451
|
Abstract
Therapeutics that block kinases, transcriptional modifiers, immune checkpoints and other biological vulnerabilities are transforming cancer treatment. As a result, many patients achieve dramatic responses, including complete radiographical or pathological remission, yet retain minimal residual disease (MRD), which results in relapse. New functional approaches can characterize clonal heterogeneity and predict therapeutic sensitivity of MRD at a single-cell level. Preliminary evidence suggests that iterative detection, profiling and targeting of MRD would meaningfully improve outcomes and may even lead to cure.
Collapse
Affiliation(s)
- Marlise R. Luskin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA,
| | - Mark A. Murakami
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA,
| | - Scott R. Manalis
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA
- Corresponding authors: (S. R. M.) and (D. M. W.)
| | - David M. Weinstock
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA,
- Broad Institute of MIT and Harvard University, Cambridge, Massachusetts, 02142, USA
- Corresponding authors: (S. R. M.) and (D. M. W.)
| |
Collapse
|
452
|
Corso S, Cargnelutti M, Durando S, Menegon S, Apicella M, Migliore C, Capeloa T, Ughetto S, Isella C, Medico E, Bertotti A, Sassi F, Sarotto I, Casorzo L, Pisacane A, Mangioni M, Sottile A, Degiuli M, Fumagalli U, Sgroi G, Molfino S, De Manzoni G, Rosati R, De Simone M, Marrelli D, Saragoni L, Rausei S, Pallabazzer G, Roviello F, Cassoni P, Sapino A, Bass A, Giordano S. Rituximab Treatment Prevents Lymphoma Onset in Gastric Cancer Patient-Derived Xenografts. Neoplasia 2018; 20:443-455. [PMID: 29574251 PMCID: PMC5915970 DOI: 10.1016/j.neo.2018.02.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 02/06/2018] [Accepted: 02/14/2018] [Indexed: 12/13/2022] Open
Abstract
Patient-Derived Xenografts (PDXs), entailing implantation of cancer specimens in immunocompromised mice, are emerging as a valuable translational model that could help validate biologically relevant targets and assist the clinical development of novel therapeutic strategies for gastric cancer. More than 30% of PDXs generated from gastric carcinoma samples developed human B-cell lymphomas instead of gastric cancer. These lymphomas were monoclonal, Epstein Barr Virus (EBV) positive, originated tumorigenic cell cultures and displayed a mutational burden and an expression profile distinct from gastric adenocarcinomas. The ability of grafted samples to develop lymphomas did not correlate with patient outcome, nor with the histotype, the lymphocyte infiltration level, or the EBV status of the original gastric tumor, impeding from foreseeing lymphoma onset. Interestingly, lymphoma development was significantly more frequent when primary rather than metastatic samples were grafted. Notably, the development of such lympho-proliferative disease could be prevented by a short rituximab treatment upon mice implant, without negatively affecting gastric carcinoma engraftment. Due to the high frequency of human lymphoma onset, our data show that a careful histologic analysis is mandatory when generating gastric cancer PDXs. Such care would avoid misleading results that could occur if testing of putative gastric cancer therapies is performed in lymphoma PDXs. We propose rituximab treatment of mice to prevent lymphoma development in PDX models, averting the loss of human-derived samples.
Collapse
Affiliation(s)
- Simona Corso
- Department of Oncology, University of Torino, Candiolo, Italy; Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy.
| | | | | | | | | | - Cristina Migliore
- Department of Oncology, University of Torino, Candiolo, Italy; Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
| | - Tania Capeloa
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy; Department of Clinical and Biological Sciences, University of Torino, Orbassano, Italy
| | - Stefano Ughetto
- Department of Oncology, University of Torino, Candiolo, Italy; Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
| | | | - Enzo Medico
- Department of Oncology, University of Torino, Candiolo, Italy; Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
| | - Andrea Bertotti
- Department of Oncology, University of Torino, Candiolo, Italy; Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
| | | | - Ivana Sarotto
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
| | - Laura Casorzo
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
| | | | | | | | | | | | - Giovanni Sgroi
- Surgical Oncology Unit, Surgical Science Department, ASST Bergamo Ovest, Treviglio (BG), Italy
| | - Sarah Molfino
- Department of Clinical and Experimental Sciences, Surgical Clinic, University of Brescia, Brescia, Italy
| | - Giovanni De Manzoni
- First Department of General Surgery, Borgo Trento Hospital, University of Verona, Italy
| | - Riccardo Rosati
- Gastroenterological Surgery Unit, IRCCS San Raffaele Hospital, Vita-Salute University, Milan, Italy
| | | | - Daniele Marrelli
- Department of Medicine, Surgery and Neurosciences, Unit of General Surgery and Surgical Oncology, University of Siena, Italy
| | - Luca Saragoni
- Pathology Unit, Morgagni-Pierantoni Hospital, Forlì, Italy
| | - Stefano Rausei
- Department of Surgery, University of Insubria, Varese, Italy
| | | | - Franco Roviello
- Department of Medicine, Surgery and Neurosciences, Unit of General Surgery and Surgical Oncology, University of Siena, Italy
| | - Paola Cassoni
- Department of Medical Sciences, University of Torino, Italy
| | - Anna Sapino
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy; Department of Medical Sciences, University of Torino, Italy
| | - Adam Bass
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Silvia Giordano
- Department of Oncology, University of Torino, Candiolo, Italy; Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy.
| |
Collapse
|
453
|
Patient-derived conditionally reprogrammed cells maintain intra-tumor genetic heterogeneity. Sci Rep 2018; 8:4097. [PMID: 29511269 PMCID: PMC5840339 DOI: 10.1038/s41598-018-22427-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Accepted: 02/22/2018] [Indexed: 02/05/2023] Open
Abstract
Preclinical in vitro models provide an essential tool to study cancer cell biology as well as aid in translational research, including drug target identification and drug discovery efforts. For any model to be clinically relevant, it needs to recapitulate the biology and cell heterogeneity of the primary tumor. We recently developed and described a conditional reprogramming (CR) cell technology that addresses many of these needs and avoids the deficiencies of most current cancer cell lines, which are usually clonal in origin. Here, we used the CR cell method to generate a collection of patient-derived cell cultures from non-small cell lung cancers (NSCLC). Whole exome sequencing and copy number variations are used for the first time to address the capability of CR cells to keep their tumor-derived heterogeneity. Our results indicated that these primary cultures largely maintained the molecular characteristics of the original tumors. Using a mutant-allele tumor heterogeneity (MATH) score, we showed that CR cells are able to keep and maintain most of the intra-tumoral heterogeneity, suggesting oligoclonality of these cultures. CR cultures therefore represent a pre-clinical lung cancer model for future basic and translational studies.
Collapse
|
454
|
Williams JA. Using PDX for Preclinical Cancer Drug Discovery: The Evolving Field. J Clin Med 2018; 7:E41. [PMID: 29498669 PMCID: PMC5867567 DOI: 10.3390/jcm7030041] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 01/19/2018] [Accepted: 02/21/2018] [Indexed: 12/21/2022] Open
Abstract
The ability to create patient derived xenografts (PDXs) has evolved considerably from the breakthrough of the development of immune compromised mice. How researchers in drug discovery have utilized PDX of certain cancer types has also changed from traditionally selecting a few models to profile a drug, to opting to assess inter-tumor response heterogeneity by screening across a broad range of tumor models, and subsequently to enable clinical stratification strategies. As with all models and methodologies, imperfections with this approach are apparent, and our understanding of the fidelity of these models continues to expand. To date though, they are still viewed as one of the most faithful modeling systems in oncology. Currently, there are many efforts ongoing to increase the utility and translatability of PDXs, including introducing a human immune component to enable immunotherapy studies.
Collapse
Affiliation(s)
- Juliet A Williams
- Novartis Institutes for Biomedical Research, Cambridge, MA 02139, USA.
| |
Collapse
|
455
|
Yada E, Wada S, Yoshida S, Sasada T. Use of patient-derived xenograft mouse models in cancer research and treatment. Future Sci OA 2018; 4:FSO271. [PMID: 29568561 PMCID: PMC5859329 DOI: 10.4155/fsoa-2017-0136] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 11/08/2017] [Indexed: 12/12/2022] Open
Affiliation(s)
- Erica Yada
- Division of Cancer Immunotherapy, Kanagawa Cancer Center Research Institute, Yokohama, Japan
| | - Satoshi Wada
- Division of Cancer Immunotherapy, Kanagawa Cancer Center Research Institute, Yokohama, Japan
- Cancer Vaccine Center, Kanagawa Cancer Center, Yokohama, Japan
| | - Shintaro Yoshida
- Division of Cancer Immunotherapy, Kanagawa Cancer Center Research Institute, Yokohama, Japan
| | - Tetsuro Sasada
- Division of Cancer Immunotherapy, Kanagawa Cancer Center Research Institute, Yokohama, Japan
- Cancer Vaccine Center, Kanagawa Cancer Center, Yokohama, Japan
| |
Collapse
|
456
|
Liu H, Murphy CJ, Karreth FA, Emdal KB, White FM, Elemento O, Toker A, Wulf GM, Cantley LC. Identifying and Targeting Sporadic Oncogenic Genetic Aberrations in Mouse Models of Triple-Negative Breast Cancer. Cancer Discov 2018; 8:354-369. [PMID: 29203461 PMCID: PMC5907916 DOI: 10.1158/2159-8290.cd-17-0679] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 11/11/2017] [Accepted: 11/27/2017] [Indexed: 02/07/2023]
Abstract
Triple-negative breast cancers (TNBC) are genetically characterized by aberrations in TP53 and a low rate of activating point mutations in common oncogenes, rendering it challenging in applying targeted therapies. We performed whole-exome sequencing (WES) and RNA sequencing (RNA-seq) to identify somatic genetic alterations in mouse models of TNBCs driven by loss of Trp53 alone or in combination with Brca1 Amplifications or translocations that resulted in elevated oncoprotein expression or oncoprotein-containing fusions, respectively, as well as frameshift mutations of tumor suppressors were identified in approximately 50% of the tumors evaluated. Although the spectrum of sporadic genetic alterations was diverse, the majority had in common the ability to activate the MAPK/PI3K pathways. Importantly, we demonstrated that approved or experimental drugs efficiently induce tumor regression specifically in tumors harboring somatic aberrations of the drug target. Our study suggests that the combination of WES and RNA-seq on human TNBC will lead to the identification of actionable therapeutic targets for precision medicine-guided TNBC treatment.Significance: Using combined WES and RNA-seq analyses, we identified sporadic oncogenic events in TNBC mouse models that share the capacity to activate the MAPK and/or PI3K pathways. Our data support a treatment tailored to the genetics of individual tumors that parallels the approaches being investigated in the ongoing NCI-MATCH, My Pathway Trial, and ESMART clinical trials. Cancer Discov; 8(3); 354-69. ©2017 AACR.See related commentary by Natrajan et al., p. 272See related article by Matissek et al., p. 336This article is highlighted in the In This Issue feature, p. 253.
Collapse
Affiliation(s)
- Hui Liu
- Department of Pathology, and Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Charles J Murphy
- Institute for Computational Biomedicine, Weill Cornell Medical College, New York, New York
- Meyer Cancer Center, Weill Cornell Medicine, New York, New York
| | - Florian A Karreth
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Kristina B Emdal
- Department of Biological Engineering, Koch Institute for Integrative Cancer Research, MIT, Cambridge, Massachusetts
| | - Forest M White
- Department of Biological Engineering, Koch Institute for Integrative Cancer Research, MIT, Cambridge, Massachusetts
| | - Olivier Elemento
- Institute for Computational Biomedicine, Weill Cornell Medical College, New York, New York
| | - Alex Toker
- Department of Pathology, and Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
- Department of Pathology, and Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, and Ludwig Center at Harvard, Boston, Massachusetts
| | - Gerburg M Wulf
- Division of Hematology/Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Lewis C Cantley
- Meyer Cancer Center, Weill Cornell Medicine, New York, New York.
| |
Collapse
|
457
|
Vlachogiannis G, Hedayat S, Vatsiou A, Jamin Y, Fernández-Mateos J, Khan K, Lampis A, Eason K, Huntingford I, Burke R, Rata M, Koh DM, Tunariu N, Collins D, Hulkki-Wilson S, Ragulan C, Spiteri I, Moorcraft SY, Chau I, Rao S, Watkins D, Fotiadis N, Bali M, Darvish-Damavandi M, Lote H, Eltahir Z, Smyth EC, Begum R, Clarke PA, Hahne JC, Dowsett M, de Bono J, Workman P, Sadanandam A, Fassan M, Sansom OJ, Eccles S, Starling N, Braconi C, Sottoriva A, Robinson SP, Cunningham D, Valeri N. Patient-derived organoids model treatment response of metastatic gastrointestinal cancers. Science 2018; 359:920-926. [PMID: 29472484 PMCID: PMC6112415 DOI: 10.1126/science.aao2774] [Citation(s) in RCA: 1277] [Impact Index Per Article: 182.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Revised: 10/26/2017] [Accepted: 01/11/2018] [Indexed: 12/20/2022]
Abstract
Patient-derived organoids (PDOs) have recently emerged as robust preclinical models; however, their potential to predict clinical outcomes in patients has remained unclear. We report on a living biobank of PDOs from metastatic, heavily pretreated colorectal and gastroesophageal cancer patients recruited in phase 1/2 clinical trials. Phenotypic and genotypic profiling of PDOs showed a high degree of similarity to the original patient tumors. Molecular profiling of tumor organoids was matched to drug-screening results, suggesting that PDOs could complement existing approaches in defining cancer vulnerabilities and improving treatment responses. We compared responses to anticancer agents ex vivo in organoids and PDO-based orthotopic mouse tumor xenograft models with the responses of the patients in clinical trials. Our data suggest that PDOs can recapitulate patient responses in the clinic and could be implemented in personalized medicine programs.
Collapse
Affiliation(s)
| | - Somaieh Hedayat
- Division of Molecular Pathology, The Institute of Cancer Research, London, UK
| | - Alexandra Vatsiou
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, UK
| | - Yann Jamin
- Cancer Research UK Cancer Imaging Centre, Division of Radiotherapy and Imaging, The Institute of Cancer Research and Royal Marsden Hospital, London, UK
| | - Javier Fernández-Mateos
- Division of Molecular Pathology, The Institute of Cancer Research, London, UK
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, UK
| | - Khurum Khan
- Division of Molecular Pathology, The Institute of Cancer Research, London, UK
- Department of Medicine, The Royal Marsden NHS Trust, London, UK
| | - Andrea Lampis
- Division of Molecular Pathology, The Institute of Cancer Research, London, UK
| | - Katherine Eason
- Division of Molecular Pathology, The Institute of Cancer Research, London, UK
| | - Ian Huntingford
- Division of Molecular Pathology, The Institute of Cancer Research, London, UK
| | - Rosemary Burke
- Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London, UK
| | - Mihaela Rata
- Cancer Research UK Cancer Imaging Centre, Division of Radiotherapy and Imaging, The Institute of Cancer Research and Royal Marsden Hospital, London, UK
| | - Dow-Mu Koh
- Cancer Research UK Cancer Imaging Centre, Division of Radiotherapy and Imaging, The Institute of Cancer Research and Royal Marsden Hospital, London, UK
- Department of Radiology, The Royal Marsden NHS Trust, London, UK
| | - Nina Tunariu
- Cancer Research UK Cancer Imaging Centre, Division of Radiotherapy and Imaging, The Institute of Cancer Research and Royal Marsden Hospital, London, UK
- Department of Radiology, The Royal Marsden NHS Trust, London, UK
| | - David Collins
- Cancer Research UK Cancer Imaging Centre, Division of Radiotherapy and Imaging, The Institute of Cancer Research and Royal Marsden Hospital, London, UK
| | - Sanna Hulkki-Wilson
- Division of Molecular Pathology, The Institute of Cancer Research, London, UK
| | - Chanthirika Ragulan
- Division of Molecular Pathology, The Institute of Cancer Research, London, UK
| | - Inmaculada Spiteri
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, UK
| | | | - Ian Chau
- Department of Medicine, The Royal Marsden NHS Trust, London, UK
| | - Sheela Rao
- Department of Medicine, The Royal Marsden NHS Trust, London, UK
| | - David Watkins
- Department of Medicine, The Royal Marsden NHS Trust, London, UK
| | - Nicos Fotiadis
- Department of Radiology, The Royal Marsden NHS Trust, London, UK
| | - Maria Bali
- Cancer Research UK Cancer Imaging Centre, Division of Radiotherapy and Imaging, The Institute of Cancer Research and Royal Marsden Hospital, London, UK
- Department of Radiology, The Royal Marsden NHS Trust, London, UK
| | | | - Hazel Lote
- Division of Molecular Pathology, The Institute of Cancer Research, London, UK
- Department of Medicine, The Royal Marsden NHS Trust, London, UK
| | - Zakaria Eltahir
- Division of Molecular Pathology, The Institute of Cancer Research, London, UK
| | | | - Ruwaida Begum
- Department of Medicine, The Royal Marsden NHS Trust, London, UK
| | - Paul A Clarke
- Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London, UK
| | - Jens C Hahne
- Division of Molecular Pathology, The Institute of Cancer Research, London, UK
| | - Mitchell Dowsett
- Ralph Lauren Centre for Breast Cancer Research, Royal Marsden Hospital NHS Trust, London, UK
| | - Johann de Bono
- Division of Clinical Studies, The Institute of Cancer Research, London, UK
| | - Paul Workman
- Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London, UK
| | - Anguraj Sadanandam
- Division of Molecular Pathology, The Institute of Cancer Research, London, UK
| | - Matteo Fassan
- Department of Medicine, Surgical Pathology and Cytopathology Unit, University of Padua, Padua, Italy
| | | | - Suzanne Eccles
- Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London, UK
| | | | - Chiara Braconi
- Department of Medicine, The Royal Marsden NHS Trust, London, UK
- Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London, UK
| | - Andrea Sottoriva
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, UK
| | - Simon P Robinson
- Cancer Research UK Cancer Imaging Centre, Division of Radiotherapy and Imaging, The Institute of Cancer Research and Royal Marsden Hospital, London, UK
| | | | - Nicola Valeri
- Division of Molecular Pathology, The Institute of Cancer Research, London, UK.
- Department of Medicine, The Royal Marsden NHS Trust, London, UK
| |
Collapse
|
458
|
Mattar M, McCarthy CR, Kulick AR, Qeriqi B, Guzman S, de Stanchina E. Establishing and Maintaining an Extensive Library of Patient-Derived Xenograft Models. Front Oncol 2018. [PMID: 29515970 PMCID: PMC5825907 DOI: 10.3389/fonc.2018.00019] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Patient-derived xenograft (PDX) models have recently emerged as a highly desirable platform in oncology and are expected to substantially broaden the way in vivo studies are designed and executed and to reshape drug discovery programs. However, acquisition of patient-derived samples, and propagation, annotation and distribution of PDXs are complex processes that require a high degree of coordination among clinic, surgery and laboratory personnel, and are fraught with challenges that are administrative, procedural and technical. Here, we examine in detail the major aspects of this complex process and relate our experience in establishing a PDX Core Laboratory within a large academic institution.
Collapse
Affiliation(s)
- Marissa Mattar
- Antitumor Assessment Core Facility, Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Craig R McCarthy
- Antitumor Assessment Core Facility, Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Amanda R Kulick
- Antitumor Assessment Core Facility, Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Besnik Qeriqi
- Antitumor Assessment Core Facility, Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Sean Guzman
- Antitumor Assessment Core Facility, Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Elisa de Stanchina
- Antitumor Assessment Core Facility, Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| |
Collapse
|
459
|
Verheecke M, Hermans E, Tuyaerts S, Souche E, Van Bree R, Verbist G, Everaert T, Cortès-Calabuig A, Van Houdt J, Van Calsteren K, Amant F. Acute Drug Effects on the Human Placental Tissue: The Development of a Placental Murine Xenograft Model. Reprod Sci 2018; 25:1637-1648. [PMID: 29439620 DOI: 10.1177/1933719118756771] [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: 11/16/2022]
Abstract
OBJECTIVE A pilot study was conducted to establish a human placental xenograft, which could serve as a model to evaluate the effect of toxic exposures during pregnancy. STUDY DESIGN The protocol consisted of engraftment of third-trimester human placental tissue in immunocompromised mice, after induction of a pseudo-pregnancy state by ovariectomy and progesterone supplementation. To validate the model, the placental tissue before and after engraftment was examined by immunohistochemistry, fluorescence-activated cell sorting (FACS), single-nucleotide polymorphism (SNP) genotyping, and whole transcriptome sequencing (WTSS). The human chorion gonadotropin (hCG) production in serum and urine was examined by enzyme-linked immunosorbent assay. RESULTS Microscopic evaluation of the placental tissue before and after engraftment revealed a stable morphology and preserved histological structure of the human tissue. Viable trophoblast was present after engraftment and remained stable over time. Vascularization and hormonal secretion (hCG) were present till 3 weeks after engraftment. Thirty-one SNPs were equally present, and there was a stable expression level for 56 451 genes evaluated by whole transcriptome sequencing. CONCLUSION Although this human placental xenograft model cannot copy the unique uterine environment in which the placenta develops and interacts between the mother and the fetus, it could be a suitable tool to evaluate the acute impact and adaptive processes of the placental tissue to environmental changes.
Collapse
Affiliation(s)
- Magali Verheecke
- 1 Department of Oncology, KU Leuven, Leuven, Belgium.,2 Department of Obstetrics and Gynecology, University Hospitals Leuven, Leuven, Belgium
| | - Els Hermans
- 3 TRACE (the patient-derived human xenograft platform), Catholic University of Leuven, KU Leuven, Leuven, Belgium.,4 Center for Human Genetics, University Hospitals Leuven, Leuven, Belgium.,5 Genomics Core, KU Leuven, Leuven, Belgium
| | | | - Erika Souche
- 4 Center for Human Genetics, University Hospitals Leuven, Leuven, Belgium.,5 Genomics Core, KU Leuven, Leuven, Belgium
| | - Rita Van Bree
- 6 Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Godelieve Verbist
- 6 Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Tina Everaert
- 1 Department of Oncology, KU Leuven, Leuven, Belgium
| | - Alvaro Cortès-Calabuig
- 4 Center for Human Genetics, University Hospitals Leuven, Leuven, Belgium.,5 Genomics Core, KU Leuven, Leuven, Belgium
| | - Jeroen Van Houdt
- 4 Center for Human Genetics, University Hospitals Leuven, Leuven, Belgium.,5 Genomics Core, KU Leuven, Leuven, Belgium
| | - Kristel Van Calsteren
- 2 Department of Obstetrics and Gynecology, University Hospitals Leuven, Leuven, Belgium.,6 Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Frederic Amant
- 1 Department of Oncology, KU Leuven, Leuven, Belgium.,7 Center for Gynecologic Oncology Amsterdam (CGOA), Netherlands Cancer Institute, (NKI), Amsterdam, the Netherlands
| |
Collapse
|
460
|
Gambara G, Gaebler M, Keilholz U, Regenbrecht CRA, Silvestri A. From Chemotherapy to Combined Targeted Therapeutics: In Vitro and in Vivo Models to Decipher Intra-tumor Heterogeneity. Front Pharmacol 2018; 9:77. [PMID: 29491834 PMCID: PMC5817069 DOI: 10.3389/fphar.2018.00077] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Accepted: 01/23/2018] [Indexed: 12/15/2022] Open
Abstract
Recent advances in next-generation sequencing and other omics technologies capable to map cell fate provide increasing evidence on the crucial role of intra-tumor heterogeneity (ITH) for cancer progression. The different facets of ITH, from genomic to microenvironmental heterogeneity and the hierarchical cellular architecture originating from the cancer stem cell compartment, contribute to the range of tumor phenotypes. Decoding these complex data resulting from the analysis of tumor tissue complexity poses a challenge for developing novel therapeutic strategies that can counteract tumor evolution and cellular plasticity. To achieve this aim, the development of in vitro and in vivo cancer models that resemble the complexity of ITH is crucial in understanding the interplay of cells and their (micro)environment and, consequently, in testing the efficacy of new targeted treatments and novel strategies of tailoring combinations of treatments to the individual composition of the tumor. This challenging approach may be an important cornerstone in overcoming the development of pharmaco-resistances during multiple lines of treatment. In this paper, we report the latest advances in patient-derived 3D (PD3D) cell cultures and patient-derived tumor xenografts (PDX) as in vitro and in vivo models that can retain the genetic and phenotypic heterogeneity of the tumor tissue.
Collapse
Affiliation(s)
- Guido Gambara
- Charité Comprehensive Cancer Center, Charité - Universitätsmedizin, Berlin, Germany.,German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Manuela Gaebler
- Department of Interdisciplinary Oncology, HELIOS Klinikum Berlin-Buch GmbH, Berlin, Germany
| | - Ulrich Keilholz
- Charité Comprehensive Cancer Center, Charité - Universitätsmedizin, Berlin, Germany.,German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | | | | |
Collapse
|
461
|
Sereti E, Karagianellou T, Kotsoni I, Magouliotis D, Kamposioras K, Ulukaya E, Sakellaridis N, Zacharoulis D, Dimas K. Patient Derived Xenografts (PDX) for personalized treatment of pancreatic cancer: emerging allies in the war on a devastating cancer? J Proteomics 2018; 188:107-118. [PMID: 29398619 DOI: 10.1016/j.jprot.2018.01.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Revised: 01/10/2018] [Accepted: 01/19/2018] [Indexed: 12/14/2022]
Abstract
The prognosis of pancreatic ductal adenocarcinoma (PDAC), the eighth most lethal cancer for men and ninth for women worldwide, remains dismal. The increasing rates of deaths by PDAC indicate that the overall management of the disease in 21st century is still insufficient. Thus it is obvious that there is an unmet need to improve management of PDAC by finding new biomarkers to screen high risk patients, confirm diagnosis, and predict response to treatment as well more efficacious and safer treatments. Patient Derived Xenografts (PDX) have been developed as a new promising tool in an effort to mirror genetics, tumor heterogeneity and cancer microenvironment of the primary tumor. Herein we aim to give an updated overview of the current status and the perspectives of PDX in the search for the identification of novel biomarkers and improved therapeutic outcomes for PDAC but also their use as a valuable tool towards individualized treatments to improve the outcome of the disease. Furthermore, we critically review the applications, advantages, limitations, and perspectives of PDX in the research towards an improved management of PDAC. SIGNIFICANCE This review provides a comprehensive overview of the current status and the potential role as well as the challenges of PDX in the road to fight one of the most lethal cancers in the developed countries, pancreatic ductal adenocarcinoma.
Collapse
Affiliation(s)
- Evangelia Sereti
- Department of Pharmacology, Faculty of Medicine, University of Thessaly, Larissa, Greece
| | | | - Ioanna Kotsoni
- Department of Pharmacology, Faculty of Medicine, University of Thessaly, Larissa, Greece
| | - Dimitrios Magouliotis
- Department of Pharmacology, Faculty of Medicine, University of Thessaly, Larissa, Greece; Department of Surgery, University Hospital of Larissa, Larissa, Greece
| | - Konstantinos Kamposioras
- Department of Medical Oncology, The Mid Yorkshire Hospitals NHS Trust, Wakefield, United Kingdom
| | - Engin Ulukaya
- Istinye University, Faculty of Medicine, Department of Clinical Biochemistry, Istanbul, Turkey
| | - Nikos Sakellaridis
- Department of Pharmacology, Faculty of Medicine, University of Thessaly, Larissa, Greece
| | | | - Konstantinos Dimas
- Department of Pharmacology, Faculty of Medicine, University of Thessaly, Larissa, Greece.
| |
Collapse
|
462
|
Jin MZ, Han RR, Qiu GZ, Ju XC, Lou G, Jin WL. Organoids: An intermediate modeling platform in precision oncology. Cancer Lett 2018; 414:174-180. [PMID: 29174804 DOI: 10.1016/j.canlet.2017.11.021] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2017] [Revised: 11/14/2017] [Accepted: 11/17/2017] [Indexed: 02/06/2023]
Abstract
Cancer harbors variable heterogeneity and plasticity. Thus far, our comprehension is greatly based on cell lines, organoids, and patient-derived tumor xenografts (PDTXs). Organoids are a three-dimensional in vitro culture platform constructed from self-organizing stem cells. They can almost accurately recapitulate tumor heterogeneity and microenvironment "in a dish," which surpass established cell lines and are not as expensive and time-consuming as PDTXs. As an intermediate model, tumor organoids are also used to study the fundamental issues of tumorigenesis and metastasis. They are specifically applied for drug testing and stored as "living biobanks." In this review, we highlight the translational applications of organoid technologies in tumor research and precision medicine, discuss the advantages and limitations compared with other mentioned methods, and provide our outlook on its future.
Collapse
Affiliation(s)
- Ming-Zhu Jin
- Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, PR China
| | - Run-Run Han
- Department of Pulmonary Circulation, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, PR China
| | - Guan-Zhong Qiu
- Department of Neurosurgery, General Hospital of Jinan Military Command, Jinan 250031, PR China
| | - Xiang-Chun Ju
- Institute of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, PR China.
| | - Ge Lou
- Department of Gynecology Oncology, The Tumor Hospital, Harbin Medical University, Harbin, 150086, PR China.
| | - Wei-Lin Jin
- Institute of Nano Biomedicine and Engineering, Shanghai Engineering Center for Intelligent Diagnosis and Treatment Instrument, Department of Instrument Science and Engineering, Key Laboratory for Thin Film and Microfabrication Technology of Ministry of Education, School of Electronic Information and Electronic Engineering, Shanghai Jiao Tong University, Shanghai 200240, PR China; National Center for Translational Medicine, Collaborative Innovational Center for System Biology, Shanghai Jiao Tong University, Shanghai 200240, PR China; Shaanxi Key Laboratory of Brain Disorders and School of Basic Medical Sciences, Xi'an Medical University, Xi'an 710021, PR China.
| |
Collapse
|
463
|
Shue YT, Lim JS, Sage J. Tumor heterogeneity in small cell lung cancer defined and investigated in pre-clinical mouse models. Transl Lung Cancer Res 2018. [PMID: 29535910 DOI: 10.21037/tlcr.2018.01.15] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Small cell lung carcinoma (SCLC) is a fast-growing, highly metastatic form of lung cancer. A major difference between SCLC and other forms of lung cancer is that SCLC tumors often respond well to chemotherapy initially; unfortunately, resistant tumors rapidly recur. In addition, despite a large number of clinical trials with a variety of therapeutic agents, little progress has been achieved in the past three decades in improving the survival of SCLC patients. These clinical observations indicate that SCLC tumors have a high degree of plasticity and rapid adaptability to changes in growth conditions. Here we consider recent evidence pointing to several levels of heterogeneity in SCLC that may explain the ability of these tumors to adjust to different microenvironment and therapeutics. In particular, we review new data pointing to the existence of several subpopulations of tumor cells that interact with each other to promote tumor growth. We also discuss how SCLC tumors that look similar at the histopathological level may actually represent distinct subtypes of tumors and how these differences impact the response to specific therapeutic agents. A better understanding of genetic and cellular heterogeneity will guide the development of personalized approaches to help SCLC patients.
Collapse
Affiliation(s)
- Yan Ting Shue
- Department of Pediatrics, Stanford University, Stanford, CA, USA.,Department of Genetics, Stanford University, Stanford, CA, USA
| | - Jing Shan Lim
- Department of Pediatrics, Stanford University, Stanford, CA, USA.,Department of Genetics, Stanford University, Stanford, CA, USA
| | - Julien Sage
- Department of Pediatrics, Stanford University, Stanford, CA, USA.,Department of Genetics, Stanford University, Stanford, CA, USA
| |
Collapse
|
464
|
Su S, Chen J, Yao H, Liu J, Yu S, Lao L, Wang M, Luo M, Xing Y, Chen F, Huang D, Zhao J, Yang L, Liao D, Su F, Li M, Liu Q, Song E. CD10 +GPR77 + Cancer-Associated Fibroblasts Promote Cancer Formation and Chemoresistance by Sustaining Cancer Stemness. Cell 2018; 172:841-856.e16. [PMID: 29395328 DOI: 10.1016/j.cell.2018.01.009] [Citation(s) in RCA: 849] [Impact Index Per Article: 121.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 12/28/2017] [Accepted: 01/04/2018] [Indexed: 12/12/2022]
Abstract
Carcinoma-associated fibroblasts (CAFs) are abundant and heterogeneous stromal cells in tumor microenvironment that are critically involved in cancer progression. Here, we demonstrate that two cell-surface molecules, CD10 and GPR77, specifically define a CAF subset correlated with chemoresistance and poor survival in multiple cohorts of breast and lung cancer patients. CD10+GPR77+ CAFs promote tumor formation and chemoresistance by providing a survival niche for cancer stem cells (CSCs). Mechanistically, CD10+GPR77+ CAFs are driven by persistent NF-κB activation via p65 phosphorylation and acetylation, which is maintained by complement signaling via GPR77, a C5a receptor. Furthermore, CD10+GPR77+ CAFs promote successful engraftment of patient-derived xenografts (PDXs), and targeting these CAFs with a neutralizing anti-GPR77 antibody abolishes tumor formation and restores tumor chemosensitivity. Our study reveals a functional CAF subset that can be defined and isolated by specific cell-surface markers and suggests that targeting the CD10+GPR77+ CAF subset could be an effective therapeutic strategy against CSC-driven solid tumors.
Collapse
Affiliation(s)
- Shicheng Su
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Jianing Chen
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Herui Yao
- Department of Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Jiang Liu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Shubin Yu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Liyan Lao
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Minghui Wang
- Department of Thoracic Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Manli Luo
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Yue Xing
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Fei Chen
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Di Huang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Jinghua Zhao
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Linbin Yang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Dan Liao
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Fengxi Su
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Mengfeng Li
- Department of Microbiology and Key Laboratory of Tropical Disease Control , Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China
| | - Qiang Liu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Erwei Song
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; Program of Molecular Medicine, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China.
| |
Collapse
|
465
|
Di Zazzo E, Galasso G, Giovannelli P, Di Donato M, Castoria G. Estrogens and Their Receptors in Prostate Cancer: Therapeutic Implications. Front Oncol 2018; 8:2. [PMID: 29404276 PMCID: PMC5778111 DOI: 10.3389/fonc.2018.00002] [Citation(s) in RCA: 88] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 01/04/2018] [Indexed: 12/21/2022] Open
Abstract
A major challenge in clinical management of prostate cancer (PC) is to limit tumor growth and prevent metastatic spreading. Considerable efforts have been made to discover new compounds for PC therapy and recent years have seen promising progress in this field. Pharmacological approaches have been designed to achieve benefits in PC treatment and avoid the negative side effects resulting from administration of antagonists or agonists or new drugs. Nonetheless, the currently available therapies frequently induce resistance and PC progresses toward castration-resistant forms that can be caused by the androgen receptor reactivation and/or mutations, or derangement of signaling pathways. Preclinical and clinical findings have also shown that other nuclear receptors are frequently altered in PC. In this review, we focus on the role of estradiol/estradiol receptor (ER) axis, which controls PC growth and progression. Selective targeting of ER subtypes (α or β) may be an attractive way to limit the growth and spreading of prostatic cancer cells.
Collapse
Affiliation(s)
- Erika Di Zazzo
- Department of Biochemistry, Biophysics and General Pathology, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Giovanni Galasso
- Department of Biochemistry, Biophysics and General Pathology, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Pia Giovannelli
- Department of Biochemistry, Biophysics and General Pathology, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Marzia Di Donato
- Department of Biochemistry, Biophysics and General Pathology, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Gabriella Castoria
- Department of Biochemistry, Biophysics and General Pathology, University of Campania Luigi Vanvitelli, Naples, Italy
| |
Collapse
|
466
|
Jungwirth U, van Weverwijk A, Melake MJ, Chambers AF, Gao Q, Fivaz M, Isacke CM. Generation and characterisation of two D2A1 mammary cancer sublines to model spontaneous and experimental metastasis in a syngeneic BALB/c host. Dis Model Mech 2018; 11:dmm.031740. [PMID: 29208627 PMCID: PMC5818081 DOI: 10.1242/dmm.031740] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Accepted: 11/17/2017] [Indexed: 12/20/2022] Open
Abstract
Studying the complex mechanisms underlying breast cancer metastasis and therapy response necessitates relevant in vivo models, particularly syngeneic models with an intact immune system. Two syngeneic spontaneously metastatic sublines, D2A1-m1 and D2A1-m2, were generated from the poorly metastasising BALB/c-derived D2A1 cell line by serial in vivo passaging. In vivo and in vitro analyses revealed distinct and shared characteristics of the metastatic D2A1-m1 and D2A1-m2 sublines. In particular, D2A1-m1 cells are more aggressive in experimental metastasis assays, while D2A1-m2 cells are more efficient at disseminating from the primary tumour in spontaneous metastasis assays. Surprisingly, classical metastasis-associated in vitro phenotypes, such as enhanced proliferation, migration and invasion, are reduced in the sublines compared to the parental cell line. Further, evasion of immune control cannot fully explain their enhanced metastatic properties. By contrast, both sublines show increased resistance to apoptosis when cultured in non-adherent conditions and, for the D2A1-m2 subline, increased 3D tumour spheroid growth. Moreover, the enhanced spontaneous metastatic phenotype of the D2A1-m2 subline is associated with an increased ability to recruit an activated tumour stroma. The metastatic D2A1-m1 and D2A1-m2 cell lines provide additional syngeneic models for investigating the different steps of the metastatic cascade and thereby represent valuable tools for breast cancer researchers. Finally, this study highlights that morphology and cell behaviour in 2D cell-based assays cannot be used as a reliable predictor of metastatic behaviour in vivo. Summary: We describe two D2A1 mouse mammary cancer sublines with enhanced spontaneous metastasis in a syngeneic host, and highlight the limitations of in vitro assays to predict in vivo metastatic behaviour.
Collapse
Affiliation(s)
- Ute Jungwirth
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, 237 Fulham Road, London, SW3 6JB, UK
| | - Antoinette van Weverwijk
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, 237 Fulham Road, London, SW3 6JB, UK
| | - Miriam J Melake
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, 237 Fulham Road, London, SW3 6JB, UK.,German Cancer Research Center, DKFZ, 69120, Heidelberg, Germany
| | - Ann F Chambers
- Department of Oncology, University of Western Ontario, London, Ontario, N6A 4L6, Canada
| | - Qiong Gao
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, 237 Fulham Road, London, SW3 6JB, UK
| | - Marc Fivaz
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, 237 Fulham Road, London, SW3 6JB, UK
| | - Clare M Isacke
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, 237 Fulham Road, London, SW3 6JB, UK
| |
Collapse
|
467
|
Callari M, Batra AS, Batra RN, Sammut SJ, Greenwood W, Clifford H, Hercus C, Chin SF, Bruna A, Rueda OM, Caldas C. Computational approach to discriminate human and mouse sequences in patient-derived tumour xenografts. BMC Genomics 2018; 19:19. [PMID: 29304755 PMCID: PMC5755132 DOI: 10.1186/s12864-017-4414-y] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Accepted: 12/22/2017] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Patient-Derived Tumour Xenografts (PDTXs) have emerged as the pre-clinical models that best represent clinical tumour diversity and intra-tumour heterogeneity. The molecular characterization of PDTXs using High-Throughput Sequencing (HTS) is essential; however, the presence of mouse stroma is challenging for HTS data analysis. Indeed, the high homology between the two genomes results in a proportion of mouse reads being mapped as human. RESULTS In this study we generated Whole Exome Sequencing (WES), Reduced Representation Bisulfite Sequencing (RRBS) and RNA sequencing (RNA-seq) data from samples with known mixtures of mouse and human DNA or RNA and from a cohort of human breast cancers and their derived PDTXs. We show that using an In silico Combined human-mouse Reference Genome (ICRG) for alignment discriminates between human and mouse reads with up to 99.9% accuracy and decreases the number of false positive somatic mutations caused by misalignment by >99.9%. We also derived a model to estimate the human DNA content in independent PDTX samples. For RNA-seq and RRBS data analysis, the use of the ICRG allows dissecting computationally the transcriptome and methylome of human tumour cells and mouse stroma. In a direct comparison with previously reported approaches, our method showed similar or higher accuracy while requiring significantly less computing time. CONCLUSIONS The computational pipeline we describe here is a valuable tool for the molecular analysis of PDTXs as well as any other mixture of DNA or RNA species.
Collapse
Affiliation(s)
- Maurizio Callari
- CRUK Cambridge Institute and Department of Oncology, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge, CB2 0RE UK
| | - Ankita Sati Batra
- CRUK Cambridge Institute and Department of Oncology, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge, CB2 0RE UK
| | - Rajbir Nath Batra
- CRUK Cambridge Institute and Department of Oncology, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge, CB2 0RE UK
| | - Stephen-John Sammut
- CRUK Cambridge Institute and Department of Oncology, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge, CB2 0RE UK
| | - Wendy Greenwood
- CRUK Cambridge Institute and Department of Oncology, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge, CB2 0RE UK
| | - Harry Clifford
- CRUK Cambridge Institute and Department of Oncology, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge, CB2 0RE UK
| | - Colin Hercus
- Novocraft Technologies Sdn Bhd, C-23A-05, 3 Two Square, Jalan 19/1, Section 19, 46300 Petaling Jaya, Selangor Darul Ehsan Malaysia
| | - Suet-Feung Chin
- CRUK Cambridge Institute and Department of Oncology, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge, CB2 0RE UK
| | - Alejandra Bruna
- CRUK Cambridge Institute and Department of Oncology, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge, CB2 0RE UK
| | - Oscar M. Rueda
- CRUK Cambridge Institute and Department of Oncology, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge, CB2 0RE UK
| | - Carlos Caldas
- CRUK Cambridge Institute and Department of Oncology, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge, CB2 0RE UK
| |
Collapse
|
468
|
Linnekamp JF, Hooff SRV, Prasetyanti PR, Kandimalla R, Buikhuisen JY, Fessler E, Ramesh P, Lee KAST, Bochove GGW, de Jong JH, Cameron K, Leersum RV, Rodermond HM, Franitza M, Nürnberg P, Mangiapane LR, Wang X, Clevers H, Vermeulen L, Stassi G, Medema JP. Consensus molecular subtypes of colorectal cancer are recapitulated in in vitro and in vivo models. Cell Death Differ 2018; 25:616-633. [PMID: 29305587 DOI: 10.1038/s41418-017-0011-5] [Citation(s) in RCA: 135] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 10/02/2017] [Accepted: 10/09/2017] [Indexed: 02/08/2023] Open
Abstract
Colorectal cancer (CRC) is a highly heterogeneous disease both from a molecular and clinical perspective. Several distinct molecular entities, such as microsatellite instability (MSI), have been defined that make up biologically distinct subgroups with their own clinical course. Recent data indicated that CRC can be best segregated into four groups called consensus molecular subtypes (CMS1-4), each of which has a unique biology and gene expression pattern. In order to develop improved, subtype-specific therapies and to gain insight into the molecular wiring and origin of these subtypes, reliable models are needed. This study was designed to determine the heterogeneity and identify the presence of CMSs in a large panel of CRC cell lines, primary cultures and patient-derived xenografts (PDX). We provide a repository encompassing this heterogeneity and moreover describe that a large part of the models can be robustly assigned to one of the four CMSs, independent of the stromal contribution. We subsequently validate our CMS stratification by functional analysis which for instance shows mesenchymal enrichment in CMS4 and metabolic dysregulation in CMS3. Finally, we observe a clear difference in sensitivity to chemotherapy-induced apoptosis, specifically between CMS2 and CMS4. This relates to the in vivo efficacy of chemotherapy, which delays outgrowth of CMS2, but not CMS4 xenografts. Combined our data indicate that molecular subtypes are faithfully modelled in CRC cell cultures and PDXs, representing tumour cell intrinsic and stable features. This repository provides researchers with a platform to study CRC using the existing heterogeneity.
Collapse
Affiliation(s)
- Janneke F Linnekamp
- Cancer Center Amsterdam, Laboratory of Experimental Oncology and Radiobiology (LEXOR), CEMM, Academic Medical Center, Meibergdreef 9, 1105AZ, Amsterdam, The Netherlands.,Cancer Genomics Netherlands, Utrecht, The Netherlands
| | - Sander R van Hooff
- Cancer Center Amsterdam, Laboratory of Experimental Oncology and Radiobiology (LEXOR), CEMM, Academic Medical Center, Meibergdreef 9, 1105AZ, Amsterdam, The Netherlands.,Cancer Genomics Netherlands, Utrecht, The Netherlands
| | - Pramudita R Prasetyanti
- Cancer Center Amsterdam, Laboratory of Experimental Oncology and Radiobiology (LEXOR), CEMM, Academic Medical Center, Meibergdreef 9, 1105AZ, Amsterdam, The Netherlands.,Cancer Genomics Netherlands, Utrecht, The Netherlands
| | - Raju Kandimalla
- Cancer Center Amsterdam, Laboratory of Experimental Oncology and Radiobiology (LEXOR), CEMM, Academic Medical Center, Meibergdreef 9, 1105AZ, Amsterdam, The Netherlands.,Cancer Genomics Netherlands, Utrecht, The Netherlands
| | - Joyce Y Buikhuisen
- Cancer Center Amsterdam, Laboratory of Experimental Oncology and Radiobiology (LEXOR), CEMM, Academic Medical Center, Meibergdreef 9, 1105AZ, Amsterdam, The Netherlands.,Cancer Genomics Netherlands, Utrecht, The Netherlands
| | - Evelyn Fessler
- Cancer Center Amsterdam, Laboratory of Experimental Oncology and Radiobiology (LEXOR), CEMM, Academic Medical Center, Meibergdreef 9, 1105AZ, Amsterdam, The Netherlands.,Cancer Genomics Netherlands, Utrecht, The Netherlands.,Gene Center and Department of Biochemistry, Ludwig-Maximilians-Universität München, Feodor-Lynen-Straße 25, 81377, Munich, Germany
| | - Prashanthi Ramesh
- Cancer Center Amsterdam, Laboratory of Experimental Oncology and Radiobiology (LEXOR), CEMM, Academic Medical Center, Meibergdreef 9, 1105AZ, Amsterdam, The Netherlands.,Cancer Genomics Netherlands, Utrecht, The Netherlands
| | - Kelly A S T Lee
- Cancer Center Amsterdam, Laboratory of Experimental Oncology and Radiobiology (LEXOR), CEMM, Academic Medical Center, Meibergdreef 9, 1105AZ, Amsterdam, The Netherlands.,Cancer Genomics Netherlands, Utrecht, The Netherlands
| | - Grehor G W Bochove
- Cancer Center Amsterdam, Laboratory of Experimental Oncology and Radiobiology (LEXOR), CEMM, Academic Medical Center, Meibergdreef 9, 1105AZ, Amsterdam, The Netherlands.,Cancer Genomics Netherlands, Utrecht, The Netherlands
| | - Johan H de Jong
- Cancer Center Amsterdam, Laboratory of Experimental Oncology and Radiobiology (LEXOR), CEMM, Academic Medical Center, Meibergdreef 9, 1105AZ, Amsterdam, The Netherlands.,Cancer Genomics Netherlands, Utrecht, The Netherlands
| | - Kate Cameron
- Cancer Center Amsterdam, Laboratory of Experimental Oncology and Radiobiology (LEXOR), CEMM, Academic Medical Center, Meibergdreef 9, 1105AZ, Amsterdam, The Netherlands.,Cancer Genomics Netherlands, Utrecht, The Netherlands
| | - Ronald van Leersum
- Cancer Center Amsterdam, Laboratory of Experimental Oncology and Radiobiology (LEXOR), CEMM, Academic Medical Center, Meibergdreef 9, 1105AZ, Amsterdam, The Netherlands.,Cancer Genomics Netherlands, Utrecht, The Netherlands
| | - Hans M Rodermond
- Cancer Center Amsterdam, Laboratory of Experimental Oncology and Radiobiology (LEXOR), CEMM, Academic Medical Center, Meibergdreef 9, 1105AZ, Amsterdam, The Netherlands.,Cancer Genomics Netherlands, Utrecht, The Netherlands
| | - Marek Franitza
- Cologne Center for Genomics (CCG), University of Cologne, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Peter Nürnberg
- Cologne Center for Genomics (CCG), University of Cologne, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Laura R Mangiapane
- Cellular and Molecular Pathophysiology Laboratory, Department of Surgical and Oncological Sciences, University of Palermo, Via del Vespro 131, Palermo, 90134, Italy
| | - Xin Wang
- Department of Biomedical Sciences, City University of Hong Kong, Kowloon Tong, Hong Kong
| | - Hans Clevers
- Cancer Genomics Netherlands, Utrecht, The Netherlands.,Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Centre (UMC), 3584 CT, Utrecht, The Netherlands.,Princess Máxima Centre for Pediatric Oncology, Utrecht, 3584 CT, The Netherlands
| | - Louis Vermeulen
- Cancer Center Amsterdam, Laboratory of Experimental Oncology and Radiobiology (LEXOR), CEMM, Academic Medical Center, Meibergdreef 9, 1105AZ, Amsterdam, The Netherlands
| | - Giorgio Stassi
- Cellular and Molecular Pathophysiology Laboratory, Department of Surgical and Oncological Sciences, University of Palermo, Via del Vespro 131, Palermo, 90134, Italy
| | - Jan Paul Medema
- Cancer Center Amsterdam, Laboratory of Experimental Oncology and Radiobiology (LEXOR), CEMM, Academic Medical Center, Meibergdreef 9, 1105AZ, Amsterdam, The Netherlands. .,Cancer Genomics Netherlands, Utrecht, The Netherlands.
| |
Collapse
|
469
|
Fumagalli A, Suijkerbuijk SJE, Begthel H, Beerling E, Oost KC, Snippert HJ, van Rheenen J, Drost J. A surgical orthotopic organoid transplantation approach in mice to visualize and study colorectal cancer progression. Nat Protoc 2018; 13:235-247. [DOI: 10.1038/nprot.2017.137] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
470
|
Abstract
<b/> A study by Pauli and colleagues in this issue of Cancer Discovery describes the creation of a precision cancer platform for patients with advanced disease, integrating DNA sequencing of patient tumors with the generation of patient-derived organoids and xenografts. They propose the use of this platform for drug testing to nominate therapeutic options for individual patients and for therapeutic biomarker discovery. Cancer Discov; 7(5); 456-8. ©2017 AACRSee related article by Pauli et al., p. 462.
Collapse
Affiliation(s)
- Gabriele Picco
- Wellcome Trust Sanger Institute, Cambridge, United Kingdom
| | | |
Collapse
|
471
|
Okada S, Vaeteewoottacharn K, Kariya R. Establishment of a Patient-Derived Tumor Xenograft Model and Application for Precision Cancer Medicine. Chem Pharm Bull (Tokyo) 2018; 66:225-230. [PMID: 29491256 DOI: 10.1248/cpb.c17-00789] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2024]
Abstract
Patient-derived xenograft (PDX) models can be created with the transplantation of cancerous cells or tissues from patients' primary tumors into immunodeficient mice. PDXs are now in the spotlight as more accurate human cancer models compared with mouse tumor and human cancer cell lines transplanted into mice. PDX technology leads to breakthroughs with the introduction of novel, highly immunodeficient mice such as NOG (NOD/Scid/IL2Rγnull), NSG (NOD/Scid/IL2Rγnull), and NOJ (NOD/Scid/Jak3null) mice. Xenograft efficiency differs by type of tumor, site of implantation, and tumor aggressiveness. Subcutaneous implantation is a standard method for PDX, and renal capsule or orthotropic implantation improves the efficiency. Despite positive test results in animal cancer models, significant numbers of novel drug candidates fail in clinical trials because conventional animal models such as murine tumor and human cancer cell line transplantation models do not always reflect the nature of human cancers. Since PDXs conserve the original tumor characteristics such as heterogeneous histology, clinical biomolecular signatures, malignant phenotypes and genotypes, tumor architecture, and tumor vasculature, they are currently believed to offer relevant predictive insights into clinical outcomes when evaluating the efficacy of novel cancer therapies. PDX banks with integrated genomic signatures are now established in many organizations including pharmaceutical companies. These PDX databases are becoming powerful tools for advancing precision cancer medicine.
Collapse
Affiliation(s)
- Seiji Okada
- Center for AIDS Resesarch, Kumamoto University
| | - Kulthida Vaeteewoottacharn
- Center for AIDS Resesarch, Kumamoto University
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University
| | | |
Collapse
|
472
|
Sulaiman A, Wang L. Bridging the divide: preclinical research discrepancies between triple-negative breast cancer cell lines and patient tumors. Oncotarget 2017; 8:113269-113281. [PMID: 29348905 PMCID: PMC5762590 DOI: 10.18632/oncotarget.22916] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Accepted: 11/13/2017] [Indexed: 12/12/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is the most refractory subtype of breast cancer and disproportionately accounts for the majority of breast cancer related deaths. Effective treatment of this disease remains an unmet medical need. Over the past several decades, TNBC cell lines have been used as the foundation for drug development and disease modeling. However, ever-mounting research demonstrates striking differences between cell lines and clinical TNBC tumors, disconnecting bench research and actual clinical responses. In this review, we discuss the limitations of cell lines and the importance of using patients' tumors for translational research, and highlight the usage of patient-derived xenograft (PDXs) models that have emerged as a clinically relevant platform for preclinical studies. PDX tumors possess tumor heterogeneity with similar cellular, molecular, genetic and epigenetic properties akin to those found within patients' tumors. Moreover, PDX and clinical tumors possess abnormal vasculature with higher blood vessel permeability, a feature that is not always demonstrated in in vivo cell line xenografts. Development of clinically relevant, novel drug-nanoparticles capable of accumulating in PDX tumors through the enhanced permeability and retention effect in tumor vasculature may lead to new and effective TNBC treatments.
Collapse
Affiliation(s)
- Andrew Sulaiman
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
- China-Canada Centre of Research for Digestive Diseases, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| | - Lisheng Wang
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
- China-Canada Centre of Research for Digestive Diseases, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, Ontario K1H 8L6, Canada
- Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| |
Collapse
|
473
|
Vincan E, Schwab RHM, Flanagan DJ, Moselen JM, Tran BM, Barker N, Phesse TJ. The Central Role of Wnt Signaling and Organoid Technology in Personalizing Anticancer Therapy. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2017; 153:299-319. [PMID: 29389521 DOI: 10.1016/bs.pmbts.2017.11.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The Wnt pathway is at the heart of organoid technology, which is set to revolutionize the cancer field. We can now predetermine a patient's response to any given anticancer therapy by exposing tumor organoids established from the patient's own tumor. This cutting-edge biomedical platform translates to patients being treated with the correct drug at the correct dose from the outset, a truly personalized and precise medical approach. A high throughput drug screen on organoids also allows drugs to be tested in limitless combinations. More recently, the tumor cells that are resistant to the therapy given to a patient were selected in culture using the patient's organoids. The resistant tumor organoids were then screened empirically to identify drugs that will kill the resistant cells. This information allows diagnosis in real-time to either prevent tumor recurrence or effectively treat the recurring tumor. Furthermore, the ability to culture stem cell-derived epithelium as organoids has enabled us to begin to understand how a stem cell becomes a cancer cell or to pin-point the genetic alteration that underlies a given genetic syndrome. Here we summarize these advances and the central role of Wnt signaling, and identify the next challenges for organoid technology.
Collapse
Affiliation(s)
- Elizabeth Vincan
- Doherty Institute of Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia; Curtin University, Perth, WA, Australia.
| | - Renate H M Schwab
- Doherty Institute of Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| | - Dustin J Flanagan
- Doherty Institute of Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| | - Jean M Moselen
- Doherty Institute of Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| | - Bang M Tran
- Doherty Institute of Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| | - Nick Barker
- A*STAR Institute of Medical Biology, Singapore
| | - Toby J Phesse
- Doherty Institute of Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia; European Cancer Stem Cell Research Institute, Cardiff University, Cardiff, United Kingdom
| |
Collapse
|
474
|
Yu J, Qin B, Moyer AM, Sinnwell JP, Thompson KJ, Copland JA, Marlow LA, Miller JL, Yin P, Gao B, Minter-Dykhouse K, Tang X, McLaughlin SA, Moreno-Aspitia A, Schweitzer A, Lu Y, Hubbard J, Northfelt DW, Gray RJ, Hunt K, Conners AL, Suman VJ, Kalari KR, Ingle JN, Lou Z, Visscher DW, Weinshilboum R, Boughey JC, Goetz MP, Wang L. Establishing and characterizing patient-derived xenografts using pre-chemotherapy percutaneous biopsy and post-chemotherapy surgical samples from a prospective neoadjuvant breast cancer study. Breast Cancer Res 2017; 19:130. [PMID: 29212525 PMCID: PMC5719923 DOI: 10.1186/s13058-017-0920-8] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 11/15/2017] [Indexed: 02/06/2023] Open
Abstract
Background Patient-derived xenografts (PDXs) are increasingly used in cancer research as a tool to inform cancer biology and drug response. Most available breast cancer PDXs have been generated in the metastatic setting. However, in the setting of operable breast cancer, PDX models both sensitive and resistant to chemotherapy are needed for drug development and prospective data are lacking regarding the clinical and molecular characteristics associated with PDX take rate in this setting. Methods The Breast Cancer Genome Guided Therapy Study (BEAUTY) is a prospective neoadjuvant chemotherapy (NAC) trial of stage I-III breast cancer patients treated with neoadjuvant weekly taxane+/-trastuzumab followed by anthracycline-based chemotherapy. Using percutaneous tumor biopsies (PTB), we established and characterized PDXs from both primary (untreated) and residual (treated) tumors. Tumor take rate was defined as percent of patients with the development of at least one stably transplantable (passed at least for four generations) xenograft that was pathologically confirmed as breast cancer. Results Baseline PTB samples from 113 women were implanted with an overall take rate of 27.4% (31/113). By clinical subtype, the take rate was 51.3% (20/39) in triple negative (TN) breast cancer, 26.5% (9/34) in HER2+, 5.0% (2/40) in luminal B and 0% (0/3) in luminal A. The take rate for those with pCR did not differ from those with residual disease in TN (p = 0.999) and HER2+ (p = 0.2401) tumors. The xenografts from 28 of these 31 patients were such that at least one of the xenografts generated had the same molecular subtype as the patient. Among the 35 patients with residual tumor after NAC adequate for implantation, the take rate was 17.1%. PDX response to paclitaxel mirrored the patients’ clinical response in all eight PDX tested. Conclusions The generation of PDX models both sensitive and resistant to standard NAC is feasible and these models exhibit similar biological and drug response characteristics as the patients’ primary tumors. Taken together, these models may be useful for biomarker discovery and future drug development. Electronic supplementary material The online version of this article (doi:10.1186/s13058-017-0920-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jia Yu
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Bo Qin
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA.,Department of Oncology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Ann M Moyer
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Jason P Sinnwell
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, 55905, USA
| | - Kevin J Thompson
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, 55905, USA
| | - John A Copland
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Laura A Marlow
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - James L Miller
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Ping Yin
- Department of Oncology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Bowen Gao
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | | | - Xiaojia Tang
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, 55905, USA
| | | | | | - Anthony Schweitzer
- Affymetrix, now part of Thermo Fisher Scientific, Santa Clara, CA, 95051, USA
| | - Yan Lu
- Affymetrix, now part of Thermo Fisher Scientific, Santa Clara, CA, 95051, USA
| | - Jason Hubbard
- Affymetrix, now part of Thermo Fisher Scientific, Santa Clara, CA, 95051, USA
| | - Donald W Northfelt
- Department of Hematology/Oncology, Mayo Clinic, Scottsdale, AZ, 85259, USA
| | - Richard J Gray
- Department of Surgery, Mayo Clinic, Scottsdale, AZ, 85259, USA
| | - Katie Hunt
- Department of Radiology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Amy L Conners
- Department of Radiology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Vera J Suman
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, 55905, USA
| | - Krishna R Kalari
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, 55905, USA
| | - James N Ingle
- Department of Oncology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Zhenkun Lou
- Department of Oncology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Daniel W Visscher
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Richard Weinshilboum
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Judy C Boughey
- Department of Surgery, Mayo Clinic, Rochester, MN, 55905, USA
| | - Matthew P Goetz
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA.,Department of Oncology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Liewei Wang
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA.
| |
Collapse
|
475
|
Gengenbacher N, Singhal M, Augustin HG. Preclinical mouse solid tumour models: status quo, challenges and perspectives. Nat Rev Cancer 2017; 17:751-765. [PMID: 29077691 DOI: 10.1038/nrc.2017.92] [Citation(s) in RCA: 214] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Oncology research in humans is limited to analytical and observational studies for obvious ethical reasons, with therapy-focused clinical trials being the one exception to this rule. Preclinical mouse tumour models therefore serve as an indispensable intermediate experimental model system bridging more reductionist in vitro research with human studies. Based on a systematic survey of preclinical mouse tumour studies published in eight scientific journals in 2016, this Analysis provides an overview of how contemporary preclinical mouse tumour biology research is pursued. It thereby identifies some of the most important challenges in this field and discusses potential ways in which preclinical mouse tumour models could be improved for better relevance, reproducibility and translatability.
Collapse
Affiliation(s)
- Nicolas Gengenbacher
- Division of Vascular Oncology and Metastasis, German Cancer Research Center (DKFZ-ZMBH Alliance), 69120 Heidelberg, Germany
| | - Mahak Singhal
- Division of Vascular Oncology and Metastasis, German Cancer Research Center (DKFZ-ZMBH Alliance), 69120 Heidelberg, Germany
- Department of Vascular Biology and Tumor Angiogenesis (CBTM), Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Hellmut G Augustin
- Division of Vascular Oncology and Metastasis, German Cancer Research Center (DKFZ-ZMBH Alliance), 69120 Heidelberg, Germany
- Department of Vascular Biology and Tumor Angiogenesis (CBTM), Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
- German Cancer Consortium, 69120 Heidelberg, Germany
| |
Collapse
|
476
|
Sakumoto M, Oyama R, Takahashi M, Takai Y, Kito F, Shiozawa K, Qiao Z, Endo M, Yoshida A, Kawai A, Kondo T. Establishment and proteomic characterization of patient-derived clear cell sarcoma xenografts and cell lines. In Vitro Cell Dev Biol Anim 2017; 54:163-176. [PMID: 29197033 DOI: 10.1007/s11626-017-0207-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Accepted: 10/10/2017] [Indexed: 01/11/2023]
Abstract
Clear cell sarcoma (CCS) is an aggressive mesenchymal malignancy characterized by the unique chimeric EWS-ATF1 fusion gene. Patient-derived cancer models are essential tools for the understanding of tumorigenesis and the development of anti-cancer drugs; however, only a limited number of CCS cell lines exist. The objective of this study was to establish patient-derived CCS models. We established patient-derived CCS models from a 43-yr-old female patient. We prepared the patient-derived xenografts (PDXs) from tumor tissues obtained through biopsy or surgery and isolated stable cell lines from PDXs and the original tumor tissue. The presence of gene fusions was examined by RT-PCR, and Sanger sequencing. The established cell lines were characterized by short tandem repeat, viability, colony and spheroid formation, and invasion analyses. Differences in gene enrichment between the primary tumor and cell lines were examined by mass spectrometry and KEGG pathway analysis. The cell lines were maintained for more than 80 passages, and had tumorigenic characteristics such as colony and spheroid formation and invasion. Mass spectrometric proteome analysis demonstrated that the cell lines were enriched for similar but distinct molecular pathways, compared to those in the xenografts and original tumor tissue. Next, tyrosine kinase inhibitors were screened for their suppressive effects on viability. We found that ponatinib, vandetanib, and doxorubicin suppressed the growth of cell lines, and had equivalent IC50 values. Further in-depth investigation and understanding of drug-sensitivity mechanisms will be important for the clinical applications of our cell lines.
Collapse
Affiliation(s)
- Marimu Sakumoto
- Department of Innovative Seeds Evaluation, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Rieko Oyama
- Department of Innovative Seeds Evaluation, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Mami Takahashi
- Central Animal Division, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Yoko Takai
- Department of Innovative Seeds Evaluation, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Fusako Kito
- Department of Innovative Seeds Evaluation, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Kumiko Shiozawa
- Division of Rare Cancer Research, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Zhiwei Qiao
- Division of Rare Cancer Research, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Makoto Endo
- Division of Musculoskeletal Oncology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Akihiko Yoshida
- Department of Pathology and Clinical Laboratories, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Akira Kawai
- Division of Musculoskeletal Oncology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Tadashi Kondo
- Department of Innovative Seeds Evaluation, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan. .,Division of Rare Cancer Research, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan.
| |
Collapse
|
477
|
Li X, Zhu D, Li N, Yang H, Zhao Z, Li M. Characterization of ascites-derived tumor cells from an endometrial cancer patient. Cancer Sci 2017; 108:2352-2357. [PMID: 28945304 PMCID: PMC5715242 DOI: 10.1111/cas.13407] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 09/14/2017] [Accepted: 09/19/2017] [Indexed: 12/13/2022] Open
Abstract
Improved treatment outcomes for the endometrial cancer patient requires precision methods to investigate the biology of this disease and clinically relevant models to test treatment drugs. Hence, we applied a personalized platform to investigate whether in vitro and in vivo models could accurately predict effective treatment regimens. We successfully expanded ascites-derived tumor cells from an endometrial cancer patient with malignant ascites using ascites collected prior to chemotherapy treatment. Hematoxylin-eosin and immunohistochemistry staining of ascites-derived tumor cells confirmed the source of endometrial cancer cells. Ascites-derived tumor cells were sensitive to cisplatin and doxorubicin single-agent treatments in CCK-8 assay and 3-D culture, a condition that more closely mimics the in vivo environment. We further showed that ascites-derived tumor cells from this patient could form tumors in NOD/SCID mice with preserved morphological characteristics. A remarkable concordance between the clinical response of cisplatin and the results of in vitro and in vivo drug tests reflected the reliability of our personalized approach in this case. Together, our results indicated that an effective platform for ex vivo and in vivo culture of ascites-derived tumor cells from our endometrial cancer patient could be applied to identify treatment options, and may be commonly used in treating cancer patients with malignant ascites in the future.
Collapse
Affiliation(s)
- Xuelu Li
- Department of Breast Surgery and OncologyThe Second Hospital of Dalian Medical UniversityDalianChina
- Department of Critical Care MedicineThe Second Hospital of Dalian Medical UniversityDalianChina
- Department of Foreign LanguageDalian Medical UniversityDalianChina
| | - Dandan Zhu
- Department of Critical Care MedicineThe Second Hospital of Dalian Medical UniversityDalianChina
| | - Ning Li
- Department of Foreign LanguageDalian Medical UniversityDalianChina
| | - Hui Yang
- Department of Foreign LanguageDalian Medical UniversityDalianChina
| | - Zuowei Zhao
- Department of Breast Surgery and OncologyThe Second Hospital of Dalian Medical UniversityDalianChina
| | - Man Li
- Department of Breast Surgery and OncologyThe Second Hospital of Dalian Medical UniversityDalianChina
| |
Collapse
|
478
|
Bräunlein E, Krackhardt AM. Identification and Characterization of Neoantigens As Well As Respective Immune Responses in Cancer Patients. Front Immunol 2017; 8:1702. [PMID: 29250075 PMCID: PMC5714868 DOI: 10.3389/fimmu.2017.01702] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Accepted: 11/17/2017] [Indexed: 12/16/2022] Open
Abstract
Cancer immunotherapy has recently emerged as a powerful tool for the treatment of diverse advanced malignancies. In particular, therapeutic application of immune checkpoint modulators, such as anti-CTLA4 or anti-PD-1/PD-L1 antibodies, have shown efficacy in a broad range of malignant diseases. Although pharmacodynamics of these immune modulators are complex, recent studies strongly support the notion that altered peptide ligands presented on tumor cells representing neoantigens may play an essential role in tumor rejection by T cells activated by anti-CTLA4 and anti-PD-1 antibodies. Neoantigens may have diverse sources as viral and mutated proteins. Moreover, posttranslational modifications and altered antigen processing may also contribute to the neoantigenic peptide ligand landscape. Different approaches of target identification are currently applied in combination with subsequent characterization of autologous and non-self T-cell responses against such neoantigens. Additional efforts are required to elucidate key characteristics and interdependences of neoantigens, immunodominance, respective T-cell responses, and the tumor microenvironment in order to define decisive determinants involved in effective T-cell-mediated tumor rejection. This review focuses on our current knowledge of identification and characterization of such neoantigens as well as respective T-cell responses. It closes with challenges to be addressed in future relevant for further improvement of immunotherapeutic strategies in malignant diseases.
Collapse
Affiliation(s)
- Eva Bräunlein
- Medizinische Klinik III, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Angela M Krackhardt
- Medizinische Klinik III, Klinikum rechts der Isar, Technische Universität München, Munich, Germany.,German Cancer Consortium of Translational Cancer Research (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
479
|
Krepler C, Sproesser K, Brafford P, Beqiri M, Garman B, Xiao M, Shannan B, Watters A, Perego M, Zhang G, Vultur A, Yin X, Liu Q, Anastopoulos IN, Wubbenhorst B, Wilson MA, Xu W, Karakousis G, Feldman M, Xu X, Amaravadi R, Gangadhar TC, Elder DE, Haydu LE, Wargo JA, Davies MA, Lu Y, Mills GB, Frederick DT, Barzily-Rokni M, Flaherty KT, Hoon DS, Guarino M, Bennett JJ, Ryan RW, Petrelli NJ, Shields CL, Terai M, Sato T, Aplin AE, Roesch A, Darr D, Angus S, Kumar R, Halilovic E, Caponigro G, Jeay S, Wuerthner J, Walter A, Ocker M, Boxer MB, Schuchter L, Nathanson KL, Herlyn M. A Comprehensive Patient-Derived Xenograft Collection Representing the Heterogeneity of Melanoma. Cell Rep 2017; 21:1953-1967. [PMID: 29141225 PMCID: PMC5726788 DOI: 10.1016/j.celrep.2017.10.021] [Citation(s) in RCA: 107] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Revised: 08/18/2017] [Accepted: 10/04/2017] [Indexed: 11/22/2022] Open
Abstract
Therapy of advanced melanoma is changing dramatically. Following mutational and biological subclassification of this heterogeneous cancer, several targeted and immune therapies were approved and increased survival significantly. To facilitate further advancements through pre-clinical in vivo modeling, we have established 459 patient-derived xenografts (PDX) and live tissue samples from 384 patients representing the full spectrum of clinical, therapeutic, mutational, and biological heterogeneity of melanoma. PDX have been characterized using targeted sequencing and protein arrays and are clinically annotated. This exhaustive live tissue resource includes PDX from 57 samples resistant to targeted therapy, 61 samples from responders and non-responders to immune checkpoint blockade, and 31 samples from brain metastasis. Uveal, mucosal, and acral subtypes are represented as well. We show examples of pre-clinical trials that highlight how the PDX collection can be used to develop and optimize precision therapies, biomarkers of response, and the targeting of rare genetic subgroups.
Collapse
Affiliation(s)
- Clemens Krepler
- Molecular and Cellular Oncogenesis Program, Wistar Institute, Philadelphia, PA 19104, USA
| | - Katrin Sproesser
- Molecular and Cellular Oncogenesis Program, Wistar Institute, Philadelphia, PA 19104, USA
| | - Patricia Brafford
- Molecular and Cellular Oncogenesis Program, Wistar Institute, Philadelphia, PA 19104, USA
| | - Marilda Beqiri
- Molecular and Cellular Oncogenesis Program, Wistar Institute, Philadelphia, PA 19104, USA
| | - Bradley Garman
- Department of Medicine, Division of Translational Medicine and Human Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Min Xiao
- Molecular and Cellular Oncogenesis Program, Wistar Institute, Philadelphia, PA 19104, USA
| | - Batool Shannan
- Molecular and Cellular Oncogenesis Program, Wistar Institute, Philadelphia, PA 19104, USA
| | - Andrea Watters
- Molecular and Cellular Oncogenesis Program, Wistar Institute, Philadelphia, PA 19104, USA
| | - Michela Perego
- Molecular and Cellular Oncogenesis Program, Wistar Institute, Philadelphia, PA 19104, USA
| | - Gao Zhang
- Molecular and Cellular Oncogenesis Program, Wistar Institute, Philadelphia, PA 19104, USA
| | - Adina Vultur
- Molecular and Cellular Oncogenesis Program, Wistar Institute, Philadelphia, PA 19104, USA
| | - Xiangfan Yin
- Molecular and Cellular Oncogenesis Program, Wistar Institute, Philadelphia, PA 19104, USA
| | - Qin Liu
- Molecular and Cellular Oncogenesis Program, Wistar Institute, Philadelphia, PA 19104, USA
| | - Ioannis N Anastopoulos
- Department of Medicine, Division of Translational Medicine and Human Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Bradley Wubbenhorst
- Department of Medicine, Division of Translational Medicine and Human Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Melissa A Wilson
- Department of Medicine, Division of Translational Medicine and Human Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Wei Xu
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Giorgos Karakousis
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Michael Feldman
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Xiaowei Xu
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ravi Amaravadi
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Tara C Gangadhar
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - David E Elder
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Lauren E Haydu
- MD Anderson Cancer Center, University of Texas, Houston, TX 77030, USA
| | - Jennifer A Wargo
- MD Anderson Cancer Center, University of Texas, Houston, TX 77030, USA
| | - Michael A Davies
- MD Anderson Cancer Center, University of Texas, Houston, TX 77030, USA
| | - Yiling Lu
- MD Anderson Cancer Center, University of Texas, Houston, TX 77030, USA
| | - Gordon B Mills
- MD Anderson Cancer Center, University of Texas, Houston, TX 77030, USA
| | | | | | - Keith T Flaherty
- Massachusetts General Hospital Cancer Center, Boston, MA 02114, USA
| | - Dave S Hoon
- Translational Molecular Medicine, John Wayne Cancer Institute, Santa Monica, CA 90404, USA
| | - Michael Guarino
- Helen F. Graham Cancer Center at Christiana Care, Newark, DE 19713, USA
| | - Joseph J Bennett
- Helen F. Graham Cancer Center at Christiana Care, Newark, DE 19713, USA
| | - Randall W Ryan
- Helen F. Graham Cancer Center at Christiana Care, Newark, DE 19713, USA
| | | | - Carol L Shields
- Ocular Oncology Service, Wills Eye Hospital, Philadelphia, PA 19107, USA
| | - Mizue Terai
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107
| | - Takami Sato
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107
| | - Andrew E Aplin
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107
| | - Alexander Roesch
- Department of Dermatology, University Duisburg-Essen, University Hospital Essen, 45147 Essen, Germany; German Consortium of Translational Cancer Research, Heidelberg, Germany
| | - David Darr
- Lineberger Cancer Center, University of North Carolina Chapel Hill, NC 27514, USA
| | - Steve Angus
- Lineberger Cancer Center, University of North Carolina Chapel Hill, NC 27514, USA
| | | | - Ensar Halilovic
- Novartis Institutes for Biomedical Research, Cambridge, MA 02139, USA
| | | | - Sebastien Jeay
- Novartis Institutes for Biomedical Research, Cambridge, MA 02139, USA
| | - Jens Wuerthner
- Novartis Institutes for Biomedical Research, Cambridge, MA 02139, USA
| | | | | | - Matthew B Boxer
- National Center for Advancing Translational Sciences, NIH, Rockville, MD 20850, USA
| | - Lynn Schuchter
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Katherine L Nathanson
- Department of Medicine, Division of Translational Medicine and Human Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Meenhard Herlyn
- Molecular and Cellular Oncogenesis Program, Wistar Institute, Philadelphia, PA 19104, USA.
| |
Collapse
|
480
|
Meehan TF, Conte N, Goldstein T, Inghirami G, Murakami MA, Brabetz S, Gu Z, Wiser JA, Dunn P, Begley DA, Krupke DM, Bertotti A, Bruna A, Brush MH, Byrne AT, Caldas C, Christie AL, Clark DA, Dowst H, Dry JR, Doroshow JH, Duchamp O, Evrard YA, Ferretti S, Frese KK, Goodwin NC, Greenawalt D, Haendel MA, Hermans E, Houghton PJ, Jonkers J, Kemper K, Khor TO, Lewis MT, Lloyd KCK, Mason J, Medico E, Neuhauser SB, Olson JM, Peeper DS, Rueda OM, Seong JK, Trusolino L, Vinolo E, Wechsler-Reya RJ, Weinstock DM, Welm A, Weroha SJ, Amant F, Pfister SM, Kool M, Parkinson H, Butte AJ, Bult CJ. PDX-MI: Minimal Information for Patient-Derived Tumor Xenograft Models. Cancer Res 2017; 77:e62-e66. [PMID: 29092942 PMCID: PMC5738926 DOI: 10.1158/0008-5472.can-17-0582] [Citation(s) in RCA: 95] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 04/20/2017] [Accepted: 08/25/2017] [Indexed: 11/16/2022]
Abstract
Patient-derived tumor xenograft (PDX) mouse models have emerged as an important oncology research platform to study tumor evolution, mechanisms of drug response and resistance, and tailoring chemotherapeutic approaches for individual patients. The lack of robust standards for reporting on PDX models has hampered the ability of researchers to find relevant PDX models and associated data. Here we present the PDX models minimal information standard (PDX-MI) for reporting on the generation, quality assurance, and use of PDX models. PDX-MI defines the minimal information for describing the clinical attributes of a patient's tumor, the processes of implantation and passaging of tumors in a host mouse strain, quality assurance methods, and the use of PDX models in cancer research. Adherence to PDX-MI standards will facilitate accurate search results for oncology models and their associated data across distributed repository databases and promote reproducibility in research studies using these models. Cancer Res; 77(21); e62-66. ©2017 AACR.
Collapse
Affiliation(s)
- Terrence F Meehan
- European Molecular Biology Laboratory-European Bioinformatics Institute, Hinxton, United Kingdom.
| | - Nathalie Conte
- European Molecular Biology Laboratory-European Bioinformatics Institute, Hinxton, United Kingdom
| | - Theodore Goldstein
- Institute for Computational Health Sciences, University of California, San Francisco, California
| | - Giorgio Inghirami
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York
| | - Mark A Murakami
- Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| | - Sebastian Brabetz
- Hopp-Children's Cancer Center at the NCT Heidelberg (KiTZ), Heidelberg, Germany
- Division of Pediatric Neuro-oncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Zhiping Gu
- Northrop Grumman Information Systems Health IT, Rockville, Maryland
| | - Jeffrey A Wiser
- Northrop Grumman Information Systems Health IT, Rockville, Maryland
| | - Patrick Dunn
- Northrop Grumman Information Systems Health IT, Rockville, Maryland
| | | | | | - Andrea Bertotti
- Candiolo Cancer Institute, FPO-IRCC, Department of Oncology, University of Torino, Torino, Italy
| | - Alejandra Bruna
- Cancer Research UK Cambridge Institute, Cambridge Cancer Centre, University of Cambridge, Cambridge, United Kingdom
| | - Matthew H Brush
- Department of Medical Informatics and Clinical Epidemiology and OHSU Library, Oregon Health and Science University, Portland, Oregon
| | | | - Carlos Caldas
- Cancer Research UK Cambridge Institute, Cambridge Cancer Centre, University of Cambridge, Cambridge, United Kingdom
| | - Amanda L Christie
- Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| | - Dominic A Clark
- European Molecular Biology Laboratory-European Bioinformatics Institute, Hinxton, United Kingdom
| | - Heidi Dowst
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, Texas
| | - Jonathan R Dry
- Oncology Innovative Medicines and Early Development, AstraZeneca R&D Boston, Waltham, Massachusetts
| | - James H Doroshow
- Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | | | - Yvonne A Evrard
- Leidos Biomedical Research, Inc, Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Stephane Ferretti
- Oncology Disease Area, Novartis Institutes for Biomedical Research, Switzerland
| | - Kristopher K Frese
- Cancer Research UK Manchester Institute, The University of Manchester, Manchester, United Kingdom
| | | | | | - Melissa A Haendel
- Department of Medical Informatics and Clinical Epidemiology and OHSU Library, Oregon Health and Science University, Portland, Oregon
| | - Els Hermans
- Katholieke Universiteit Leuven, Leuven, Belgium
| | - Peter J Houghton
- Greehey Children's Cancer Research Institute, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Jos Jonkers
- The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Kristel Kemper
- The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Tin O Khor
- Institute for Applied Cancer Science, Center for Co-Clinical Trial, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Michael T Lewis
- The Lester and Sue Smith Breast Center, Departments of Molecular and Cellular Biology and Radiology, Baylor College of Medicine, Houston, Texas
| | - K C Kent Lloyd
- Department of Surgery, School of Medicine, and Mouse Biology Program, University of California Davis, Davis, California
| | - Jeremy Mason
- European Molecular Biology Laboratory-European Bioinformatics Institute, Hinxton, United Kingdom
| | - Enzo Medico
- Candiolo Cancer Institute, FPO-IRCC, Department of Oncology, University of Torino, Torino, Italy
| | | | - James M Olson
- Fred Hutchinson Cancer Research Center, Seattle Children's Hospital, Seattle, Washington
| | - Daniel S Peeper
- The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Oscar M Rueda
- Cancer Research UK Cambridge Institute, Cambridge Cancer Centre, University of Cambridge, Cambridge, United Kingdom
| | - Je Kyung Seong
- Research Institute for Veterinary Science and Korea Mouse Phenotyping Center, Seoul, Republic of Korea
| | - Livio Trusolino
- Candiolo Cancer Institute, FPO-IRCC, Department of Oncology, University of Torino, Torino, Italy
| | | | - Robert J Wechsler-Reya
- Tumor Initiation and Maintenance Program, NCI-Designated Cancer Center, La Jolla, California
| | - David M Weinstock
- Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| | - Alana Welm
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah
| | - S John Weroha
- Department of Oncology, Mayo Clinic College of Medicine, Rochester, Minnesota
| | - Frédéric Amant
- The Netherlands Cancer Institute, Amsterdam, the Netherlands
- University of Leuven, Leuven, Belgium
| | - Stefan M Pfister
- Hopp-Children's Cancer Center at the NCT Heidelberg (KiTZ), Heidelberg, Germany
- Division of Pediatric Neuro-oncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany
- Department of Pediatric Hematology and Oncology, Heidelberg University Hospital, Heidelberg, Germany
| | - Marcel Kool
- Hopp-Children's Cancer Center at the NCT Heidelberg (KiTZ), Heidelberg, Germany
- Division of Pediatric Neuro-oncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Helen Parkinson
- European Molecular Biology Laboratory-European Bioinformatics Institute, Hinxton, United Kingdom
| | - Atul J Butte
- Institute for Computational Health Sciences, University of California, San Francisco, California
| | | |
Collapse
|
481
|
Abstract
INTRODUCTION Proteomics has been used in soft tissue sarcoma (STS) research in the attempts to improve the understanding of the disease background and develop novel clinical applications. Using various proteomics modalities, aberrant regulations of numerous intriguing proteins were identified in STSs, and the possible utilities of identified proteins as biomarkers or therapeutic targets have been explored. STS is an exceptionally diverse group of malignant diseases with highly complex molecular backgrounds and, therefore, an overview of the achievements and prospects of STS proteomics could enhance our knowledge of the possibilities and limitations of cancer proteomics. Areas covered: This review examines all STSs that have been examined using proteomics modalities, discussing unique aspects, limitations, and possible improvements of individual reports. To contribute to the current progress in cancer treatment development using novel anti-cancer drugs, proteomics plays a central role in linking cutting-edge technologies, application of proteogenomics, patient-derived cancer models, and biobanking system. Expert commentary: Therefore, proteomic-based STS research will be developed as an interdisciplinary science. STS proteomics will be further developed based on the interaction of oncologists with basic researchers in various fields, aimed at obtaining an enhanced understanding of the biology of the disease and achieving superior clinical outcomes for patients.
Collapse
Affiliation(s)
- Tadashi Kondo
- a Division of Rare Cancer Research , National Cancer Center Research Institute , Tokyo , Japan
| |
Collapse
|
482
|
Venetsanakos E, Brameld KA, Phan VT, Verner E, Owens TD, Xing Y, Tam D, LaStant J, Leung K, Karr DE, Hill RJ, Gerritsen ME, Goldstein DM, Funk JO, Bradshaw JM. The Irreversible Covalent Fibroblast Growth Factor Receptor Inhibitor PRN1371 Exhibits Sustained Inhibition of FGFR after Drug Clearance. Mol Cancer Ther 2017; 16:2668-2676. [DOI: 10.1158/1535-7163.mct-17-0309] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Revised: 07/11/2017] [Accepted: 09/22/2017] [Indexed: 11/16/2022]
|
483
|
Luraghi P, Bigatto V, Cipriano E, Reato G, Orzan F, Sassi F, De Bacco F, Isella C, Bellomo SE, Medico E, Comoglio PM, Bertotti A, Trusolino L, Boccaccio C. A Molecularly Annotated Model of Patient-Derived Colon Cancer Stem-Like Cells to Assess Genetic and Nongenetic Mechanisms of Resistance to Anti-EGFR Therapy. Clin Cancer Res 2017; 24:807-820. [PMID: 28974546 DOI: 10.1158/1078-0432.ccr-17-2151] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Revised: 09/14/2017] [Accepted: 09/27/2017] [Indexed: 11/16/2022]
Abstract
Purpose: Patient-derived xenografts ("xenopatients") of colorectal cancer metastases have been essential to identify genetic determinants of resistance to the anti-EGFR antibody cetuximab and to explore new therapeutic strategies. From xenopatients, a genetically annotated collection of stem-like cultures ("xenospheres") was generated and characterized for response to targeted therapies.Experimental Design: Xenospheres underwent exome-sequencing analysis, gene expression profile, and in vitro targeted treatments to assess genetic, biological, and pharmacologic correspondence with xenopatients, and to investigate nongenetic biomarkers of therapeutic resistance. The outcome of EGFR family inhibition was tested in an NRG1-expressing in vivo model.Results: Xenospheres faithfully retained the genetic make-up of their matched xenopatients over in vitro and in vivo passages. Frequent and rare genetic lesions triggering primary resistance to cetuximab through constitutive activation of the RAS signaling pathway were conserved, as well as the vulnerability to their respective targeted treatments. Xenospheres lacking such alterations (RASwt) were highly sensitive to cetuximab, but were protected by ligands activating the EGFR family, mostly NRG1. Upon reconstitution of NRG1 expression, xenospheres displayed increased tumorigenic potential in vivo and generated tumors completely resistant to cetuximab, and sensitive only to comprehensive EGFR family inhibition.Conclusions: Xenospheres are a reliable model to identify both genetic and nongenetic mechanisms of response and resistance to targeted therapies in colorectal cancer. In the absence of RAS pathway mutations, NRG1 and other EGFR ligands can play a major role in conferring primary cetuximab resistance, indicating that comprehensive inhibition of the EGFR family is required to achieve a significant therapeutic response. Clin Cancer Res; 24(4); 807-20. ©2017 AACRSee related commentary by Napolitano and Ciardiello, p. 727.
Collapse
Affiliation(s)
- Paolo Luraghi
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Torino, Italy
| | - Viola Bigatto
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Torino, Italy
| | - Elia Cipriano
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Torino, Italy.,Department of Oncology, University of Torino, Candiolo, Torino, Italy
| | - Gigliola Reato
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Torino, Italy.,Department of Oncology, University of Torino, Candiolo, Torino, Italy
| | - Francesca Orzan
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Torino, Italy
| | - Francesco Sassi
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Torino, Italy
| | | | - Claudio Isella
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Torino, Italy
| | - Sara E Bellomo
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Torino, Italy
| | - Enzo Medico
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Torino, Italy.,Department of Oncology, University of Torino, Candiolo, Torino, Italy
| | | | - Andrea Bertotti
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Torino, Italy.,Department of Oncology, University of Torino, Candiolo, Torino, Italy
| | - Livio Trusolino
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Torino, Italy.,Department of Oncology, University of Torino, Candiolo, Torino, Italy
| | - Carla Boccaccio
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Torino, Italy. .,Department of Oncology, University of Torino, Candiolo, Torino, Italy
| |
Collapse
|
484
|
Liu Z, Delavan B, Roberts R, Tong W. Lessons Learned from Two Decades of Anticancer Drugs. Trends Pharmacol Sci 2017; 38:852-872. [DOI: 10.1016/j.tips.2017.06.005] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Revised: 06/09/2017] [Accepted: 06/19/2017] [Indexed: 02/08/2023]
|
485
|
Senft D, Leiserson MDM, Ruppin E, Ronai ZA. Precision Oncology: The Road Ahead. Trends Mol Med 2017; 23:874-898. [PMID: 28887051 PMCID: PMC5718207 DOI: 10.1016/j.molmed.2017.08.003] [Citation(s) in RCA: 98] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Revised: 08/06/2017] [Accepted: 08/08/2017] [Indexed: 02/06/2023]
Abstract
Current efforts in precision oncology largely focus on the benefit of genomics-guided therapy. Yet, advances in sequencing techniques provide an unprecedented view of the complex genetic and nongenetic heterogeneity within individual tumors. Herein, we outline the benefits of integrating genomic and transcriptomic analyses for advanced precision oncology. We summarize relevant computational approaches to detect novel drivers and genetic vulnerabilities, suitable for therapeutic exploration. Clinically relevant platforms to functionally test predicted drugs/drug combinations for individual patients are reviewed. Finally, we highlight the technological advances in single cell analysis of tumor specimens. These may ultimately lead to the development of next-generation cancer drugs, capable of tackling the hurdles imposed by genetic and phenotypic heterogeneity on current anticancer therapies.
Collapse
Affiliation(s)
- Daniela Senft
- Tumor Initiation and Maintenance Program, NCI designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Mark D M Leiserson
- Microsoft Research New England, Cambridge, MA 02142, USA; Center for Bioinformatics and Computational Biology, University of Maryland, College Park, MD 20742, USA
| | - Eytan Ruppin
- School of Computer Sciences and Sackler School of Medicine, Tel Aviv University, Tel Aviv, 69978, Israel; Center for Bioinformatics and Computational Biology, University of Maryland, College Park, MD 20742, USA
| | - Ze'ev A Ronai
- Tumor Initiation and Maintenance Program, NCI designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA; Technion Integrated Cancer Center, Faculty of Medicine, Technion, Israel Institute of Technology, Haifa, 31096, Israel.
| |
Collapse
|
486
|
Moro M, Casanova M, Roz L. Patient-derived xenografts, a multi-faceted in vivo model enlightening research on rare liver cancer biology. Hepatobiliary Surg Nutr 2017; 6:344-346. [PMID: 29152485 DOI: 10.21037/hbsn.2017.06.03] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Affiliation(s)
- Massimo Moro
- Tumor Genomics Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Michela Casanova
- Pediatric Oncology Unit, Department of Medical Oncology and Hematology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Luca Roz
- Tumor Genomics Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| |
Collapse
|
487
|
Shin SH, Lim DY, Reddy K, Malakhova M, Liu F, Wang T, Song M, Chen H, Bae KB, Ryu J, Liu K, Lee MH, Bode AM, Dong Z. A Small Molecule Inhibitor of the β-Catenin-TCF4 Interaction Suppresses Colorectal Cancer Growth In Vitro and In Vivo. EBioMedicine 2017; 25:22-31. [PMID: 29033371 PMCID: PMC5704052 DOI: 10.1016/j.ebiom.2017.09.029] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 09/21/2017] [Accepted: 09/22/2017] [Indexed: 01/14/2023] Open
Abstract
Colorectal cancer is associated with aberrant activation of the Wnt pathway. β-Catenin plays essential roles in the Wnt pathway by interacting with T-cell factor 4 (TCF4) to transcribe oncogenes. We synthesized a small molecule, referred to as HI-B1, and evaluated signaling changes and biological consequences induced by the compound. HI-B1 inhibited β-catenin/TCF4 luciferase activity and preferentially caused apoptosis of cancer cells in which the survival is dependent on β-catenin. The formation of the β-catenin/TCF4 complex was disrupted by HI-B1 due to the direct interaction of HI-B1 with β-catenin. Colon cancer patient-derived xenograft (PDX) studies showed that a tumor with higher levels of β-catenin expression was more sensitive to HI-B1 treatment, compared to a tumor with lower expression levels of β-catenin. The different sensitivities of PDX tumors to HI-B1 were dependent on the β-catenin expression level and potentially could be further exploited for biomarker development and therapeutic applications against colon cancer. HI-B1 is a synthesized compound identified as a β-catenin inhibitor suppressing the β-catenin-TCF4 protein interaction. HI-B1 preferentially causes apoptosis in β-catenin-dependent cancer cells. A colon cancer PDX mouse model with a high level of β-catenin is sensitive to HI-B1.
β-catenin is an important protein that facilitates colon cancer. Shin et al. synthesized and identified HI-B1 as a direct β-catenin inhibitor. HI-B1 disrupted formation of the β-catenin-TCF4 protein complex. HI-B1 preferentially caused apoptosis of cancer cells in which the survival is dependent on β-catenin. In a comparison of two colon cancer PDX models with different β-catenin levels, they showed that β-catenin-high PDX is more sensitive to HI-B1 treatment than β-catenin-low PDX. HI-B1 could thus be further developed as a colon cancer drug, and β-catenin expression levels might be a predictive biomarker for colon cancer therapy using β-catenin inhibitors.
Collapse
Affiliation(s)
- Seung Ho Shin
- The Hormel Institute, University of Minnesota, Austin, MN 55912, USA; Program in Bioinformatics and Computational Biology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Do Young Lim
- The Hormel Institute, University of Minnesota, Austin, MN 55912, USA
| | - Kanamata Reddy
- The Hormel Institute, University of Minnesota, Austin, MN 55912, USA
| | | | - Fangfang Liu
- The China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, China; The Pathophysiology Department, The School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Ting Wang
- The China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, China
| | - Mengqiu Song
- The China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, China; The Pathophysiology Department, The School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Hanyong Chen
- The Hormel Institute, University of Minnesota, Austin, MN 55912, USA
| | - Ki Beom Bae
- The Hormel Institute, University of Minnesota, Austin, MN 55912, USA
| | - Joohyun Ryu
- The Hormel Institute, University of Minnesota, Austin, MN 55912, USA
| | - Kangdong Liu
- The China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, China; The Pathophysiology Department, The School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China; The Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou, China; The Affiliated Cancer Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Mee-Hyun Lee
- The China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, China
| | - Ann M Bode
- The Hormel Institute, University of Minnesota, Austin, MN 55912, USA; Program in Bioinformatics and Computational Biology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Zigang Dong
- The Hormel Institute, University of Minnesota, Austin, MN 55912, USA; Program in Bioinformatics and Computational Biology, University of Minnesota, Minneapolis, MN 55455, USA; The China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, China; The Pathophysiology Department, The School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China; The Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou, China; The Affiliated Cancer Hospital, Zhengzhou University, Zhengzhou, Henan, China.
| |
Collapse
|
488
|
Byrne AT, Alférez DG, Amant F, Annibali D, Arribas J, Biankin AV, Bruna A, Budinská E, Caldas C, Chang DK, Clarke RB, Clevers H, Coukos G, Dangles-Marie V, Eckhardt SG, Gonzalez-Suarez E, Hermans E, Hidalgo M, Jarzabek MA, de Jong S, Jonkers J, Kemper K, Lanfrancone L, Mælandsmo GM, Marangoni E, Marine JC, Medico E, Norum JH, Palmer HG, Peeper DS, Pelicci PG, Piris-Gimenez A, Roman-Roman S, Rueda OM, Seoane J, Serra V, Soucek L, Vanhecke D, Villanueva A, Vinolo E, Bertotti A, Trusolino L. Interrogating open issues in cancer medicine with patient-derived xenografts. Nat Rev Cancer 2017; 17:632. [PMID: 28912576 DOI: 10.1038/nrc.2017.85] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
This corrects the article DOI: 10.1038/nrc.2016.140.
Collapse
|
489
|
Addressing the challenges of applying precision oncology. NPJ Precis Oncol 2017; 1:28. [PMID: 29872710 PMCID: PMC5871855 DOI: 10.1038/s41698-017-0032-z] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Revised: 07/25/2017] [Accepted: 07/26/2017] [Indexed: 02/07/2023] Open
Abstract
Precision oncology is described as the matching of the most accurate and effective treatments with the individual cancer patient. Identification of important gene mutations, such as BRCA1/2 that drive carcinogenesis, helped pave the way for precision diagnosis in cancer. Oncoproteins and their signaling pathways have been extensively studied, leading to the development of target-based precision therapies against several types of cancers. Although many challenges exist that could hinder the success of precision oncology, cutting-edge tools for precision diagnosis and precision therapy will assist in overcoming many of these difficulties. Based on the continued rapid progression of genomic analysis, drug development, and clinical trial design, precision oncology will ultimately become the standard of care in cancer therapeutics.
Collapse
|
490
|
Perales-Patón J, Piñeiro-Yañez E, Tejero H, López-Casas PP, Hidalgo M, Gómez-López G, Al-Shahrour F. Pancreas Cancer Precision Treatment Using Avatar Mice from a Bioinformatics Perspective. Public Health Genomics 2017; 20:81-91. [PMID: 28858862 DOI: 10.1159/000479812] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Accepted: 07/27/2017] [Indexed: 01/03/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a leading cause of cancer-related death among solid malignancies. Unfortunately, PDAC lethality has not substantially decreased over the past 20 years. This aggressiveness is related to the genomic complexity and heterogeneity of PDAC, but also to the absence of an effective screening for the detection of early-stage tumors and a lack of efficient therapeutic options. Therefore, there is an urgent need to improve the arsenal of anti-PDAC drugs for an effective treatment of these patients. Patient-derived xenograft (PDX) mouse models represent a promising strategy to personalize PDAC treatment, offering a bench testing of candidate treatments and helping to select empirical treatments in PDAC patients with no therapeutic targets. Moreover, bioinformatics-based approaches have the potential to offer systematic insights into PDAC etiology predicting putatively actionable tumor-specific genomic alterations, identifying novel biomarkers and generating disease-associated gene expression signatures. This review focuses on recent efforts to individualize PDAC treatments using PDX models. Additionally, we discuss the current understanding of the PDAC genomic landscape and the putative druggable targets derived from mutational studies. PDAC molecular subclassifications and gene expression profiling studies are reviewed as well. Finally, latest bioinformatics methodologies based on somatic variant detection and prioritization, in silico drug response prediction, and drug repositioning to improve the treatment of advanced PDAC tumors are also covered.
Collapse
Affiliation(s)
- Javier Perales-Patón
- Bioinformatics Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | | | | | | | | | | | | |
Collapse
|
491
|
Establishment and characterization of uterine sarcoma and carcinosarcoma patient-derived xenograft models. Gynecol Oncol 2017. [DOI: 10.1016/j.ygyno.2017.06.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
492
|
Gómez-Cuadrado L, Tracey N, Ma R, Qian B, Brunton VG. Mouse models of metastasis: progress and prospects. Dis Model Mech 2017; 10:1061-1074. [PMID: 28883015 PMCID: PMC5611969 DOI: 10.1242/dmm.030403] [Citation(s) in RCA: 211] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Metastasis is the spread of cancer cells from a primary tumor to distant sites within the body to establish secondary tumors. Although this is an inefficient process, the consequences are devastating as metastatic disease accounts for >90% of cancer-related deaths. The formation of metastases is the result of a series of events that allow cancer cells to escape from the primary site, survive in the lymphatic system or blood vessels, extravasate and grow at distant sites. The metastatic capacity of a tumor is determined by genetic and epigenetic changes within the cancer cells as well as contributions from cells in the tumor microenvironment. Mouse models have proven to be an important tool for unraveling the complex interactions involved in the metastatic cascade and delineating its many stages. Here, we critically appraise the strengths and weaknesses of the current mouse models and highlight the recent advances that have been made using these models in our understanding of metastasis. We also discuss the use of these models for testing potential therapies and the challenges associated with the translation of these findings into the provision of new and effective treatments for cancer patients.
Collapse
Affiliation(s)
- Laura Gómez-Cuadrado
- Edinburgh Cancer Research Centre, Institute for Genetics and Molecular Medicine, Edinburgh, EH4 2XR, UK
| | - Natasha Tracey
- Edinburgh Cancer Research Centre, Institute for Genetics and Molecular Medicine, Edinburgh, EH4 2XR, UK
| | - Ruoyu Ma
- MRC Centre for Reproductive Health, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Binzhi Qian
- MRC Centre for Reproductive Health, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
- Edinburgh Cancer Research UK Centre, Queen's Medical Research Institute, Edinburgh, EH16 4TJ, UK
| | - Valerie G Brunton
- Edinburgh Cancer Research Centre, Institute for Genetics and Molecular Medicine, Edinburgh, EH4 2XR, UK
| |
Collapse
|
493
|
Vanpouille-Box C, Lhuillier C, Bezu L, Aranda F, Yamazaki T, Kepp O, Fucikova J, Spisek R, Demaria S, Formenti SC, Zitvogel L, Kroemer G, Galluzzi L. Trial watch: Immune checkpoint blockers for cancer therapy. Oncoimmunology 2017; 6:e1373237. [PMID: 29147629 DOI: 10.1080/2162402x.2017.1373237] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Accepted: 08/25/2017] [Indexed: 02/08/2023] Open
Abstract
Immune checkpoint blockers (ICBs) are literally revolutionizing the clinical management of an ever more diversified panel of oncological indications. Although considerable attention persists around the inhibition of cytotoxic T lymphocyte-associated protein 4 (CTLA4) and programmed cell death 1 (PDCD1, best known as PD-1) signaling, several other co-inhibitory T-cell receptors are being evaluated as potential targets for the development of novel ICBs. Moreover, substantial efforts are being devoted to the identification of biomarkers that reliably predict the likelihood of each patient to obtain clinical benefits from ICBs in the absence of severe toxicity. Tailoring the delivery of specific ICBs or combinations thereof to selected patient populations in the context of precision medicine programs constitutes indeed a major objective of the future of ICB-based immunotherapy. Here, we discuss recent preclinical and clinical advances on the development of ICBs for oncological indications.
Collapse
Affiliation(s)
| | - Claire Lhuillier
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
| | - Lucillia Bezu
- Université Paris Descartes/Paris V, Paris, France.,Université Pierre et Marie Curie/Paris VI, Paris, France.,Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, Paris, France.,INSERM, U1138, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, Villejuif, France
| | - Fernando Aranda
- Immunoreceptors of the Innate and Adaptive System Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Takahiro Yamazaki
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
| | - Oliver Kepp
- Université Paris Descartes/Paris V, Paris, France.,Université Pierre et Marie Curie/Paris VI, Paris, France.,Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, Paris, France.,INSERM, U1138, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, Villejuif, France
| | - Jitka Fucikova
- Sotio a.c., Prague, Czech Republic.,Dept. of Immunology, 2nd Faculty of Medicine and University Hospital Motol, Charles University, Prague, Czech Republic
| | - Radek Spisek
- Sotio a.c., Prague, Czech Republic.,Dept. of Immunology, 2nd Faculty of Medicine and University Hospital Motol, Charles University, Prague, Czech Republic
| | - Sandra Demaria
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA.,Sandra and Edward Meyer Cancer Center, New York, NY, USA
| | - Silvia C Formenti
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA.,Sandra and Edward Meyer Cancer Center, New York, NY, USA
| | - Laurence Zitvogel
- Gustave Roussy Comprehensive Cancer Institute, Villejuif, France.,INSERM, U1015, Villejuif, France.,Center of Clinical Investigations in Biotherapies of Cancer (CICBT) 1428, Villejuif, France.,Université Paris Sud/Paris XI, Le Kremlin-Bicêtre, France
| | - Guido Kroemer
- Université Paris Descartes/Paris V, Paris, France.,Université Pierre et Marie Curie/Paris VI, Paris, France.,Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, Paris, France.,INSERM, U1138, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, Villejuif, France.,Department of Women's and Children's Health, Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden.,Pôle de Biologie, Hopitâl Européen George Pompidou, AP-HP, Paris, France
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA.,Université Paris Descartes/Paris V, Paris, France.,Sandra and Edward Meyer Cancer Center, New York, NY, USA
| |
Collapse
|
494
|
|
495
|
Farooqi AA, Naqvi SKUH, Perk AA, Yanar O, Tabassum S, Ahmad MS, Mansoor Q, Ashry MS, Ismail M, Naoum GE, Arafat WO. Natural Agents-Mediated Targeting of Histone Deacetylases. Arch Immunol Ther Exp (Warsz) 2017; 66:31-44. [PMID: 28852775 DOI: 10.1007/s00005-017-0488-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2017] [Accepted: 03/03/2017] [Indexed: 02/07/2023]
Abstract
In the past few years, basic and clinical scientists have witnessed landmark achievements in many research projects, such as those conducted by the US National Institutes of Health Roadmap Epigenomics Mapping Consortium, the International Human Epigenome Consortium, The Cancer Genome Atlas Network and the International Cancer Genome Consortium, which have provided near-complete resolution of epigenetic landscape in different diseases. Furthermore, genome sequencing of tumors has provided compelling evidence related to frequent existence of mutations in readers, erasers and writers of epigenome in different cancers. Histone acetylation is an intricate mechanism modulated by two opposing sets of enzymes and deeply studied as a key biological phenomenon in 1964 by Vincent Allfrey and colleagues. The research group suggested that this protein modification contributed substantially in transcriptional regulation. Subsequently, histone deacetylases (HDACs), histone acetyltransferases and acetyl-Lys-binding proteins were identified as transcriptional mediators, which further deepened our comprehension regarding biochemical modifications. Overwhelmingly increasing high-impact research is improving our understanding of this molecularly controlled mechanism; moreover, quantification and identification of lysine acetylation by mass spectrometry has added new layers of information. We partition this multi-component review into how both activity and expression of HDAC are targeted using natural agents. We also set spotlight on how oncogenic fusion proteins tactfully utilize HDAC-associated nano-machinery to modulate expression of different genes and how HDAC inhibitors regulate TRAIL-induced apoptosis in cancer cells. HDAC inhibitors have been reported to upregulate expression of TRAIL receptors and protect TRAIL from proteasomal degradation. Deeper understanding of HDAC biology will be useful for stratification and selection of patients who are responders, non-responders and poor-responders for HDACi therapy, and for the rational design of combination studies using HDACi.
Collapse
Affiliation(s)
| | | | - Aliye Aras Perk
- Division of Botany, Department of Biology, Faculty of Science, Istanbul University, Istanbul, Turkey
| | - Onur Yanar
- Division of Botany, Department of Biology, Faculty of Science, Istanbul University, Istanbul, Turkey
| | - Sobia Tabassum
- Department of Bioinformatics and Biotechnology, International Islamic University, Islamabad, Pakistan
| | - Muhammad Sheeraz Ahmad
- Institute of Biochemistry and Biotechnology, PMAS Arid Agriculture University, Rawalpindi, Pakistan
| | - Qaisar Mansoor
- Institute of Biomedical and Genetic Engineering, Islamabad, Pakistan
| | - Mohamed S Ashry
- Clinical Oncology Department, Mansoura University, Mansoura, Egypt
| | - Muhammad Ismail
- Institute of Biomedical and Genetic Engineering, Islamabad, Pakistan
| | - George E Naoum
- Alexandria Comprehensive Cancer Center, Alexandria, Egypt.,Department of radiation Oncology, Harvard Medical School, Massachusetts General Hospital, Boston, USA
| | - Waleed O Arafat
- Clinical Oncology Department, Alexandria University, Alexandria, Egypt.
| |
Collapse
|
496
|
Russell TA, Eckardt MA, Murakami T, Elliott IA, Kawaguchi K, Kiyuna T, Igarashi K, Li Y, Crompton JG, Graham DS, Dry SM, Bernthal N, Yanagawa J, Kalbasi A, Federman N, Chmielowski B, Singh AS, Hoffman RM, Eilber FC. Clinical Factors That Affect the Establishment of Soft Tissue Sarcoma Patient-Derived Orthotopic Xenografts: A University of California, Los Angeles, Sarcoma Program Prospective Clinical Trial. JCO Precis Oncol 2017; 2017. [PMID: 30613825 DOI: 10.1200/po.17.00071] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Purpose Given the diverse and aggressive nature of soft tissue sarcomas (STSs), a need exists for more-precise therapy. Patient-derived orthotopic xenografts (PDOXs) provide a unique platform for personalized treatment. Thus, identification of patient and treatment factors that predict PDOX establishment is important. This study assessed the feasibility of incorporating PDOXs into the clinical setting and identifying factors associated with PDOX establishment. Patients and Methods From May 2015 to May 2016, 107 patients with biopsy-proven or potential STS were enrolled. Tumor samples were obtained intraoperatively and orthotopically implanted into nude mice in the corresponding anatomic location. PDOXs were considered established after engraftment and serial passage. Factors associated with establishment were analyzed by logistic regression and time to establishment by time-to-event analysis. Results Only high-grade tumors established (32 of 72 [44.4%]). The establishment rate (ER) varied by neoadjuvant therapy and treatment response, with the highest ER among untreated high-grade tumors (26 of 42 [61.9%]). Tumors exposed to radiation preoperatively did not establish (zero of 11 [0%]), and tumors exposed to neoadjuvant chemotherapy had a lower ER(31.9%) than untreated tumors. Only STSs with minimal pathologic response to neoadjuvant treatment (≤ 30%) established a PDOX (six of 18 [33.3%]). Median establishment time was 54 days, which varied by neoadjuvant therapy but was not statistically significant (P = .180). Conclusion To our knowledge, in the largest STS PDOX study to date, we demonstrate a 62% ER among untreated high-grade tumors with a median establishment time of 54 days. Neoadjuvant therapy, particularly radiation, and pathologic response to treatment were associated with a reduced rate of PDOX establishment.
Collapse
Affiliation(s)
- Tara A Russell
- Tara A. Russell, Irmina A. Elliott, Yungfeng Li, Joseph G. Crompton, Danielle S. Graham, Sarah M. Dry, Nicholas Bernthal, Jane Yanagawa, Anusha Kalbasi, Noah Federman, Bartosz Chmielowski, Arun S. Singh, and Fritz C. Eilber, University of California, Los Angeles; Tara A. Russell, Veterans Affairs Los Angeles Health Services Research & Development Center of Innovation, Los Angeles, CA; Mark A. Eckardt, Yale School of Medicine, New Haven, CT; Takashi Murakami, Kei Kawaguchi, Tasuku Kiyuna, Kentaro Igarashi, and Robert M. Hoffman, AntiCancer; and Takashi Murakami, Kei Kawaguchi, Tasuku Kiyuna, Kentaro Igarashi, and Robert M. Hoffman, University of California, San Diego, San Diego, CA
| | - Mark A Eckardt
- Tara A. Russell, Irmina A. Elliott, Yungfeng Li, Joseph G. Crompton, Danielle S. Graham, Sarah M. Dry, Nicholas Bernthal, Jane Yanagawa, Anusha Kalbasi, Noah Federman, Bartosz Chmielowski, Arun S. Singh, and Fritz C. Eilber, University of California, Los Angeles; Tara A. Russell, Veterans Affairs Los Angeles Health Services Research & Development Center of Innovation, Los Angeles, CA; Mark A. Eckardt, Yale School of Medicine, New Haven, CT; Takashi Murakami, Kei Kawaguchi, Tasuku Kiyuna, Kentaro Igarashi, and Robert M. Hoffman, AntiCancer; and Takashi Murakami, Kei Kawaguchi, Tasuku Kiyuna, Kentaro Igarashi, and Robert M. Hoffman, University of California, San Diego, San Diego, CA
| | - Takashi Murakami
- Tara A. Russell, Irmina A. Elliott, Yungfeng Li, Joseph G. Crompton, Danielle S. Graham, Sarah M. Dry, Nicholas Bernthal, Jane Yanagawa, Anusha Kalbasi, Noah Federman, Bartosz Chmielowski, Arun S. Singh, and Fritz C. Eilber, University of California, Los Angeles; Tara A. Russell, Veterans Affairs Los Angeles Health Services Research & Development Center of Innovation, Los Angeles, CA; Mark A. Eckardt, Yale School of Medicine, New Haven, CT; Takashi Murakami, Kei Kawaguchi, Tasuku Kiyuna, Kentaro Igarashi, and Robert M. Hoffman, AntiCancer; and Takashi Murakami, Kei Kawaguchi, Tasuku Kiyuna, Kentaro Igarashi, and Robert M. Hoffman, University of California, San Diego, San Diego, CA
| | - Irmina A Elliott
- Tara A. Russell, Irmina A. Elliott, Yungfeng Li, Joseph G. Crompton, Danielle S. Graham, Sarah M. Dry, Nicholas Bernthal, Jane Yanagawa, Anusha Kalbasi, Noah Federman, Bartosz Chmielowski, Arun S. Singh, and Fritz C. Eilber, University of California, Los Angeles; Tara A. Russell, Veterans Affairs Los Angeles Health Services Research & Development Center of Innovation, Los Angeles, CA; Mark A. Eckardt, Yale School of Medicine, New Haven, CT; Takashi Murakami, Kei Kawaguchi, Tasuku Kiyuna, Kentaro Igarashi, and Robert M. Hoffman, AntiCancer; and Takashi Murakami, Kei Kawaguchi, Tasuku Kiyuna, Kentaro Igarashi, and Robert M. Hoffman, University of California, San Diego, San Diego, CA
| | - Kei Kawaguchi
- Tara A. Russell, Irmina A. Elliott, Yungfeng Li, Joseph G. Crompton, Danielle S. Graham, Sarah M. Dry, Nicholas Bernthal, Jane Yanagawa, Anusha Kalbasi, Noah Federman, Bartosz Chmielowski, Arun S. Singh, and Fritz C. Eilber, University of California, Los Angeles; Tara A. Russell, Veterans Affairs Los Angeles Health Services Research & Development Center of Innovation, Los Angeles, CA; Mark A. Eckardt, Yale School of Medicine, New Haven, CT; Takashi Murakami, Kei Kawaguchi, Tasuku Kiyuna, Kentaro Igarashi, and Robert M. Hoffman, AntiCancer; and Takashi Murakami, Kei Kawaguchi, Tasuku Kiyuna, Kentaro Igarashi, and Robert M. Hoffman, University of California, San Diego, San Diego, CA
| | - Tasuku Kiyuna
- Tara A. Russell, Irmina A. Elliott, Yungfeng Li, Joseph G. Crompton, Danielle S. Graham, Sarah M. Dry, Nicholas Bernthal, Jane Yanagawa, Anusha Kalbasi, Noah Federman, Bartosz Chmielowski, Arun S. Singh, and Fritz C. Eilber, University of California, Los Angeles; Tara A. Russell, Veterans Affairs Los Angeles Health Services Research & Development Center of Innovation, Los Angeles, CA; Mark A. Eckardt, Yale School of Medicine, New Haven, CT; Takashi Murakami, Kei Kawaguchi, Tasuku Kiyuna, Kentaro Igarashi, and Robert M. Hoffman, AntiCancer; and Takashi Murakami, Kei Kawaguchi, Tasuku Kiyuna, Kentaro Igarashi, and Robert M. Hoffman, University of California, San Diego, San Diego, CA
| | - Kentaro Igarashi
- Tara A. Russell, Irmina A. Elliott, Yungfeng Li, Joseph G. Crompton, Danielle S. Graham, Sarah M. Dry, Nicholas Bernthal, Jane Yanagawa, Anusha Kalbasi, Noah Federman, Bartosz Chmielowski, Arun S. Singh, and Fritz C. Eilber, University of California, Los Angeles; Tara A. Russell, Veterans Affairs Los Angeles Health Services Research & Development Center of Innovation, Los Angeles, CA; Mark A. Eckardt, Yale School of Medicine, New Haven, CT; Takashi Murakami, Kei Kawaguchi, Tasuku Kiyuna, Kentaro Igarashi, and Robert M. Hoffman, AntiCancer; and Takashi Murakami, Kei Kawaguchi, Tasuku Kiyuna, Kentaro Igarashi, and Robert M. Hoffman, University of California, San Diego, San Diego, CA
| | - Yungfeng Li
- Tara A. Russell, Irmina A. Elliott, Yungfeng Li, Joseph G. Crompton, Danielle S. Graham, Sarah M. Dry, Nicholas Bernthal, Jane Yanagawa, Anusha Kalbasi, Noah Federman, Bartosz Chmielowski, Arun S. Singh, and Fritz C. Eilber, University of California, Los Angeles; Tara A. Russell, Veterans Affairs Los Angeles Health Services Research & Development Center of Innovation, Los Angeles, CA; Mark A. Eckardt, Yale School of Medicine, New Haven, CT; Takashi Murakami, Kei Kawaguchi, Tasuku Kiyuna, Kentaro Igarashi, and Robert M. Hoffman, AntiCancer; and Takashi Murakami, Kei Kawaguchi, Tasuku Kiyuna, Kentaro Igarashi, and Robert M. Hoffman, University of California, San Diego, San Diego, CA
| | - Joseph G Crompton
- Tara A. Russell, Irmina A. Elliott, Yungfeng Li, Joseph G. Crompton, Danielle S. Graham, Sarah M. Dry, Nicholas Bernthal, Jane Yanagawa, Anusha Kalbasi, Noah Federman, Bartosz Chmielowski, Arun S. Singh, and Fritz C. Eilber, University of California, Los Angeles; Tara A. Russell, Veterans Affairs Los Angeles Health Services Research & Development Center of Innovation, Los Angeles, CA; Mark A. Eckardt, Yale School of Medicine, New Haven, CT; Takashi Murakami, Kei Kawaguchi, Tasuku Kiyuna, Kentaro Igarashi, and Robert M. Hoffman, AntiCancer; and Takashi Murakami, Kei Kawaguchi, Tasuku Kiyuna, Kentaro Igarashi, and Robert M. Hoffman, University of California, San Diego, San Diego, CA
| | - Danielle S Graham
- Tara A. Russell, Irmina A. Elliott, Yungfeng Li, Joseph G. Crompton, Danielle S. Graham, Sarah M. Dry, Nicholas Bernthal, Jane Yanagawa, Anusha Kalbasi, Noah Federman, Bartosz Chmielowski, Arun S. Singh, and Fritz C. Eilber, University of California, Los Angeles; Tara A. Russell, Veterans Affairs Los Angeles Health Services Research & Development Center of Innovation, Los Angeles, CA; Mark A. Eckardt, Yale School of Medicine, New Haven, CT; Takashi Murakami, Kei Kawaguchi, Tasuku Kiyuna, Kentaro Igarashi, and Robert M. Hoffman, AntiCancer; and Takashi Murakami, Kei Kawaguchi, Tasuku Kiyuna, Kentaro Igarashi, and Robert M. Hoffman, University of California, San Diego, San Diego, CA
| | - Sarah M Dry
- Tara A. Russell, Irmina A. Elliott, Yungfeng Li, Joseph G. Crompton, Danielle S. Graham, Sarah M. Dry, Nicholas Bernthal, Jane Yanagawa, Anusha Kalbasi, Noah Federman, Bartosz Chmielowski, Arun S. Singh, and Fritz C. Eilber, University of California, Los Angeles; Tara A. Russell, Veterans Affairs Los Angeles Health Services Research & Development Center of Innovation, Los Angeles, CA; Mark A. Eckardt, Yale School of Medicine, New Haven, CT; Takashi Murakami, Kei Kawaguchi, Tasuku Kiyuna, Kentaro Igarashi, and Robert M. Hoffman, AntiCancer; and Takashi Murakami, Kei Kawaguchi, Tasuku Kiyuna, Kentaro Igarashi, and Robert M. Hoffman, University of California, San Diego, San Diego, CA
| | - Nicholas Bernthal
- Tara A. Russell, Irmina A. Elliott, Yungfeng Li, Joseph G. Crompton, Danielle S. Graham, Sarah M. Dry, Nicholas Bernthal, Jane Yanagawa, Anusha Kalbasi, Noah Federman, Bartosz Chmielowski, Arun S. Singh, and Fritz C. Eilber, University of California, Los Angeles; Tara A. Russell, Veterans Affairs Los Angeles Health Services Research & Development Center of Innovation, Los Angeles, CA; Mark A. Eckardt, Yale School of Medicine, New Haven, CT; Takashi Murakami, Kei Kawaguchi, Tasuku Kiyuna, Kentaro Igarashi, and Robert M. Hoffman, AntiCancer; and Takashi Murakami, Kei Kawaguchi, Tasuku Kiyuna, Kentaro Igarashi, and Robert M. Hoffman, University of California, San Diego, San Diego, CA
| | - Jane Yanagawa
- Tara A. Russell, Irmina A. Elliott, Yungfeng Li, Joseph G. Crompton, Danielle S. Graham, Sarah M. Dry, Nicholas Bernthal, Jane Yanagawa, Anusha Kalbasi, Noah Federman, Bartosz Chmielowski, Arun S. Singh, and Fritz C. Eilber, University of California, Los Angeles; Tara A. Russell, Veterans Affairs Los Angeles Health Services Research & Development Center of Innovation, Los Angeles, CA; Mark A. Eckardt, Yale School of Medicine, New Haven, CT; Takashi Murakami, Kei Kawaguchi, Tasuku Kiyuna, Kentaro Igarashi, and Robert M. Hoffman, AntiCancer; and Takashi Murakami, Kei Kawaguchi, Tasuku Kiyuna, Kentaro Igarashi, and Robert M. Hoffman, University of California, San Diego, San Diego, CA
| | - Anusha Kalbasi
- Tara A. Russell, Irmina A. Elliott, Yungfeng Li, Joseph G. Crompton, Danielle S. Graham, Sarah M. Dry, Nicholas Bernthal, Jane Yanagawa, Anusha Kalbasi, Noah Federman, Bartosz Chmielowski, Arun S. Singh, and Fritz C. Eilber, University of California, Los Angeles; Tara A. Russell, Veterans Affairs Los Angeles Health Services Research & Development Center of Innovation, Los Angeles, CA; Mark A. Eckardt, Yale School of Medicine, New Haven, CT; Takashi Murakami, Kei Kawaguchi, Tasuku Kiyuna, Kentaro Igarashi, and Robert M. Hoffman, AntiCancer; and Takashi Murakami, Kei Kawaguchi, Tasuku Kiyuna, Kentaro Igarashi, and Robert M. Hoffman, University of California, San Diego, San Diego, CA
| | - Noah Federman
- Tara A. Russell, Irmina A. Elliott, Yungfeng Li, Joseph G. Crompton, Danielle S. Graham, Sarah M. Dry, Nicholas Bernthal, Jane Yanagawa, Anusha Kalbasi, Noah Federman, Bartosz Chmielowski, Arun S. Singh, and Fritz C. Eilber, University of California, Los Angeles; Tara A. Russell, Veterans Affairs Los Angeles Health Services Research & Development Center of Innovation, Los Angeles, CA; Mark A. Eckardt, Yale School of Medicine, New Haven, CT; Takashi Murakami, Kei Kawaguchi, Tasuku Kiyuna, Kentaro Igarashi, and Robert M. Hoffman, AntiCancer; and Takashi Murakami, Kei Kawaguchi, Tasuku Kiyuna, Kentaro Igarashi, and Robert M. Hoffman, University of California, San Diego, San Diego, CA
| | - Bartosz Chmielowski
- Tara A. Russell, Irmina A. Elliott, Yungfeng Li, Joseph G. Crompton, Danielle S. Graham, Sarah M. Dry, Nicholas Bernthal, Jane Yanagawa, Anusha Kalbasi, Noah Federman, Bartosz Chmielowski, Arun S. Singh, and Fritz C. Eilber, University of California, Los Angeles; Tara A. Russell, Veterans Affairs Los Angeles Health Services Research & Development Center of Innovation, Los Angeles, CA; Mark A. Eckardt, Yale School of Medicine, New Haven, CT; Takashi Murakami, Kei Kawaguchi, Tasuku Kiyuna, Kentaro Igarashi, and Robert M. Hoffman, AntiCancer; and Takashi Murakami, Kei Kawaguchi, Tasuku Kiyuna, Kentaro Igarashi, and Robert M. Hoffman, University of California, San Diego, San Diego, CA
| | - Arun S Singh
- Tara A. Russell, Irmina A. Elliott, Yungfeng Li, Joseph G. Crompton, Danielle S. Graham, Sarah M. Dry, Nicholas Bernthal, Jane Yanagawa, Anusha Kalbasi, Noah Federman, Bartosz Chmielowski, Arun S. Singh, and Fritz C. Eilber, University of California, Los Angeles; Tara A. Russell, Veterans Affairs Los Angeles Health Services Research & Development Center of Innovation, Los Angeles, CA; Mark A. Eckardt, Yale School of Medicine, New Haven, CT; Takashi Murakami, Kei Kawaguchi, Tasuku Kiyuna, Kentaro Igarashi, and Robert M. Hoffman, AntiCancer; and Takashi Murakami, Kei Kawaguchi, Tasuku Kiyuna, Kentaro Igarashi, and Robert M. Hoffman, University of California, San Diego, San Diego, CA
| | - Robert M Hoffman
- Tara A. Russell, Irmina A. Elliott, Yungfeng Li, Joseph G. Crompton, Danielle S. Graham, Sarah M. Dry, Nicholas Bernthal, Jane Yanagawa, Anusha Kalbasi, Noah Federman, Bartosz Chmielowski, Arun S. Singh, and Fritz C. Eilber, University of California, Los Angeles; Tara A. Russell, Veterans Affairs Los Angeles Health Services Research & Development Center of Innovation, Los Angeles, CA; Mark A. Eckardt, Yale School of Medicine, New Haven, CT; Takashi Murakami, Kei Kawaguchi, Tasuku Kiyuna, Kentaro Igarashi, and Robert M. Hoffman, AntiCancer; and Takashi Murakami, Kei Kawaguchi, Tasuku Kiyuna, Kentaro Igarashi, and Robert M. Hoffman, University of California, San Diego, San Diego, CA
| | - Fritz C Eilber
- Tara A. Russell, Irmina A. Elliott, Yungfeng Li, Joseph G. Crompton, Danielle S. Graham, Sarah M. Dry, Nicholas Bernthal, Jane Yanagawa, Anusha Kalbasi, Noah Federman, Bartosz Chmielowski, Arun S. Singh, and Fritz C. Eilber, University of California, Los Angeles; Tara A. Russell, Veterans Affairs Los Angeles Health Services Research & Development Center of Innovation, Los Angeles, CA; Mark A. Eckardt, Yale School of Medicine, New Haven, CT; Takashi Murakami, Kei Kawaguchi, Tasuku Kiyuna, Kentaro Igarashi, and Robert M. Hoffman, AntiCancer; and Takashi Murakami, Kei Kawaguchi, Tasuku Kiyuna, Kentaro Igarashi, and Robert M. Hoffman, University of California, San Diego, San Diego, CA
| |
Collapse
|
497
|
Fiori ME, Villanova L, De Maria R. Cancer stem cells: at the forefront of personalized medicine and immunotherapy. Curr Opin Pharmacol 2017; 35:1-11. [DOI: 10.1016/j.coph.2017.04.006] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Revised: 04/05/2017] [Accepted: 04/24/2017] [Indexed: 01/21/2023]
|
498
|
Weeber F, Ooft SN, Dijkstra KK, Voest EE. Tumor Organoids as a Pre-clinical Cancer Model for Drug Discovery. Cell Chem Biol 2017; 24:1092-1100. [PMID: 28757181 DOI: 10.1016/j.chembiol.2017.06.012] [Citation(s) in RCA: 349] [Impact Index Per Article: 43.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Revised: 05/24/2017] [Accepted: 06/20/2017] [Indexed: 01/21/2023]
Abstract
Tumor organoids are 3D cultures of cancer cells that can be derived on an individual patient basis with a high success rate. This creates opportunities to build large biobanks with relevant patient material that can be used to perform drug screens and facilitate drug development. The high take rate will also allow side-by-side comparison to evaluate the translational potential of this model system to the patient. These tumors-in-a-dish can be established for a variety of tumor types including colorectal, pancreas, stomach, prostate, and breast cancers. In this review, we highlight what is currently known about tumor organoid culture, the advantages and challenges of the model system, compare it with other pre-clinical cancer models, and evaluate its value for drug development.
Collapse
Affiliation(s)
- Fleur Weeber
- Department of Molecular Oncology and Immunology, The Netherlands Cancer Institute, Amsterdam, Noord-Holland 1066CX, the Netherlands
| | - Salo N Ooft
- Department of Molecular Oncology and Immunology, The Netherlands Cancer Institute, Amsterdam, Noord-Holland 1066CX, the Netherlands
| | - Krijn K Dijkstra
- Department of Molecular Oncology and Immunology, The Netherlands Cancer Institute, Amsterdam, Noord-Holland 1066CX, the Netherlands
| | - Emile E Voest
- Department of Molecular Oncology and Immunology, The Netherlands Cancer Institute, Amsterdam, Noord-Holland 1066CX, the Netherlands; Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, Noord-Holland 1066CX, the Netherlands; Foundation Hubrecht Organoid Technology (HUB), Utrecht, Utrecht 3584CM, the Netherlands; Cancer Genomics.nl, Utrecht, Utrecht 3584 CG, the Netherlands.
| |
Collapse
|
499
|
Gomez-Miragaya J, González-Suárez E. Tumor-initiating CD49f cells are a hallmark of chemoresistant triple negative breast cancer. Mol Cell Oncol 2017; 4:e1338208. [PMID: 28868349 DOI: 10.1080/23723556.2017.1338208] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Revised: 05/30/2017] [Accepted: 05/30/2017] [Indexed: 12/24/2022]
Abstract
Taxanes are mainstay treatment of triple negative breast cancer (TNBC) patients but resistance often develops. Using TNBC patient-derived orthoxenografts (PDX) we have recently discovered that a CD49f+ chemoresistant population with tumor-initiating ability is present in sensitive tumors and expands in tumors that have acquired resistance. Importantly, sensitivity to taxanes is recovered after long-term drug interruption. The characterization of this chemoresistant CD49f+ cells provides a unique opportunity to identify novel targets for the treatment of chemoresistant TNBC.
Collapse
Affiliation(s)
- Jorge Gomez-Miragaya
- Transformation and Metastasis laboratory, Cancer Epigenetics and Biology Program (PEBC), Biomedical Research Institute of Bellvitge (IDIBELL), Barcelona, Spain, Av.Gran Via de L'Hospitalet, 199 - 203. L'Hospitalet de Llobregat, Barcelona, Spain
| | - Eva González-Suárez
- Transformation and Metastasis laboratory, Cancer Epigenetics and Biology Program (PEBC), Biomedical Research Institute of Bellvitge (IDIBELL), Barcelona, Spain, Av.Gran Via de L'Hospitalet, 199 - 203. L'Hospitalet de Llobregat, Barcelona, Spain
| |
Collapse
|
500
|
Pancreatic cancer heterogeneity and response to Mek inhibition. Oncogene 2017; 36:5639-5647. [DOI: 10.1038/onc.2017.174] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2016] [Revised: 03/29/2017] [Accepted: 04/19/2017] [Indexed: 12/22/2022]
|