1
|
Catozzi A, Peiris-Pagès M, Humphrey S, Revill M, Morgan D, Roebuck J, Chen Y, Davies-Williams B, Lallo A, Galvin M, Pearce SP, Kerr A, Priest L, Foy V, Carter M, Caeser R, Chan J, Rudin CM, Blackhall F, Frese KK, Dive C, Simpson KL. Functional Characterisation of the ATOH1 Molecular Subtype Indicates a Pro-Metastatic Role in Small Cell Lung Cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.16.580247. [PMID: 38405859 PMCID: PMC10888785 DOI: 10.1101/2024.02.16.580247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
Molecular subtypes of Small Cell Lung Cancer (SCLC) have been described based on differential expression of transcription factors (TFs) ASCL1, NEUROD1, POU2F3 and immune-related genes. We previously reported an additional subtype based on expression of the neurogenic TF ATOH1 within our SCLC Circulating tumour cell-Derived eXplant (CDX) model biobank. Here we show that ATOH1 protein was detected in 7/81 preclinical models and 16/102 clinical samples of SCLC. In CDX models, ATOH1 directly regulated neurogenesis and differentiation programs consistent with roles in normal tissues. In ex vivo cultures of ATOH1-positive CDX, ATOH1 was required for cell survival. In vivo, ATOH1 depletion slowed tumour growth and suppressed liver metastasis. Our data validate ATOH1 as a bona fide oncogenic driver of SCLC with tumour cell survival and pro-metastatic functions. Further investigation to explore ATOH1 driven vulnerabilities for targeted treatment with predictive biomarkers is warranted.
Collapse
Affiliation(s)
- Alessia Catozzi
- Cancer Research UK Manchester Institute, University of Manchester, Manchester, United Kingdom
- Cancer Research UK Lung Cancer Centre of Excellence, Manchester, United Kingdom
| | - Maria Peiris-Pagès
- Cancer Research UK Manchester Institute, University of Manchester, Manchester, United Kingdom
- Cancer Research UK Lung Cancer Centre of Excellence, Manchester, United Kingdom
| | - Sam Humphrey
- Cancer Research UK Manchester Institute, University of Manchester, Manchester, United Kingdom
- Cancer Research UK Lung Cancer Centre of Excellence, Manchester, United Kingdom
| | - Mitchell Revill
- Cancer Research UK Lung Cancer Centre of Excellence, Manchester, United Kingdom
- Cancer Research UK National Biomarker Centre, University of Manchester, Manchester, United Kingdom
| | - Derrick Morgan
- Cancer Research UK Lung Cancer Centre of Excellence, Manchester, United Kingdom
- Cancer Research UK National Biomarker Centre, University of Manchester, Manchester, United Kingdom
| | - Jordan Roebuck
- Cancer Research UK Lung Cancer Centre of Excellence, Manchester, United Kingdom
- Cancer Research UK National Biomarker Centre, University of Manchester, Manchester, United Kingdom
| | - Yitao Chen
- Cancer Research UK Manchester Institute, University of Manchester, Manchester, United Kingdom
- Cancer Research UK Lung Cancer Centre of Excellence, Manchester, United Kingdom
| | - Bethan Davies-Williams
- Cancer Research UK Manchester Institute, University of Manchester, Manchester, United Kingdom
- Cancer Research UK Lung Cancer Centre of Excellence, Manchester, United Kingdom
| | - Alice Lallo
- Cancer Research UK Manchester Institute, University of Manchester, Manchester, United Kingdom
- Cancer Research UK Lung Cancer Centre of Excellence, Manchester, United Kingdom
| | - Melanie Galvin
- Cancer Research UK Lung Cancer Centre of Excellence, Manchester, United Kingdom
- Cancer Research UK National Biomarker Centre, University of Manchester, Manchester, United Kingdom
| | - Simon P Pearce
- Cancer Research UK National Biomarker Centre, University of Manchester, Manchester, United Kingdom
| | - Alastair Kerr
- Cancer Research UK Lung Cancer Centre of Excellence, Manchester, United Kingdom
- Cancer Research UK National Biomarker Centre, University of Manchester, Manchester, United Kingdom
| | - Lynsey Priest
- Cancer Research UK National Biomarker Centre, University of Manchester, Manchester, United Kingdom
- Medical Oncology, The Christie NHS Foundation Trust, Manchester, United Kingdom
| | - Victoria Foy
- Cancer Research UK Manchester Institute, University of Manchester, Manchester, United Kingdom
- Medical Oncology, The Christie NHS Foundation Trust, Manchester, United Kingdom
| | - Mathew Carter
- Cancer Research UK National Biomarker Centre, University of Manchester, Manchester, United Kingdom
- Medical Oncology, The Christie NHS Foundation Trust, Manchester, United Kingdom
| | - Rebecca Caeser
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Joseph Chan
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Charles M. Rudin
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Fiona Blackhall
- Cancer Research UK Lung Cancer Centre of Excellence, Manchester, United Kingdom
- Medical Oncology, The Christie NHS Foundation Trust, Manchester, United Kingdom
- Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Kristopher K Frese
- Cancer Research UK Manchester Institute, University of Manchester, Manchester, United Kingdom
- Cancer Research UK Lung Cancer Centre of Excellence, Manchester, United Kingdom
- Cancer Research UK National Biomarker Centre, University of Manchester, Manchester, United Kingdom
| | - Caroline Dive
- Cancer Research UK Manchester Institute, University of Manchester, Manchester, United Kingdom
- Cancer Research UK Lung Cancer Centre of Excellence, Manchester, United Kingdom
- Cancer Research UK National Biomarker Centre, University of Manchester, Manchester, United Kingdom
| | - Kathryn L Simpson
- Cancer Research UK Manchester Institute, University of Manchester, Manchester, United Kingdom
- Cancer Research UK Lung Cancer Centre of Excellence, Manchester, United Kingdom
- Cancer Research UK National Biomarker Centre, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
2
|
Martynov I, Dhaka L, Wilke B, Hoyer P, Vahdad MR, Seitz G. Contemporary preclinical mouse models for pediatric rhabdomyosarcoma: from bedside to bench to bedside. Front Oncol 2024; 14:1333129. [PMID: 38371622 PMCID: PMC10869630 DOI: 10.3389/fonc.2024.1333129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Accepted: 01/02/2024] [Indexed: 02/20/2024] Open
Abstract
Background Rhabdomyosarcoma (RMS) is the most common pediatric soft-tissue malignancy, characterized by high clinicalopathological and molecular heterogeneity. Preclinical in vivo models are essential for advancing our understanding of RMS oncobiology and developing novel treatment strategies. However, the diversity of scholarly data on preclinical RMS studies may challenge scientists and clinicians. Hence, we performed a systematic literature survey of contemporary RMS mouse models to characterize their phenotypes and assess their translational relevance. Methods We identified papers published between 01/07/2018 and 01/07/2023 by searching PubMed and Web of Science databases. Results Out of 713 records screened, 118 studies (26.9%) were included in the qualitative synthesis. Cell line-derived xenografts (CDX) were the most commonly utilized (n = 75, 63.6%), followed by patient-derived xenografts (PDX) and syngeneic models, each accounting for 11.9% (n = 14), and genetically engineered mouse models (GEMM) (n = 7, 5.9%). Combinations of different model categories were reported in 5.9% (n = 7) of studies. One study employed a virus-induced RMS model. Overall, 40.0% (n = 30) of the studies utilizing CDX models established alveolar RMS (aRMS), while 38.7% (n = 29) were embryonal phenotypes (eRMS). There were 20.0% (n = 15) of studies that involved a combination of both aRMS and eRMS subtypes. In one study (1.3%), the RMS phenotype was spindle cell/sclerosing. Subcutaneous xenografts (n = 66, 55.9%) were more frequently used compared to orthotopic models (n = 29, 24.6%). Notably, none of the employed cell lines were derived from primary untreated tumors. Only a minority of studies investigated disseminated RMS phenotypes (n = 16, 13.6%). The utilization areas of RMS models included testing drugs (n = 64, 54.2%), studying tumorigenesis (n = 56, 47.5%), tumor modeling (n = 19, 16.1%), imaging (n = 9, 7.6%), radiotherapy (n = 6, 5.1%), long-term effects related to radiotherapy (n = 3, 2.5%), and investigating biomarkers (n = 1, 0.8%). Notably, no preclinical studies focused on surgery. Conclusions This up-to-date review highlights the need for mouse models with dissemination phenotypes and cell lines from primary untreated tumors. Furthermore, efforts should be directed towards underexplored areas such as surgery, radiotherapy, and biomarkers.
Collapse
Affiliation(s)
- Illya Martynov
- Department of Pediatric Surgery and Urology, University Hospital Giessen-Marburg, Marburg, Germany
- Department of Pediatric Surgery, University Hospital Giessen-Marburg, Giessen, Germany
| | - Lajwanti Dhaka
- Department of Pediatric Surgery and Urology, University Hospital Giessen-Marburg, Marburg, Germany
| | - Benedikt Wilke
- Department of Pediatric Surgery and Urology, University Hospital Giessen-Marburg, Marburg, Germany
| | - Paul Hoyer
- Department of Pediatric Surgery and Urology, University Hospital Giessen-Marburg, Marburg, Germany
| | - M. Reza Vahdad
- Department of Pediatric Surgery and Urology, University Hospital Giessen-Marburg, Marburg, Germany
- Department of Pediatric Surgery, University Hospital Giessen-Marburg, Giessen, Germany
| | - Guido Seitz
- Department of Pediatric Surgery and Urology, University Hospital Giessen-Marburg, Marburg, Germany
- Department of Pediatric Surgery, University Hospital Giessen-Marburg, Giessen, Germany
| |
Collapse
|
3
|
Carcaboso AM. Results from the Children's Oncology Group phase III trial of a monoclonal antibody against the insulin-like growth factor-1 receptor in patients with newly diagnosed metastatic Ewing sarcoma. Transl Pediatr 2023; 12:1916-1919. [PMID: 37969125 PMCID: PMC10644028 DOI: 10.21037/tp-23-388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 09/14/2023] [Indexed: 11/17/2023] Open
Affiliation(s)
- Angel M. Carcaboso
- SJD Pediatric Cancer Center Barcelona, Hospital Sant Joan de Deu, Barcelona, Spain
- Pediatric Cancer Program, Institut de Recerca Sant Joan de Deu, Barcelona, Spain
| |
Collapse
|
4
|
Hingorani P, Zhang W, Zhang Z, Xu Z, Wang WL, Roth ME, Wang Y, Gill JB, Harrison DJ, Teicher BA, Erickson SW, Gatto G, Kolb EA, Smith MA, Kurmasheva RT, Houghton PJ, Gorlick R. Trastuzumab Deruxtecan, Antibody-Drug Conjugate Targeting HER2, Is Effective in Pediatric Malignancies: A Report by the Pediatric Preclinical Testing Consortium. Mol Cancer Ther 2022; 21:1318-1325. [PMID: 35657346 DOI: 10.1158/1535-7163.mct-21-0758] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 01/31/2022] [Accepted: 05/18/2022] [Indexed: 11/16/2022]
Abstract
HER2 is expressed in many pediatric solid tumors and is a target for innovative immune therapies including CAR-T cells and antibody-drug conjugates (ADC). We evaluated the preclinical efficacy of trastuzumab deruxtecan (T-DXd, DS-8201a), a humanized monoclonal HER2-targeting antibody conjugated to a topoisomerase 1 inhibitor, DXd, in patient- and cell line-derived xenograft (PDX/CDX) models. HER2 mRNA expression was determined using RNA-seq and protein expression via IHC across multiple pediatric tumor PDX models. Osteosarcoma (OS), malignant rhabdoid tumor (MRT), and Wilms tumor (WT) models with varying HER2 expression were tested using 10 mice per group. Additional histologies such as Ewing sarcoma (EWS), rhabdomyosarcoma (RMS), neuroblastoma (NB), and brain tumors were evaluated using single mouse testing (SMT) experiments. T-DXd or vehicle control was administered intravenously to mice harboring established flank tumors at a dose of 5 mg/kg on day 1. Event-free survival (EFS) and objective response were compared between treatment and control groups. HER2 mRNA expression was observed across histologies, with the highest expression in WT (median = 22 FPKM), followed by MRT, OS, and EWS. The relationship between HER2 protein and mRNA expression was inconsistent. T-DXd significantly prolonged EFS in 6/7 OS, 2/2 MRT, and 3/3 WT PDX models. Complete response (CR) or maintained CR (MCR) were observed for 4/5 WT and MRT models, whereas stable disease was the best response among OS models. SMT experiments also demonstrated activity across multiple solid tumors. Clinical trials assessing the efficacy of a HER2-directed ADC in pediatric patients with HER2-expressing tumors should be considered.
Collapse
Affiliation(s)
- Pooja Hingorani
- Division of Pediatrics, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Wendong Zhang
- Division of Pediatrics, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Zhongting Zhang
- Division of Pediatrics, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Zhaohui Xu
- Division of Pediatrics, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Wei-Lien Wang
- Division of Pathology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Michael E Roth
- Division of Pediatrics, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Yifei Wang
- Division of Pediatrics, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jonathan B Gill
- Division of Pediatrics, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Douglas J Harrison
- Division of Pediatrics, University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | | | - Gregory Gatto
- Global Health Technologies, RTI International, Durham, NC, USA
| | - Edward A Kolb
- Division of Pediatric Hematology/Oncology, Nemours/Alfred I. duPont Hospital for Children, Wilmington, Delaware
| | - Malcolm A Smith
- Cancer Therapeutics Evaluation Program, NCI, Bethesda, Maryland
| | | | - Peter J Houghton
- Greehey Children's Research Cancer Institute, San Antonio, Texas
| | - Richard Gorlick
- Division of Pediatrics, University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
5
|
Zou Z, Sun W, Xu Y, Liu W, Zhong J, Lin X, Chen Y. Application of Multi-Omics Approach in Sarcomas: A Tool for Studying Mechanism, Biomarkers, and Therapeutic Targets. Front Oncol 2022; 12:946022. [PMID: 35875106 PMCID: PMC9304858 DOI: 10.3389/fonc.2022.946022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 06/16/2022] [Indexed: 12/18/2022] Open
Abstract
Sarcomas are rare, heterogeneous mesenchymal neoplasms with various subtypes, each exhibiting unique genetic characteristics. Although studies have been conducted to improve the treatment for sarcomas, the specific development from normal somatic cells to sarcoma cells is still unclear and needs further research. The diagnosis of sarcomas depends heavily on the pathological examination, which is yet a difficult work and requires expert analysis. Advanced treatment like precise medicine optimizes the efficacy of treatment and the prognosis of sarcoma patients, yet, in sarcomas, more studies should be done to put such methods in clinical practice. The revolution of advanced technology has pushed the multi-omics approach to the front, and more could be learnt in sarcomas with such methods. Multi-omics combines the character of each omics techniques, analyzes the mechanism of tumor cells from different levels, which makes up for the shortage of single-omics, and gives us an integrated picture of bioactivities inside tumor cells. Multi-omics research of sarcomas has reached appreciable progress in recent years, leading to a better understanding of the mutation, proliferation, and metastasis of sarcomas. With the help of multi-omics approach, novel biomarkers were found, with promising effects in improving the process of diagnosis, prognosis anticipation, and treatment decision. By analyzing large amounts of biological features, subtype clustering could be done in a better precision, which may be useful in the clinical procedure. In this review, we summarized recent discoveries using multi-omics approach in sarcomas, discussed their merits and challenges, and concluded with future perspectives of the sarcoma research.
Collapse
Affiliation(s)
- Zijian Zou
- Department of Musculoskeletal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wei Sun
- Department of Musculoskeletal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yu Xu
- Department of Musculoskeletal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wanlin Liu
- Department of Musculoskeletal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jingqin Zhong
- Department of Musculoskeletal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xinyi Lin
- Department of Musculoskeletal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yong Chen
- Department of Musculoskeletal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
6
|
Barbet V, Broutier L. Future Match Making: When Pediatric Oncology Meets Organoid Technology. Front Cell Dev Biol 2021; 9:674219. [PMID: 34327198 PMCID: PMC8315550 DOI: 10.3389/fcell.2021.674219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Accepted: 05/10/2021] [Indexed: 11/13/2022] Open
Abstract
Unlike adult cancers that frequently result from the accumulation in time of mutational “hits” often linked to lifestyle, childhood cancers are emerging as diseases of dysregulated development through massive epigenetic alterations. The ability to reconstruct these differences in cancer models is therefore crucial for better understanding the uniqueness of pediatric cancer biology. Cancer organoids (i.e., tumoroids) represent a promising approach for creating patient-derived in vitro cancer models that closely recapitulate the overall pathophysiological features of natural tumorigenesis, including intra-tumoral heterogeneity and plasticity. Though largely applied to adult cancers, this technology is scarcely used for childhood cancers, with a notable delay in technological transfer. However, tumoroids could provide an unprecedented tool to unravel the biology of pediatric cancers and improve their therapeutic management. We herein present the current state-of-the-art of a long awaited and much needed matchmaking.
Collapse
Affiliation(s)
- Virginie Barbet
- Childhood Cancer & Cell Death (C3), Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon (CRCL), Lyon, France
| | - Laura Broutier
- Childhood Cancer & Cell Death (C3), Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon (CRCL), Lyon, France
| |
Collapse
|
7
|
Ghilu S, Kurmasheva RT, Houghton PJ. Developing New Agents for Treatment of Childhood Cancer: Challenges and Opportunities for Preclinical Testing. J Clin Med 2021; 10:jcm10071504. [PMID: 33916592 PMCID: PMC8038510 DOI: 10.3390/jcm10071504] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 03/29/2021] [Accepted: 03/29/2021] [Indexed: 12/26/2022] Open
Abstract
Developing new therapeutics for the treatment of childhood cancer has challenges not usually associated with adult malignancies. Firstly, childhood cancer is rare, with approximately 12,500 new diagnoses annually in the U.S. in children 18 years or younger. With current multimodality treatments, the 5-year event-free survival exceeds 80%, and 70% of patients achieve long-term “cure”, hence the overall number of patients eligible for experimental drugs is small. Childhood cancer comprises many disease entities, the most frequent being acute lymphoblastic leukemias (25% of cancers) and brain tumors (21%), and each of these comprises multiple molecular subtypes. Hence, the numbers of diagnoses even for the more frequently occurring cancers of childhood are small, and undertaking clinical trials remains a significant challenge. Consequently, development of preclinical models that accurately represent each molecular entity can be valuable in identifying those agents or combinations that warrant clinical evaluation. Further, new regulations under the Research to Accelerate Cures and Equity for Children Act (RACE For Children Act) will change the way in which drugs are developed. Here, we will consider some of the limitations of preclinical models and consider approaches that may improve their ability to translate therapy to clinical trial more accurately.
Collapse
|
8
|
Hingorani P, Roth ME, Wang Y, Zhang W, Gill JB, Harrison DJ, Teicher B, Erickson S, Gatto G, Smith MA, Kolb EA, Gorlick R. ABBV-085, Antibody-Drug Conjugate Targeting LRRC15, Is Effective in Osteosarcoma: A Report by the Pediatric Preclinical Testing Consortium. Mol Cancer Ther 2020; 20:535-540. [PMID: 33298592 DOI: 10.1158/1535-7163.mct-20-0406] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 09/03/2020] [Accepted: 12/02/2020] [Indexed: 11/16/2022]
Abstract
Membrane protein leucine-rich repeat containing 15 (LRRC15) is known to be expressed in several solid tumors including osteosarcoma. ABBV-085, an antibody-drug conjugate against LRRC15, conjugated to monomethyl auristatin E (MMAE), was studied in osteosarcoma patient-derived xenografts (PDXs) by the Pediatric Preclinical Testing Consortium (PPTC). LRRC15 expression data were obtained from PPTC RNA-sequencing data for the PDX models. The TARGET database was mined for LRRC15 expression in human osteosarcoma. Protein expression was confirmed via IHC in three PDX models. Seven osteosarcoma PDX models (OS1, OS9, OS33, OS34, OS42, OS55, and OS60) with varying LRRC15 gene expression were studied. ABBV-085 was administered at 3 mg/kg (OS33), 6 mg/kg (all seven PDXs), and 12 mg/kg (OS60) weekly for 4 consecutive weeks via intraperitoneal injection. Control cohorts included vehicle and an isotype MMAE-linked antibody. Tumor volumes and responses were reported using PPTC statistical analysis. OS1, OS33, OS42, OS55, and OS60 had high LRRC15 expression while OS9 and OS34 had low LRRC15 expression. ABBV-085 inhibited tumor growth in six of seven PDX models as compared with vehicle control and significantly improved event-free survival in five of seven models as compared with isotype controls. Two models showed maintained complete responses while all others showed progressive disease. Response correlated with LRRC15 expression. ABBV-085's antitumor activity against osteosarcoma PDX suggests LRRC15 may be a rational target for pursuing clinical trials in patients with this disease.
Collapse
Affiliation(s)
- Pooja Hingorani
- Division of Pediatrics, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Michael E Roth
- Division of Pediatrics, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Yifei Wang
- Division of Pediatrics, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Wendong Zhang
- Division of Pediatrics, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jonathan B Gill
- Division of Pediatrics, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Douglas J Harrison
- Division of Pediatrics, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Beverly Teicher
- Cancer Therapeutics Evaluation Program, National Cancer Institute, Bethesda, Maryland
| | - Stephen Erickson
- Global Health Technologies, RTI International, Research Triangle Park, North Carolina
| | - Gregory Gatto
- Global Health Technologies, RTI International, Research Triangle Park, North Carolina
| | - Malcolm A Smith
- Cancer Therapeutics Evaluation Program, National Cancer Institute, Bethesda, Maryland
| | - Edward A Kolb
- Division of Pediatric Hematology/Oncology, Nemours/Alfred I. duPont Hospital for Children, Wilmington, Delaware
| | - Richard Gorlick
- Division of Pediatrics, University of Texas MD Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
9
|
Robles AJ, Kurmasheva RT, Bandyopadhyay A, Phelps DA, Erickson SW, Lai Z, Kurmashev D, Chen Y, Smith MA, Houghton PJ. Evaluation of Eribulin Combined with Irinotecan for Treatment of Pediatric Cancer Xenografts. Clin Cancer Res 2020; 26:3012-3023. [PMID: 32184294 PMCID: PMC7299830 DOI: 10.1158/1078-0432.ccr-19-1822] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 01/13/2020] [Accepted: 03/12/2020] [Indexed: 01/17/2023]
Abstract
PURPOSE Vincristine combined with camptothecin derivatives showed synergy in preclinical pediatric cancer models, and the combinations are effective in treatment of childhood solid tumors. We determined whether the synergy between vincristine and irinotecan extends to eribulin, another microtubule inhibitor. EXPERIMENTAL DESIGN Vincristine or eribulin, alone or combined with irinotecan, was studied in 12 xenograft models. Tumor regression and time to event were used to assess antitumor activity. Pharmacodynamic studies and RNA sequencing (RNA-seq) were conducted 24 and 144 hours after single-agent or combination treatment. Effects on vascular development were studied in Matrigel plugs implanted in mice. The interaction between binary combinations was examined in vitro. RESULTS Eribulin combined with irinotecan was more effective than vincristine-irinotecan in 6 of 12 models. Pharmacodynamic markers induced by eribulin (phospho-histone H3) and irinotecan (γ-H2A.X) were abrogated in combination-treated tumors. The predominant RNA-seq signature in combination-treated tumors was activation of the TP53 pathway with increased nuclear TP53. Massive apoptosis was observed 24 hours only after treatment with the eribulin combination. In vitro, neither combination showed interaction using combination index analysis. Eribulin alone and the combination caused alterations in developing vasculature. CONCLUSIONS The eribulin combination is very active in these xenograft models, but not synergistic in vitro. The combination reduced pharmacodynamic markers indicative of single-agent mechanisms but in tumors, dramatically activated the TP53 pathway. Although a mechanism for in vivo synergy requires further study, it is possible that eribulin-induced inhibition of microtubule dynamics enhances irinotecan-induced nuclear accumulation of TP53, leading to rapid cell death.
Collapse
Affiliation(s)
- Andrew J Robles
- Greehey Children's Cancer Research Institute, UT Health San Antonio, San Antonio, Texas
| | - Raushan T Kurmasheva
- Greehey Children's Cancer Research Institute, UT Health San Antonio, San Antonio, Texas
| | - Abhik Bandyopadhyay
- Greehey Children's Cancer Research Institute, UT Health San Antonio, San Antonio, Texas
| | - Doris A Phelps
- Greehey Children's Cancer Research Institute, UT Health San Antonio, San Antonio, Texas
| | | | - Zhao Lai
- Greehey Children's Cancer Research Institute, UT Health San Antonio, San Antonio, Texas
| | - Dias Kurmashev
- Greehey Children's Cancer Research Institute, UT Health San Antonio, San Antonio, Texas
| | - Yidong Chen
- Greehey Children's Cancer Research Institute, UT Health San Antonio, San Antonio, Texas
| | - Malcom A Smith
- Cancer Therapy Evaluation Program, National Cancer Institute, Bethesda, Maryland
| | - Peter J Houghton
- Greehey Children's Cancer Research Institute, UT Health San Antonio, San Antonio, Texas.
| |
Collapse
|
10
|
Simpson KL, Stoney R, Frese KK, Simms N, Rowe W, Pearce SP, Humphrey S, Booth L, Morgan D, Dynowski M, Trapani F, Catozzi A, Revill M, Helps T, Galvin M, Girard L, Nonaka D, Carter L, Krebs MG, Cook N, Carter M, Priest L, Kerr A, Gazdar AF, Blackhall F, Dive C. A biobank of small cell lung cancer CDX models elucidates inter- and intratumoral phenotypic heterogeneity. NATURE CANCER 2020; 1:437-451. [PMID: 35121965 DOI: 10.1038/s43018-020-0046-2] [Citation(s) in RCA: 98] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 02/26/2020] [Indexed: 12/17/2022]
Abstract
Although small cell lung cancer (SCLC) is treated as a homogeneous disease, biopsies and preclinical models reveal heterogeneity in transcriptomes and morphology. SCLC subtypes were recently defined by neuroendocrine transcription factor (NETF) expression. Circulating-tumor-cell-derived explant models (CDX) recapitulate donor patients' tumor morphology, diagnostic NE marker expression and chemotherapy responses. We describe a biobank of 38 CDX models, including six CDX pairs generated pretreatment and at disease progression revealing complex intra- and intertumoral heterogeneity. Transcriptomic analysis confirmed three of four previously described subtypes based on ASCL1, NEUROD1 and POU2F3 expression and identified a previously unreported subtype based on another NETF, ATOH1. We document evolution during disease progression exemplified by altered MYC and NOTCH gene expression, increased 'variant' cell morphology, and metastasis without strong evidence of epithelial to mesenchymal transition. This CDX biobank provides a research resource to facilitate SCLC personalized medicine.
Collapse
Affiliation(s)
- Kathryn L Simpson
- Cancer Research UK Manchester Institute Cancer Biomarker Centre, University of Manchester, Manchester, UK
| | - Ruth Stoney
- Cancer Research UK Manchester Institute Cancer Biomarker Centre, University of Manchester, Manchester, UK
- Manchester Institute of Biotechnology, The University of Manchester, Manchester, UK
| | - Kristopher K Frese
- Cancer Research UK Manchester Institute Cancer Biomarker Centre, University of Manchester, Manchester, UK
| | - Nicole Simms
- Cancer Research UK Manchester Institute Cancer Biomarker Centre, University of Manchester, Manchester, UK
| | - William Rowe
- Cancer Research UK Manchester Institute Cancer Biomarker Centre, University of Manchester, Manchester, UK
- Manchester Centre for Synthetic Biology of Fine and Speciality Chemicals (SYNBIOCHEM), Manchester Institute of Biotechnology, The University of Manchester, Manchester, UK
| | - Simon P Pearce
- Cancer Research UK Manchester Institute Cancer Biomarker Centre, University of Manchester, Manchester, UK
| | - Sam Humphrey
- Cancer Research UK Manchester Institute Cancer Biomarker Centre, University of Manchester, Manchester, UK
| | - Laura Booth
- Cancer Research UK Manchester Institute Cancer Biomarker Centre, University of Manchester, Manchester, UK
| | - Derrick Morgan
- Cancer Research UK Manchester Institute Cancer Biomarker Centre, University of Manchester, Manchester, UK
| | - Marek Dynowski
- Scientific Computing Core Facility, Cancer Research UK Manchester Institute, The University of Manchester, Manchester, UK
| | - Francesca Trapani
- Cancer Research UK Manchester Institute Cancer Biomarker Centre, University of Manchester, Manchester, UK
| | - Alessia Catozzi
- Cancer Research UK Manchester Institute Cancer Biomarker Centre, University of Manchester, Manchester, UK
| | - Mitchell Revill
- Cancer Research UK Manchester Institute Cancer Biomarker Centre, University of Manchester, Manchester, UK
| | - Thomas Helps
- Cancer Research UK Manchester Institute Cancer Biomarker Centre, University of Manchester, Manchester, UK
| | - Melanie Galvin
- Cancer Research UK Manchester Institute Cancer Biomarker Centre, University of Manchester, Manchester, UK
| | - Luc Girard
- Hamon Center for Therapeutic Oncology Research, Department of Pharmacology, UT Southwestern Medical Center, Dallas, TX, USA
| | | | - Louise Carter
- The Christie NHS Foundation Trust, Manchester, UK
- Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Matthew G Krebs
- The Christie NHS Foundation Trust, Manchester, UK
- Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Natalie Cook
- The Christie NHS Foundation Trust, Manchester, UK
- Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Mathew Carter
- Cancer Research UK Manchester Institute Cancer Biomarker Centre, University of Manchester, Manchester, UK
| | - Lynsey Priest
- Cancer Research UK Manchester Institute Cancer Biomarker Centre, University of Manchester, Manchester, UK
| | - Alastair Kerr
- Cancer Research UK Manchester Institute Cancer Biomarker Centre, University of Manchester, Manchester, UK
| | - Adi F Gazdar
- Hamon Center for Therapeutic Oncology Research, Department of Pharmacology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Fiona Blackhall
- The Christie NHS Foundation Trust, Manchester, UK
- Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
- Cancer Research UK Lung Cancer Centre of Excellence, Manchester, UK
| | - Caroline Dive
- Cancer Research UK Manchester Institute Cancer Biomarker Centre, University of Manchester, Manchester, UK.
- Cancer Research UK Lung Cancer Centre of Excellence, Manchester, UK.
| |
Collapse
|
11
|
Houghton PJ, Kurmasheva RT. Challenges and Opportunities for Childhood Cancer Drug Development. Pharmacol Rev 2019; 71:671-697. [PMID: 31558580 PMCID: PMC6768308 DOI: 10.1124/pr.118.016972] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Cancer in children is rare with approximately 15,700 new cases diagnosed in the United States annually. Through use of multimodality therapy (surgery, radiation therapy, and aggressive chemotherapy), 70% of patients will be "cured" of their disease, and 5-year event-free survival exceeds 80%. However, for patients surviving their malignancy, therapy-related long-term adverse effects are severe, with an estimated 50% having chronic life-threatening toxicities related to therapy in their fourth or fifth decade of life. While overall intensive therapy with cytotoxic agents continues to reduce cancer-related mortality, new understanding of the molecular etiology of many childhood cancers offers an opportunity to redirect efforts to develop effective, less genotoxic therapeutic options, including agents that target oncogenic drivers directly, and the potential for use of agents that target the tumor microenvironment and immune-directed therapies. However, for many high-risk cancers, significant challenges remain.
Collapse
Affiliation(s)
- Peter J Houghton
- Greehey Children's Cancer Research Institute, University of Texas Health, San Antonio, Texas
| | - Raushan T Kurmasheva
- Greehey Children's Cancer Research Institute, University of Texas Health, San Antonio, Texas
| |
Collapse
|
12
|
Grünewald TGP, Cidre-Aranaz F, Surdez D, Tomazou EM, de Álava E, Kovar H, Sorensen PH, Delattre O, Dirksen U. Ewing sarcoma. Nat Rev Dis Primers 2018; 4:5. [PMID: 29977059 DOI: 10.1038/s41572-018-0003-x] [Citation(s) in RCA: 449] [Impact Index Per Article: 74.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Ewing sarcoma is the second most frequent bone tumour of childhood and adolescence that can also arise in soft tissue. Ewing sarcoma is a highly aggressive cancer, with a survival of 70-80% for patients with standard-risk and localized disease and ~30% for those with metastatic disease. Treatment comprises local surgery, radiotherapy and polychemotherapy, which are associated with acute and chronic adverse effects that may compromise quality of life in survivors. Histologically, Ewing sarcomas are composed of small round cells expressing high levels of CD99. Genetically, they are characterized by balanced chromosomal translocations in which a member of the FET gene family is fused with an ETS transcription factor, with the most common fusion being EWSR1-FLI1 (85% of cases). Ewing sarcoma breakpoint region 1 protein (EWSR1)-Friend leukaemia integration 1 transcription factor (FLI1) is a tumour-specific chimeric transcription factor (EWSR1-FLI1) with neomorphic effects that massively rewires the transcriptome. Additionally, EWSR1-FLI1 reprogrammes the epigenome by inducing de novo enhancers at GGAA microsatellites and by altering the state of gene regulatory elements, creating a unique epigenetic signature. Additional mutations at diagnosis are rare and mainly involve STAG2, TP53 and CDKN2A deletions. Emerging studies on the molecular mechanisms of Ewing sarcoma hold promise for improvements in early detection, disease monitoring, lower treatment-related toxicity, overall survival and quality of life.
Collapse
Affiliation(s)
- Thomas G P Grünewald
- Max-Eder Research Group for Pediatric Sarcoma Biology, Institute of Pathology, Faculty of Medicine, LMU Munich, Munich, Germany. .,Institute of Pathology, Faculty of Medicine, LMU Munich, Munich, Germany. .,German Cancer Consortium, partner site Munich, Munich, Germany. .,German Cancer Research Center, Heidelberg, Germany.
| | - Florencia Cidre-Aranaz
- Max-Eder Research Group for Pediatric Sarcoma Biology, Institute of Pathology, Faculty of Medicine, LMU Munich, Munich, Germany. .,Institute of Pathology, Faculty of Medicine, LMU Munich, Munich, Germany. .,German Cancer Consortium, partner site Munich, Munich, Germany. .,German Cancer Research Center, Heidelberg, Germany.
| | - Didier Surdez
- INSERM U830, Équipe Labellisé LNCC, PSL Université, SIREDO Oncology Centre, Institut Curie, Paris, France
| | - Eleni M Tomazou
- Children's Cancer Research Institute, St Anna Kinderkrebsforschung, Vienna, Austria
| | - Enrique de Álava
- Institute of Biomedicine of Seville, Virgen del Rocío University Hospital/CSIC/University of Seville/CIBERONC, Seville, Spain
| | - Heinrich Kovar
- Children's Cancer Research Institute, St Anna Kinderkrebsforschung, Vienna, Austria.,Department of Pediatrics, Medical University Vienna, Vienna, Austria
| | - Poul H Sorensen
- British Columbia Cancer Research Centre and University of British Columbia, Vancouver, Canada
| | - Olivier Delattre
- INSERM U830, Équipe Labellisé LNCC, PSL Université, SIREDO Oncology Centre, Institut Curie, Paris, France
| | - Uta Dirksen
- German Cancer Research Center, Heidelberg, Germany.,Cooperative Ewing Sarcoma Study group, Essen University Hospital, Essen, Germany.,German Cancer Consortium, partner site Essen, Essen, Germany
| |
Collapse
|
13
|
Shapiro MC, Tang T, Dasgupta A, Kurenbekova L, Shuck R, Gaber MW, Yustein JT. In Vitro and In Vivo Characterization of a Preclinical Irradiation-Adapted Model for Ewing Sarcoma. Int J Radiat Oncol Biol Phys 2018. [DOI: 10.1016/j.ijrobp.2018.01.095] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
14
|
Zarzosa P, Navarro N, Giralt I, Molist C, Almazán-Moga A, Vidal I, Soriano A, Segura MF, Hladun R, Villanueva A, Gallego S, Roma J. Patient-derived xenografts for childhood solid tumors: a valuable tool to test new drugs and personalize treatments. Clin Transl Oncol 2016; 19:44-50. [PMID: 27718156 DOI: 10.1007/s12094-016-1557-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Accepted: 09/22/2016] [Indexed: 12/23/2022]
Abstract
The use of preclinical models is essential in translational cancer research and especially important in pediatric cancer given the low incidence of each particular type of cancer. Cell line cultures have led to significant advances in cancer biology. However, cell lines have adapted to growth in artificial culture conditions, thereby undergoing genetic and phenotypic changes which may hinder the translational application. Tumor grafts developed in mice from patient tumor tissues, generally known as patient-derived xenografts (PDXs), are interesting alternative approaches to reproducing the biology of the original tumor. This review is focused on highlighting the interest of PDX models in pediatric cancer research and supporting strategies of personalized medicine. This review provides: (1) a description of the background of PDX in cancer, (2) the particular case of PDX in pediatric cancer, (3) how PDX can improve personalized medicine strategies, (4) new methods to increase engraftment, and, finally, (5) concluding remarks.
Collapse
Affiliation(s)
- P Zarzosa
- Laboratory of Translational Research in Childhood and Adolescent Cancer, Hospital Universitari Vall d'Hebron, Vall d'Hebron Research Institute. Universitat Autònoma de Barcelona, Barcelona, Spain
| | - N Navarro
- Laboratory of Translational Research in Childhood and Adolescent Cancer, Hospital Universitari Vall d'Hebron, Vall d'Hebron Research Institute. Universitat Autònoma de Barcelona, Barcelona, Spain
| | - I Giralt
- Laboratory of Translational Research in Childhood and Adolescent Cancer, Hospital Universitari Vall d'Hebron, Vall d'Hebron Research Institute. Universitat Autònoma de Barcelona, Barcelona, Spain
| | - C Molist
- Laboratory of Translational Research in Childhood and Adolescent Cancer, Hospital Universitari Vall d'Hebron, Vall d'Hebron Research Institute. Universitat Autònoma de Barcelona, Barcelona, Spain
| | - A Almazán-Moga
- Laboratory of Translational Research in Childhood and Adolescent Cancer, Hospital Universitari Vall d'Hebron, Vall d'Hebron Research Institute. Universitat Autònoma de Barcelona, Barcelona, Spain
| | - I Vidal
- Laboratory of Translational Research in Childhood and Adolescent Cancer, Hospital Universitari Vall d'Hebron, Vall d'Hebron Research Institute. Universitat Autònoma de Barcelona, Barcelona, Spain
| | - A Soriano
- Laboratory of Translational Research in Childhood and Adolescent Cancer, Hospital Universitari Vall d'Hebron, Vall d'Hebron Research Institute. Universitat Autònoma de Barcelona, Barcelona, Spain
| | - M F Segura
- Laboratory of Translational Research in Childhood and Adolescent Cancer, Hospital Universitari Vall d'Hebron, Vall d'Hebron Research Institute. Universitat Autònoma de Barcelona, Barcelona, Spain
| | - R Hladun
- Pediatric Oncology and Hematology Department, Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - A Villanueva
- Chemoresistance and Predicitive Factors Laboratory, Program Against Cancer Therapeutic Resistance (ProCURE), Catalan Institute of Oncology (ICO), Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, 08908, Barcelona, Spain
- Xenopat S.L. Business Bioincubator Bellvitge Health Science Campus, L'Hospitalet de Llobregat, 08908, Barcelona, Spain
| | - S Gallego
- Laboratory of Translational Research in Childhood and Adolescent Cancer, Hospital Universitari Vall d'Hebron, Vall d'Hebron Research Institute. Universitat Autònoma de Barcelona, Barcelona, Spain
- Pediatric Oncology and Hematology Department, Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - J Roma
- Laboratory of Translational Research in Childhood and Adolescent Cancer, Hospital Universitari Vall d'Hebron, Vall d'Hebron Research Institute. Universitat Autònoma de Barcelona, Barcelona, Spain.
| |
Collapse
|