implications that extend beyond cancer cells themselves. Aberrant tumour cell signalling has various effects on the tumour microenvironment and anti-tumour immunity, with important consequences for therapy response and resistance. We provide an overview of how mutant RAS, one of the most prevalent oncogenic drivers in cancer, can instigate immune evasion programs at the tumour cell level and through remodelling interactions with the innate and adaptive immune cell compartments. Finally, we describe how immune evasion networks focused on RAS, and the immune checkpoint molecule PD-L1 can be disrupted through therapeutic intervention, and discuss potential strategies for combinatorial treatment. Video abstract.
Collapse
Affiliation(s)
- Alex Watterson
- Translational Cancer Genomics, Wellcome Sanger Institute, Hinxton, UK.,Open Targets, Cambridge, UK
| | - Matthew A Coelho
- Translational Cancer Genomics, Wellcome Sanger Institute, Hinxton, UK. .,Open Targets, Cambridge, UK.
| |
Collapse
|
5
|
De Thoré MG, Meziani L, Deutsch E, Mondini M. Cytofluorometric characterization of the myeloid compartment of irradiated mouse tumors. Methods Cell Biol 2023; 174:17-30. [PMID: 36710048 DOI: 10.1016/bs.mcb.2022.08.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
The use of ionizing radiation (IR) is a cornerstone for the treatment of cancer and radiotherapy (RT) is used in roughly 50% of cancer patients. It is now well established that RT exerts widespread effects on the tumor stroma, including the immune environment. Together with its deeply characterized effects on the lymphoid compartment, RT also deeply affects the myeloid cell compartment. Fluorescence-activated flow cytometry is one of the most widely used technologies in immunology, allowing the multiparametric analysis of cells on a cell-by-cell basis. Here, we provide a detailed flow cytometry protocol to analyze the myeloid cell populations of human papillomavirus (HPV)-positive TC1/Luc tumors engrafted in the oral mucosa of immunocompetent mice, and to evaluate their modulations in response to RT. The same method, with slight modifications, can be used to study the tumor myeloid cells from a variety of other mouse tumors.
Collapse
Affiliation(s)
| | - Lydia Meziani
- Gustave Roussy, INSERM U1030, Université Paris-Saclay, Villejuif, France
| | - Eric Deutsch
- Gustave Roussy, INSERM U1030, Université Paris-Saclay, Villejuif, France.
| | - Michele Mondini
- Gustave Roussy, INSERM U1030, Université Paris-Saclay, Villejuif, France.
| |
Collapse
|
6
|
Milic M, Mondini M, Deutsch E. How to Improve SBRT Outcomes in NSCLC: From Pre-Clinical Modeling to Successful Clinical Translation. Cancers (Basel) 2022; 14:cancers14071705. [PMID: 35406477 PMCID: PMC8997119 DOI: 10.3390/cancers14071705] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 03/14/2022] [Accepted: 03/22/2022] [Indexed: 02/01/2023] Open
Abstract
Simple Summary Despite major research and clinical efforts, lung cancer remains the leading cause of cancer-related death. Stereotactic body radiotherapy (SBRT) has emerged as a major treatment modality for lung cancer in the last decade. Additional research is needed to elucidate underlying mechanisms of resistance and to develop improved therapeutic strategies. Clinical progress relies on accurate preclinical modelling of human disease in order to yield clinically meaningful results; however, successful translation of pre-clinical research is still lagging behind. In this review, we summarize the major clinical developments of radiation therapy for non-small-cell lung cancer (NSCLC), and we discuss the pre-clinical research models at our disposal, highlighting ongoing translational challenges and future perspectives. Abstract Despite major research and clinical efforts, lung cancer remains the leading cause of cancer-related death. While the delivery of conformal radiotherapy and image guidance of stereotactic body radiotherapy (SBRT) have revolutionized the treatment of early-stage non-small-cell lung cancer (NSCLC), additional research is needed to elucidate underlying mechanisms of resistance and identify novel therapeutic combinations. Clinical progress relies on the successful translation of pre-clinical work, which so far has not always yielded expected results. Improved clinical modelling involves characterizing the preclinical models and selecting appropriate experimental designs that faithfully mimic precise clinical scenarios. Here, we review the current role of SBRT and the scope of pre-clinical armamentarium at our disposal to improve successful clinical translation of pre-clinical research in the radiation oncology of NSCLC.
Collapse
Affiliation(s)
- Marina Milic
- Gustave Roussy, Université Paris-Saclay, INSERM U1030, F-94805 Villejuif, France;
| | - Michele Mondini
- Gustave Roussy, Université Paris-Saclay, INSERM U1030, F-94805 Villejuif, France;
- Correspondence: (M.M.); (E.D.)
| | - Eric Deutsch
- Gustave Roussy, Université Paris-Saclay, INSERM U1030, F-94805 Villejuif, France;
- Gustave Roussy, Département d’Oncologie-Radiothérapie, F-94805 Villejuif, France
- Correspondence: (M.M.); (E.D.)
| |
Collapse
|
7
|
Hamon P, Gerbé De Thoré M, Classe M, Signolle N, Liu W, Bawa O, Meziani L, Clémenson C, Milliat F, Deutsch E, Mondini M. TGFβ receptor inhibition unleashes interferon-β production by tumor-associated macrophages and enhances radiotherapy efficacy. J Immunother Cancer 2022; 10:jitc-2021-003519. [PMID: 35301235 PMCID: PMC8932273 DOI: 10.1136/jitc-2021-003519] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/25/2022] [Indexed: 01/18/2023] Open
Abstract
Background Transforming growth factor-beta (TGFβ) can limit the efficacy of cancer treatments, including radiotherapy (RT), by inducing an immunosuppressive tumor environment. The association of TGFβ with impaired T cell infiltration and antitumor immunity is known, but the mechanisms by which TGFβ participates in immune cell exclusion and limits the efficacy of antitumor therapies warrant further investigations. Methods We used the clinically relevant TGFβ receptor 2 (TGFβR2)-neutralizing antibody MT1 and the small molecule TGFβR1 inhibitor LY3200882 and evaluated their efficacy in combination with RT against murine orthotopic models of head and neck and lung cancer. Results We demonstrated that TGFβ pathway inhibition strongly increased the efficacy of RT. TGFβR2 antibody upregulated interferon beta expression in tumor-associated macrophages within the irradiated tumors and favored T cell infiltration at the periphery and within the core of the tumor lesions. We highlighted that both the antitumor efficacy and the increased lymphocyte infiltration observed with the combination of MT1 and RT were dependent on type I interferon signaling. Conclusions These data shed new light on the role of TGFβ in limiting the efficacy of RT, identifying a novel mechanism involving the inhibition of macrophage-derived type I interferon production, and fostering the use of TGFβR inhibition in combination with RT in therapeutic strategies for the management of head and neck and lung cancer.
Collapse
Affiliation(s)
- Pauline Hamon
- INSERM U1030, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | | | - Marion Classe
- INSERM U1030, Gustave Roussy, Université Paris-Saclay, Villejuif, France.,Départment de Pathologie, Gustave Roussy, Villejuif, France
| | - Nicolas Signolle
- Plateforme de pathologie expérimentale et translationnelle, UMS AMMICA, Gustave Roussy, Villejuif, France
| | - Winchygn Liu
- INSERM U1030, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Olivia Bawa
- Départment de Pathologie, Gustave Roussy, Villejuif, France
| | - Lydia Meziani
- INSERM U1030, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Céline Clémenson
- INSERM U1030, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Fabien Milliat
- Department of RAdiobiology and Regenerative MEDicine (SERAMED), Laboratory of MEDical Radiobiology (LRMed), Institute for Radiological Protection and Nuclear Safety (IRSN), Fontenay-aux-Roses, France
| | - Eric Deutsch
- INSERM U1030, Gustave Roussy, Université Paris-Saclay, Villejuif, France .,Département d'Oncologie-Radiothérapie, Gustave Roussy, Villejuif, France
| | - Michele Mondini
- INSERM U1030, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| |
Collapse
|
8
|
Wan Q, Bao Y, Xia X, Liu J, Wang P, Peng Y, Xie X, He J, Li X. Intravoxel Incoherent Motion Diffusion-Weighted Imaging for Predicting and Monitoring the Response of Anti-Angiogenic Treatment in the Orthotopic Nude Mouse Model of Lung Adenocarcinoma. J Magn Reson Imaging 2021; 55:1202-1210. [PMID: 34570394 DOI: 10.1002/jmri.27920] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 08/30/2021] [Accepted: 08/31/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND The treatment efficacy of angiogenesis inhibitor could be underestimated at an early stage based on tumor volume changes. Intravoxel incoherent motion diffusion-weighted imaging (IVIM-DWI) can quantitatively assess tumors at the cellular level, but it is unclear whether it can provide useful information for assessing treatment response of anti-angiogenic treatment for lung adenocarcinoma. PURPOSE To determine the use of IVIM-DWI for non-invasive monitoring of the early response to anti-angiogenic treatment in the orthotopic transplantation of lung adenocarcinoma model. STUDY TYPE Prospective. POPULATION Thirty-seven nude mice were randomized into two groups: treatment group (received bevacizumab + cisplatin, N = 20) and control group (received saline, N = 17). FIELD STRENGTH/SEQUENCE Single-shot turbo spin-echo (TSE) IVIM-DWI, TSE T2-weighted imaging at 3.0 T. ASSESSMENT Tumor volume, IVIM parameters (apparent diffusion coefficient [ADC], diffusivity [D], perfusion fraction [f], and pseudo-diffusion coefficient [D*]) were measured before and 2 hours, 3, 7, 10 and 14 days after treatment. Regions of interest were manually drawn along the inner edge of the tumor by two radiologists with 5 and 10-year experience in magnetic resonance imaging. Pathological examinations (hematoxylin and eosin stain, cluster of differentiation 34) were performed. STATISTICAL TESTS Kolmogorov-Smirnov test, repeated-measure two-way analysis of variance test, Mann-Whitney U test, Pearson correlation analysis, receiver operating characteristic curve. P < 0.05 was considered statistically significant. RESULTS The tumor volume of the two groups was significantly different only on day 14 (control group vs. treatment group, 43.15 ± 18.28 mm3 vs. 28.41 ± 1.71 mm3 ). ADC2h , ADC10d , D2h , D7d , D10d , and D14d were significantly higher, while f10d and f14d were significantly lower in the treatment group compared to those of the control group. Both the △ADC2h (r = -0.631) and △D2h (r = -0.700) showed moderate correlations with the relative tumor volume on day 14. DATA CONCLUSION IVIM has the potential to predict and monitor the early response to anti-angiogenic treatment, earlier than size changes, for lung adenocarcinoma. LEVEL OF EVIDENCE 2 TECHNICAL EFFICACY: Stage 4.
Collapse
Affiliation(s)
- Qi Wan
- Department of Radiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yingying Bao
- Department of Radiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xiaoying Xia
- Department of Radiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jieqiong Liu
- Department of Radiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Peng Wang
- Department of Radiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yu Peng
- Department of Radiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xiaobin Xie
- Department of Radiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jianxing He
- Department of Thoracic Oncology and Surgery, China State Key Laboratory of Respiratory Disease and National Clinical Research Center for Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xinchun Li
- Department of Radiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
9
|
Wu NF, Yamamoto J, Bouvet M, Hoffman RM. A Novel Procedure for Orthotopic Tibia Implantation for Establishment of a More Clinical Osteosarcoma PDOX Mouse Model. In Vivo 2021; 35:105-109. [PMID: 33402455 DOI: 10.21873/invivo.12237] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 11/21/2020] [Accepted: 11/25/2020] [Indexed: 12/18/2022]
Abstract
BACKGROUND/AIM Osteosarcoma is a rare type of malignancy that affects mostly children and adolescents. A new procedure was designed to create an improved patient-derived orthotopic xenograft (PDOX) mouse model of osteosarcoma that more closely mimics osteosarcoma in clinical settings. Previous osteosarcoma PDOX models involved implanting a tumor fragment near the femur of nude mice in a space created by separating muscle. MATERIALS AND METHODS A hole was created in the tibia of nude mice and an osteosarcoma tumor fragment was implanted directly into the bone. RESULTS This procedure resulted in tumor growth in the bone similar to osteosarcoma tumors found in clinical patients. CONCLUSION The establishment ratio for this procedure is 80% making it a practical and clinically-relevant model for screening effective therapies for osteosarcoma patients.
Collapse
Affiliation(s)
- Nathaniel F Wu
- AntiCancer Inc, San Diego, CA, U.S.A.,Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, U.S.A
| | - Jun Yamamoto
- AntiCancer Inc, San Diego, CA, U.S.A.,Department of Surgery, University of California, San Diego, CA, U.S.A
| | - Michael Bouvet
- Department of Surgery, University of California, San Diego, CA, U.S.A
| | - Robert M Hoffman
- AntiCancer Inc, San Diego, CA, U.S.A.; .,Department of Surgery, University of California, San Diego, CA, U.S.A
| |
Collapse
|
10
|
Hollandsworth HM, Nishino H, Turner M, Amirfakhri S, Filemoni F, Hoffman RM, Yazaki PJ, Bouvet M. Humanized Fluorescent Tumor-associated Glycoprotein-72 Antibody Selectively Labels Colon-cancer Liver Metastases in Orthotopic Mouse Models. In Vivo 2020; 34:2303-2307. [PMID: 32871754 PMCID: PMC7652487 DOI: 10.21873/invivo.12042] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 06/07/2020] [Accepted: 06/11/2020] [Indexed: 11/10/2022]
Abstract
BACKGROUND/AIM Fluorescence imaging has been shown to improve intra-operative detection of liver metastasis. The present study aimed to determine whether humanized anti-TAG-72 antibody (huCC49) conjugated to a near-infrared dye provides selective labeling of colorectal-cancer liver metastasis in orthotopic mouse models. MATERIALS AND METHODS Humanized anti-TAG-72 (huCC49) was conjugated to IRDye800CW (huCC49-IR800). Orthotopic liver-metastasis nude-mouse models (n=5) were established with the human colon-cancer LS174T cell-line. Three weeks later, mice were administered huCC49-IR800 and intra-vital imaging was performed 48 h later. The mean tumor-to-liver ratio (TLR) was calculated. RESULTS Intra-vital imaging demonstrated clear tumor margins with minimal liver fluorescence 48 h after administration of 50 μg huCC49-IR800 with mean TLR=7.53 (SD±2.76). CONCLUSION Anti-TAG-72 monoclonal antibody conjugated to IRDye800 provides distinct and bright labeling of colorectal tumors in orthotopic nude-mouse models of liver metastasis. TAG-72 may be a useful target for intra-operative imaging of colorectal cancer liver metastasis in the clinic.
Collapse
Affiliation(s)
- Hannah M Hollandsworth
- Department of Surgery, University of California San Diego, San Diego, CA, U.S.A.,Moores Cancer Center, University of California San Diego, San Diego, CA, U.S.A.,Department of Surgery, VA San Diego Healthcare System, San Diego, CA, U.S.A
| | - Hiroto Nishino
- Department of Surgery, University of California San Diego, San Diego, CA, U.S.A.,AntiCancer, Inc., San Diego, CA, U.S.A
| | - Michael Turner
- Department of Surgery, University of California San Diego, San Diego, CA, U.S.A.,Moores Cancer Center, University of California San Diego, San Diego, CA, U.S.A
| | - Siamak Amirfakhri
- Department of Surgery, University of California San Diego, San Diego, CA, U.S.A.,Moores Cancer Center, University of California San Diego, San Diego, CA, U.S.A.,Department of Surgery, VA San Diego Healthcare System, San Diego, CA, U.S.A
| | - Filemoni Filemoni
- Department of Surgery, University of California San Diego, San Diego, CA, U.S.A.,Moores Cancer Center, University of California San Diego, San Diego, CA, U.S.A.,Department of Surgery, VA San Diego Healthcare System, San Diego, CA, U.S.A
| | - Robert M Hoffman
- Department of Surgery, University of California San Diego, San Diego, CA, U.S.A.,Moores Cancer Center, University of California San Diego, San Diego, CA, U.S.A.,Department of Surgery, VA San Diego Healthcare System, San Diego, CA, U.S.A.,AntiCancer, Inc., San Diego, CA, U.S.A
| | - Paul J Yazaki
- Department of Molecular Imaging and Therapy, Beckman Research Institute of the City of Hope, Duarte, CA, U.S.A
| | - Michael Bouvet
- Department of Surgery, University of California San Diego, San Diego, CA, U.S.A. .,Department of Surgery, VA San Diego Healthcare System, San Diego, CA, U.S.A.,Department of Surgery, VA San Diego Healthcare System, San Diego, CA, U.S.A
| |
Collapse
|
11
|
Guffanti F, Alvisi MF, Caiola E, Ricci F, De Maglie M, Soldati S, Ganzinelli M, Decio A, Giavazzi R, Rulli E, Damia G. Impact of ERCC1, XPF and DNA Polymerase β Expression on Platinum Response in Patient-Derived Ovarian Cancer Xenografts. Cancers (Basel) 2020; 12:cancers12092398. [PMID: 32847049 PMCID: PMC7564949 DOI: 10.3390/cancers12092398] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 08/12/2020] [Accepted: 08/20/2020] [Indexed: 12/17/2022] Open
Abstract
Platinum resistance is an unmet medical need in ovarian carcinoma. Molecular biomarkers to predict the response to platinum-based therapy could allow patient stratification and alternative therapeutic strategies early in clinical management. Sensitivity and resistance to platinum therapy are partially determined by the tumor’s intrinsic DNA repair activities, including nucleotide excision repair (NER) and base excision repair (BER). We investigated the role of the NER proteins—ERCC1, XPF, ERCC1/XPF complex—and of the BER protein DNA polymerase β, as possible biomarkers of cisplatin (DDP) response in a platform of recently established patient-derived ovarian carcinoma xenografts (OC-PDXs). ERCC1 and DNA polymerase β protein expressions were measured by immunohistochemistry, the ERCC1/XPF foci number was detected by proximity ligation assay (PLA) and their mRNA levels by real-time PCR. We then correlated the proteins, gene expression and ERCC1/XPF complexes with OC-PDXs’ response to platinum. To the best of our knowledge, this is the first investigation of the role of the ERCC1/XPF complex, detected by PLA, in relation to the response to DDP in ovarian carcinoma. None of the proteins in the BER and NER pathways studied predicted platinum activity in this panel of OC-PDXs, nor did the ERCC1/XPF foci number. These results were partially explained by the experimental evidence that the ERCC1/XPF complex increases after DDP treatment and this possibly better associates with the cancer cells’ abilities to activate the NER pathway to repair platinum-induced damage than its basal level. Our findings highlight the need for DNA functional assays to predict the response to platinum-based therapy.
Collapse
Affiliation(s)
- Federica Guffanti
- Laboratory of Molecular Pharmacology, Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156 Milan, Italy; (F.G.); (E.C.); (F.R.)
| | - Maria Francesca Alvisi
- Laboratory of Methodology for Clinical Research, Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156 Milan, Italy; (M.F.A.); (E.R.)
| | - Elisa Caiola
- Laboratory of Molecular Pharmacology, Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156 Milan, Italy; (F.G.); (E.C.); (F.R.)
| | - Francesca Ricci
- Laboratory of Molecular Pharmacology, Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156 Milan, Italy; (F.G.); (E.C.); (F.R.)
| | - Marcella De Maglie
- Mouse and Animal Pathology Lab (MAPLab), Filarete Foundation, Department of Veterinary Medicine, University of Milan, 20139 Milan, Italy;
| | - Sabina Soldati
- Department of Veterinary Pathology, University of Milan, 20133 Milan, Italy;
| | - Monica Ganzinelli
- Unit of Thoracic Oncology, Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy;
| | - Alessandra Decio
- Laboratory of Cancer Metastasis Therapeutics, Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156 Milan, Italy; (A.D.); (R.G.)
| | - Raffaella Giavazzi
- Laboratory of Cancer Metastasis Therapeutics, Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156 Milan, Italy; (A.D.); (R.G.)
| | - Eliana Rulli
- Laboratory of Methodology for Clinical Research, Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156 Milan, Italy; (M.F.A.); (E.R.)
| | - Giovanna Damia
- Laboratory of Molecular Pharmacology, Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156 Milan, Italy; (F.G.); (E.C.); (F.R.)
- Correspondence: ; Tel.: +39-0239014234
| |
Collapse
|