51
|
Bartucci M, Ferrari AC, Kim IY, Ploss A, Yarmush M, Sabaawy HE. Corrigendum: Personalized Medicine Approaches in Prostate Cancer Employing Patient Derived 3D Organoids and Humanized Mice. Front Cell Dev Biol 2016; 4:74. [PMID: 27583246 PMCID: PMC4999522 DOI: 10.3389/fcell.2016.00074] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Accepted: 06/28/2016] [Indexed: 11/13/2022] Open
Affiliation(s)
- Monica Bartucci
- Rutgers Cancer Institute of New Jersey, Rutgers University New Brunswick, NJ, USA
| | - Anna C Ferrari
- Rutgers Cancer Institute of New Jersey, Rutgers University New Brunswick, NJ, USA
| | - Isaac Yi Kim
- Rutgers Cancer Institute of New Jersey, Rutgers University New Brunswick, NJ, USA
| | - Alexander Ploss
- Rutgers Cancer Institute of New Jersey, Rutgers UniversityNew Brunswick, NJ, USA; Department of Molecular Biology, Princeton UniversityPrinceton, NJ, USA
| | - Martin Yarmush
- Center for Engineering in Medicine, Shriners Hospitals for Children and Department of Surgery, Massachusetts General Hospital, Harvard Medical SchoolBoston, MA, USA; Department of Biomedical Engineering, Rutgers UniversityNew Brunswick, NJ, USA
| | - Hatem E Sabaawy
- Rutgers Cancer Institute of New Jersey, Rutgers UniversityNew Brunswick, NJ, USA; Department of Medicine, Rutgers Biomedical and Health Sciences (RBHS)-Robert Wood Johnson Medical School, Rutgers UniversityNew Brunswick, NJ, USA
| |
Collapse
|
52
|
Ernst W. Humanized mice in infectious diseases. Comp Immunol Microbiol Infect Dis 2016; 49:29-38. [PMID: 27865261 DOI: 10.1016/j.cimid.2016.08.006] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Revised: 08/12/2016] [Accepted: 08/12/2016] [Indexed: 02/06/2023]
Abstract
The pathogenesis of infectious agents with human tropism can only be properly studied in an in vivo model featuring human cells or tissue. Humanized mice represent a small animal model featuring human cells or tissue that can be infected by human-specific viruses, bacteria, and parasites and also providing a functional human immune system. This makes the analysis of a human immune response to infection possible and allows for preclinical testing of new vaccines and therapeutic agents. Results of various studies using humanized mice to investigate pathogens with human tropism are presented in this review. In addition, the limitations of humanized mice and methods to improve this valuable animal model are discussed.
Collapse
Affiliation(s)
- W Ernst
- Clinic of Gynecology and Obstetrics St. Hedwig, University of Regensburg, Regensburg, Bavaria, Germany.
| |
Collapse
|
53
|
Bartucci M, Ferrari AC, Kim IY, Ploss A, Yarmush M, Sabaawy HE. Personalized Medicine Approaches in Prostate Cancer Employing Patient Derived 3D Organoids and Humanized Mice. Front Cell Dev Biol 2016; 4:64. [PMID: 27446916 PMCID: PMC4917534 DOI: 10.3389/fcell.2016.00064] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Accepted: 06/08/2016] [Indexed: 01/21/2023] Open
Abstract
Prostate cancer (PCa) is the most common malignancy and the second most common cause of cancer death in Western men. Despite its prevalence, PCa has proven very difficult to propagate in vitro. PCa represents a complex organ-like multicellular structure maintained by the dynamic interaction of tumoral cells with parenchymal stroma, endothelial and immune cells, and components of the extracellular matrix (ECM). The lack of PCa models that recapitulate this intricate system has hampered progress toward understanding disease progression and lackluster therapeutic responses. Tissue slices, monolayer cultures and genetically engineered mouse models (GEMM) fail to mimic the complexities of the PCa microenvironment or reproduce the diverse mechanisms of therapy resistance. Moreover, patient derived xenografts (PDXs) are expensive, time consuming, difficult to establish for prostate cancer, lack immune cell-tumor regulation, and often tumors undergo selective engraftments. Here, we describe an interdisciplinary approach using primary PCa and tumor initiating cells (TICs), three-dimensional (3D) tissue engineering, genetic and morphometric profiling, and humanized mice to generate patient-derived organoids for examining personalized therapeutic responses in vitro and in mice co-engrafted with a human immune system (HIS), employing adaptive T-cell- and chimeric antigen receptor- (CAR) immunotherapy. The development of patient specific therapies targeting the vulnerabilities of cancer, when combined with antiproliferative and immunotherapy approaches could help to achieve the full transformative power of cancer precision medicine.
Collapse
Affiliation(s)
- Monica Bartucci
- Rutgers Cancer Institute of New Jersey, Rutgers University New Brunswick, NJ, USA
| | - Anna C Ferrari
- Rutgers Cancer Institute of New Jersey, Rutgers University New Brunswick, NJ, USA
| | - Isaac Yi Kim
- Rutgers Cancer Institute of New Jersey, Rutgers University New Brunswick, NJ, USA
| | - Alexander Ploss
- Rutgers Cancer Institute of New Jersey, Rutgers UniversityNew Brunswick, NJ, USA; Department of Molecular Biology, Princeton UniversityPrinceton, NJ, USA
| | - Martin Yarmush
- Center for Engineering in Medicine, Shriners Hospitals for Children and Department of Surgery, Massachusetts General Hospital, Harvard Medical SchoolBoston, MA, USA; Department of Biomedical Engineering, Rutgers UniversityNew Brunswick, NJ, USA
| | - Hatem E Sabaawy
- Rutgers Cancer Institute of New Jersey, Rutgers UniversityNew Brunswick, NJ, USA; Department of Medicine, Rutgers Biomedical and Health Sciences (RBHS)-Robert Wood Johnson Medical School, Rutgers UniversityNew Brunswick, NJ, USA
| |
Collapse
|
54
|
Kang YK, Ko Y, Choi A, Choi HJ, Seo JH, Lee M, Lee JA. Humanizing NOD/SCID/IL-2Rγnull (NSG) mice using busulfan and retro-orbital injection of umbilical cord blood-derived CD34(+) cells. Blood Res 2016; 51:31-6. [PMID: 27104189 PMCID: PMC4828526 DOI: 10.5045/br.2016.51.1.31] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Revised: 03/02/2016] [Accepted: 03/07/2016] [Indexed: 12/11/2022] Open
Abstract
Background Humanized mouse models are still under development, and various protocols exist to improve human cell engraftment and function. Methods Fourteen NOD/SCID/IL-2Rγnull (NSG) mice (4‒5 wk old) were conditioned with busulfan and injected with human umbilical cord blood (hUCB)-derived CD34+ hematopoietic stem cells (HSC) via retro-orbital sinuses. The bone marrow (BM), spleen, and peripheral blood (PB) were analyzed 8 and 12 weeks after HSC transplantation. Results Most of the NSG mice tolerated the regimen well. The percentage of hCD45+ and CD19+ cells rose significantly in a time-dependent manner. The median percentage of hCD45+cells in the BM was 55.5% at week 8, and 67.2% at week 12. The median percentage of hCD45+ cells in the spleen at weeks 8 and 12 was 42% and 51%, respectively. The median percentage of hCD19+ cells in BM at weeks 8 and 12 was 21.5% and 39%, respectively (P=0.04). Similarly, the median percentage of hCD19+ cells in the spleen at weeks 8 and 12 was 10% and 24%, respectively (P=0.04). The percentage of hCD19+ B cells in PB was 23% at week 12. At week 8, hCD3+ T cells were barely detectable, while hCD7+ was detected in the BM and spleen. The percentage of hCD3+ T cells was 2‒3% at week 12 in the BM, spleen, and PB of humanized NSG mice. Conclusion We adopted a simplified protocol for establishing humanized NSG mice. We observed a higher engraftment rate of human CD45+ cells than earlier studies without any significant toxicity. And human CD45+ cell engraftment at week 8 was comparable to that of week 12.
Collapse
Affiliation(s)
- Young Kyung Kang
- Department of Pediatrics, Korea Cancer Center Hospital, Seoul, Korea
| | - Yunmi Ko
- Department of Pediatrics, Korea Cancer Center Hospital, Seoul, Korea
| | - Aery Choi
- Department of Pediatrics, Korea Cancer Center Hospital, Seoul, Korea
| | - Hyeong Jwa Choi
- Division of Radiation Effect, Korea Institute of Radiological and Medical Sciences, Seoul, Korea
| | - Jin-Hee Seo
- Laboratory Animal Facility, Korea Institute of Radiological and Medical Sciences, Seoul, Korea
| | - Minyoung Lee
- Division of Radiation Effect, Korea Institute of Radiological and Medical Sciences, Seoul, Korea.; Laboratory Animal Facility, Korea Institute of Radiological and Medical Sciences, Seoul, Korea
| | - Jun Ah Lee
- Department of Pediatrics, Korea Cancer Center Hospital, Seoul, Korea
| |
Collapse
|
55
|
Limitations of Current in Vivo Mouse Models for the Study of Chikungunya Virus Pathogenesis. Med Sci (Basel) 2015; 3:64-77. [PMID: 29083392 PMCID: PMC5635755 DOI: 10.3390/medsci3030064] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Revised: 07/20/2015] [Accepted: 07/20/2015] [Indexed: 12/19/2022] Open
Abstract
Chikungunya virus (CHIKV) is an arthropod-borne alphavirus that causes febrile chikungunya fever (CHIKF) in humans. This disease is debilitating and characterized by acute fever onset and chronic incapacitating polyarthralgia. CHIKF pathogenesis remains poorly defined with no approved vaccines and therapies. Recent outbreaks in the Caribbean islands have elevated concerns over the possibility of a global pandemic. Tremendous efforts have been made to develop relevant mouse models to enable the study of infection and immunity against this viral disease. Among them, the more common C57BL/6 mouse model demonstrated the ability to recapitulate the symptoms shown in infected humans, including self-limiting arthritis, myositis, and tenosynovitis. This has facilitated the unraveling of some key factors involved in disease pathogenesis of CHIKF. However, the stark differences in immune response between humans and mouse models necessitate the development of an animal model with an immune system that is more genetically similar to the human system for a better representation. In this paper, we aim to uncover the limitations of the C57BL/6 model and discuss alternative mouse models for CHIKV research.
Collapse
|
56
|
Abstract
During the past decade, the development of humanized mouse models and their general applications in biomedical research greatly accelerated the translation of outcomes obtained from basic research into potential diagnostic and therapeutic strategies in clinic. In this chapter, we firstly present an overview on the history and current progress of diverse humanized mouse models and then focus on those equipped with reconstituted human immune system. The update advancement in the establishment of humanized immune system mice and their applications in the studies of the development of human immune system and the pathogenesis of multiple human immune-related diseases are intensively reviewed here, while the shortcoming and perspective of these potent tools are discussed as well. As a valuable bridge across the gap between bench work and clinical trial, progressive humanized mouse models will undoubtedly continue to play an indispensable role in the wide area of biomedical research.
Collapse
|
57
|
Bae KH, Lee F, Xu K, Keng CT, Tan SY, Tan YJ, Chen Q, Kurisawa M. Microstructured dextran hydrogels for burst-free sustained release of PEGylated protein drugs. Biomaterials 2015; 63:146-57. [PMID: 26100344 DOI: 10.1016/j.biomaterials.2015.06.008] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Revised: 06/08/2015] [Accepted: 06/10/2015] [Indexed: 12/01/2022]
Abstract
Hydrogels have gained significant attention as ideal delivery vehicles for protein drugs. However, the use of hydrogels for protein delivery has been restricted because their porous structures inevitably cause a premature leakage of encapsulated proteins. Here, we report a simple yet effective approach to regulate the protein release kinetics of hydrogels through the creation of microstructures, which serve as a reservoir, releasing their payloads in a controlled manner. Microstructured dextran hydrogels enable burst-free sustained release of PEGylated interferon over 3 months without compromising its bioactivity. These hydrogels substantially extend the circulation half-life of PEGylated interferon, allowing for less frequent dosing in a humanized mouse model of hepatitis C. The present approach opens up possibilities for the development of sustained protein delivery systems for a broad range of pharmaceutical and biomedical applications.
Collapse
Affiliation(s)
- Ki Hyun Bae
- Institute of Bioengineering and Nanotechnology, 31 Biopolis Way, The Nanos, Singapore 138669, Singapore
| | - Fan Lee
- Institute of Bioengineering and Nanotechnology, 31 Biopolis Way, The Nanos, Singapore 138669, Singapore
| | - Keming Xu
- Institute of Bioengineering and Nanotechnology, 31 Biopolis Way, The Nanos, Singapore 138669, Singapore
| | - Choong Tat Keng
- Institute of Molecular and Cell Biology, 61 Biopolis Drive, The Proteos, Singapore 138673, Singapore
| | - Sue Yee Tan
- Institute of Molecular and Cell Biology, 61 Biopolis Drive, The Proteos, Singapore 138673, Singapore
| | - Yee Joo Tan
- Institute of Molecular and Cell Biology, 61 Biopolis Drive, The Proteos, Singapore 138673, Singapore; Department of Microbiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
| | - Qingfeng Chen
- Institute of Molecular and Cell Biology, 61 Biopolis Drive, The Proteos, Singapore 138673, Singapore; Department of Microbiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore; Interdisciplinary Research Group in Infectious Diseases, Singapore-Massachusetts Institute of Technology Alliance for Research and Technology, Singapore 138602, Singapore.
| | - Motoichi Kurisawa
- Institute of Bioengineering and Nanotechnology, 31 Biopolis Way, The Nanos, Singapore 138669, Singapore.
| |
Collapse
|
58
|
Mac Keon S, Ruiz MS, Gazzaniga S, Wainstok R. Dendritic cell-based vaccination in cancer: therapeutic implications emerging from murine models. Front Immunol 2015; 6:243. [PMID: 26042126 PMCID: PMC4438595 DOI: 10.3389/fimmu.2015.00243] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Accepted: 05/06/2015] [Indexed: 01/29/2023] Open
Abstract
Dendritic cells (DCs) play a pivotal role in the orchestration of immune responses, and are thus key targets in cancer vaccine design. Since the 2010 FDA approval of the first cancer DC-based vaccine (Sipuleucel-T), there has been a surge of interest in exploiting these cells as a therapeutic option for the treatment of tumors of diverse origin. In spite of the encouraging results obtained in the clinic, many elements of DC-based vaccination strategies need to be optimized. In this context, the use of experimental cancer models can help direct efforts toward an effective vaccine design. This paper reviews recent findings in murine models regarding the antitumoral mechanisms of DC-based vaccination, covering issues related to antigen sources, the use of adjuvants and maturing agents, and the role of DC subsets and their interaction in the initiation of antitumoral immune responses. The summary of such diverse aspects will highlight advantages and drawbacks in the use of murine models, and contribute to the design of successful DC-based translational approaches for cancer treatment.
Collapse
Affiliation(s)
- Soledad Mac Keon
- Laboratorio de Cancerología, Fundación Instituto Leloir, Instituto de Investigaciones Bioquímicas de Buenos Aires IIBBA-CONICET , Buenos Aires , Argentina
| | - María Sol Ruiz
- Centro de Investigaciones Oncológicas, Fundación para la Investigación, Docencia y Prevención del Cáncer (FUCA) , Buenos Aires , Argentina
| | - Silvina Gazzaniga
- Laboratorio de Biología Tumoral, Departamento de Química Biológica IQUIBICEN-CONICET, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires , Buenos Aires , Argentina
| | - Rosa Wainstok
- Laboratorio de Cancerología, Fundación Instituto Leloir, Instituto de Investigaciones Bioquímicas de Buenos Aires IIBBA-CONICET , Buenos Aires , Argentina ; Laboratorio de Biología Tumoral, Departamento de Química Biológica IQUIBICEN-CONICET, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires , Buenos Aires , Argentina
| |
Collapse
|
59
|
Ebina W, Rossi DJ. Transcription factor-mediated reprogramming toward hematopoietic stem cells. EMBO J 2015; 34:694-709. [PMID: 25712209 DOI: 10.15252/embj.201490804] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
De novo generation of human hematopoietic stem cells (HSCs) from renewable cell types has been a long sought-after but elusive goal in regenerative medicine. Paralleling efforts to guide pluripotent stem cell differentiation by manipulating developmental cues, substantial progress has been made recently toward HSC generation via combinatorial transcription factor (TF)-mediated fate conversion, a paradigm established by Yamanaka's induction of pluripotency in somatic cells by mere four TFs. This review will integrate the recently reported strategies to directly convert a variety of starting cell types toward HSCs in the context of hematopoietic transcriptional regulation and discuss how these findings could be further developed toward the ultimate generation of therapeutic human HSCs.
Collapse
Affiliation(s)
- Wataru Ebina
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA Program in Cellular and Molecular Medicine, Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA, USA
| | - Derrick J Rossi
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA Program in Cellular and Molecular Medicine, Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA, USA Department of Pediatrics, Harvard Medical School, Boston, MA, USA Harvard Stem Cell Institute, Cambridge, MA, USA
| |
Collapse
|
60
|
Villaudy J, Schotte R, Legrand N, Spits H. Critical assessment of human antibody generation in humanized mouse models. J Immunol Methods 2014; 410:18-27. [PMID: 24952244 DOI: 10.1016/j.jim.2014.06.010] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Revised: 06/10/2014] [Accepted: 06/10/2014] [Indexed: 12/31/2022]
Abstract
Immunodeficient mice reconstituted with human hematopoietic stem cells provide a small-animal model for the study of development and function of human hematopoietic cells in vivo. However, in the current models, the immune response, and especially the humoral response by the human immune cells is far from optimal. The B cells found in these mice exhibit an immature and abnormal phenotype correlating with a reduced capacity to produce antigen-specific affinity matured antibodies upon infection or immunization. Herein, we review the current state of knowledge of development, function and antibody production of human B cells and discuss the obstacles for the improvement of these models.
Collapse
Affiliation(s)
- Julien Villaudy
- AIMM Therapeutics, Meibergdreef 59, 1105 BA Amsterdam Zuidoost, Netherlands; Department of Medical Microbiology, Academic Medical Center, University of Amsterdam, Meibergdreef 9, 1105 BA Amsterdam Zuidoost, Netherlands.
| | - Remko Schotte
- AIMM Therapeutics, Meibergdreef 59, 1105 BA Amsterdam Zuidoost, Netherlands.
| | - Nicolas Legrand
- AXENIS, Institut Pasteur, Centre Francois Jacob, 28, rue du Dr. Roux, 75015 Paris, France.
| | - Hergen Spits
- AIMM Therapeutics, Meibergdreef 59, 1105 BA Amsterdam Zuidoost, Netherlands; Department of Cell Biology and Histology, Academic Medical Center, University of Amsterdam, Meibergdreef 9, 1105 BA Amsterdam Zuidoost, Netherlands.
| |
Collapse
|
61
|
Rongvaux A, Willinger T, Martinek J, Strowig T, Gearty SV, Teichmann LL, Saito Y, Marches F, Halene S, Palucka AK, Manz MG, Flavell RA. Development and function of human innate immune cells in a humanized mouse model. Nat Biotechnol 2014; 32:364-72. [PMID: 24633240 PMCID: PMC4017589 DOI: 10.1038/nbt.2858] [Citation(s) in RCA: 597] [Impact Index Per Article: 54.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2013] [Accepted: 02/24/2014] [Indexed: 12/22/2022]
Abstract
Mice repopulated with human hematopoietic cells are a powerful tool for the study of human hematopoiesis and immune function in vivo. However, existing humanized mouse models are unable to support development of human innate immune cells, including myeloid cells and NK cells. Here we describe a mouse strain, called MI(S)TRG, in which human versions of four genes encoding cytokines important for innate immune cell development are knocked in to their respective mouse loci. The human cytokines support the development and function of monocytes/macrophages and natural killer cells derived from human fetal liver or adult CD34+ progenitor cells injected into the mice. Human macrophages infiltrated a human tumor xenograft in MI(S)TRG mice in a manner resembling that observed in tumors obtained from human patients. This humanized mouse model may be used to model the human immune system in scenarios of health and pathology, and may enable evaluation of therapeutic candidates in an in vivo setting relevant to human physiology.
Collapse
Affiliation(s)
- Anthony Rongvaux
- 1] Department of Immunobiology, Yale University, New Haven, Connecticut, USA. [2]
| | - Tim Willinger
- 1] Department of Immunobiology, Yale University, New Haven, Connecticut, USA. [2]
| | - Jan Martinek
- 1] Baylor Institute for Immunology Research, Dallas, Texas, USA. [2] Biomedical studies program, Baylor University, Waco, Texas, USA
| | - Till Strowig
- 1] Department of Immunobiology, Yale University, New Haven, Connecticut, USA. [2]
| | - Sofia V Gearty
- Department of Immunobiology, Yale University, New Haven, Connecticut, USA
| | - Lino L Teichmann
- 1] Department of Laboratory Medicine, Yale University, New Haven, Connecticut, USA. [2] Department of Medicine III, University Hospital Bonn, Bonn, Germany
| | - Yasuyuki Saito
- Division of Hematology, University Hospital Zurich, Zurich, Switzerland
| | | | - Stephanie Halene
- Section of Hematology, Department of Internal Medicine and Yale Comprehensive Cancer Center, Yale University, New Haven, Connecticut, USA
| | | | - Markus G Manz
- Division of Hematology, University Hospital Zurich, Zurich, Switzerland
| | - Richard A Flavell
- 1] Department of Immunobiology, Yale University, New Haven, Connecticut, USA. [2] Howard Hughes Medical Institute, Yale University, New Haven, Connecticut, USA
| |
Collapse
|
62
|
Drake A, Joshi NS, Szeto GL, Zhu E, Eisen HN, Irvine DJ. Koch Institute Symposium on Cancer Immunology and Immunotherapy. Cancer Immunol Res 2014; 1:217-222. [PMID: 24466562 DOI: 10.1158/2326-6066.cir-13-0116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The 12th annual summer symposium of The Koch Institute for Integrative Cancer Research at MIT was held in Cambridge, MA, on June 14th, 1023. The symposium entitled "Cancer Immunology and Immunotherapy" focused on recent advances in preclinical research in basic immunology and biomedical engineering, and their clinical application in cancer therapies. The day-long gathering also provided a forum for discussion and potential collaborations between engineers and clinical investigators. The major topics presented include: (i) enhancement of adoptive cell therapy by engineering to improve the ability and functionality of T-cells against tumor cells; (ii) current therapies using protein and antibody therapeutics to modulate endogenous anti-tumor immunity; and (iii) new technologies to identify molecular targets and assess therapeutic efficacy, and devices to control and target drug delivery more effectively and efficiently.
Collapse
Affiliation(s)
- Adam Drake
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Nikhil S Joshi
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Gregory L Szeto
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Eric Zhu
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139 ; Dept. of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Herman N Eisen
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139 ; Dept. of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Darrell J Irvine
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139 ; Dept. of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139 ; Dept. of Materials Science & Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139 ; Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139 ; Howard Hughes Medical Institute, Chevy Chase, MD 20815
| |
Collapse
|
63
|
Wartha K, Herting F, Hasmann M. Fit-for purpose use of mouse models to improve predictivity of cancer therapeutics evaluation. Pharmacol Ther 2014; 142:351-61. [PMID: 24412280 DOI: 10.1016/j.pharmthera.2014.01.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Accepted: 12/19/2013] [Indexed: 12/21/2022]
Abstract
UNLABELLED Preclinical animal models are useful tools to better understand tumor initiation and progression and to predict the activity of an anticancer agent in the clinic. Ideally, these models should recapitulate the biological characteristics of the tumor and of the related tumor microenvironment (e.g. vasculature, immune cells) in patients. Even if several examples of translational success have been reported it is a matter of fact that clinical trials in oncology often fail to meet their primary endpoints despite encouraging preclinical data. For this reason, there is an increasing need of improved and more predictive models. This review aims to give an overview on existing mouse models for preclinical evaluation of cancer therapeutics and their applicability. Different types of mouse models commonly used for the evaluation of cancer therapeutics are described and considerations for a "fit-for purpose" application of these models for the evaluation of different cancer therapeutics dependent on their mode of action are outlined. Furthermore, considerations for study design and data interpretation to translatability of findings into the clinics are given. CONCLUSION Detailed knowledge of the molecular/biological properties of the respective model, diligent experimental setup, and awareness of its limitations are indispensable prerequisites for the successful translational use of animal models.
Collapse
Affiliation(s)
- K Wartha
- Discovery Oncology, Pharmaceutical Research and Early Development (pRED), Roche Diagnostics GmbH, Penzberg, Germany.
| | - F Herting
- Discovery Oncology, Pharmaceutical Research and Early Development (pRED), Roche Diagnostics GmbH, Penzberg, Germany
| | - M Hasmann
- Discovery Oncology, Pharmaceutical Research and Early Development (pRED), Roche Diagnostics GmbH, Penzberg, Germany
| |
Collapse
|
64
|
Of mice and men: what rodent models don't tell us. Cell Mol Immunol 2013; 10:284-5. [PMID: 23686228 DOI: 10.1038/cmi.2013.21] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2013] [Accepted: 04/20/2013] [Indexed: 02/07/2023] Open
|
65
|
Shultz LD, Brehm MA, Garcia-Martinez JV, Greiner DL. Humanized mice for immune system investigation: progress, promise and challenges. Nat Rev Immunol 2012; 12:786-98. [PMID: 23059428 DOI: 10.1038/nri3311] [Citation(s) in RCA: 715] [Impact Index Per Article: 55.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Significant advances in our understanding of the in vivo functions of human cells and tissues and the human immune system have resulted from the development of 'humanized' mouse strains that are based on severely immunodeficient mice with mutations in the interleukin-2 receptor common γ-chain locus. These mouse strains support the engraftment of a functional human immune system and permit detailed analyses of human immune biology, development and functions. In this Review, we discuss recent advances in the development and utilization of humanized mice, the lessons learnt, the remaining challenges and the promise of using humanized mice for the in vivo study of human immunology.
Collapse
Affiliation(s)
- Leonard D Shultz
- Jackson Laboratory, 600 Main Street, Bar Harbor, Maine 04609, USA.
| | | | | | | |
Collapse
|
66
|
Li P, Xiao Y, Liu Z, Liu P. Using mouse models to study function of transcriptional factors in T cell development. CELL REGENERATION (LONDON, ENGLAND) 2012; 1:8. [PMID: 25408871 PMCID: PMC4230505 DOI: 10.1186/2045-9769-1-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/08/2012] [Accepted: 10/08/2012] [Indexed: 02/03/2023]
Abstract
Laboratory mice have widely been used as tools for basic biological research and models for studying human diseases. With the advances of genetic engineering and conditional knockout (CKO) mice, we now understand hematopoiesis is a dynamic stepwise process starting from hematopoietic stem cells (HSCs) which are responsible for replenishing all blood cells. Transcriptional factors play important role in hematopoiesis. In this review we compile several studies on using genetic modified mice and humanized mice to study function of transcriptional factors in lymphopoiesis, including T lymphocyte and Natural killer (NK) cell development. Finally, we focused on the key transcriptional factor Bcl11b and its function in regulating T cell specification and commitment.
Collapse
Affiliation(s)
- Peng Li
- Key Laboratory of Regenerative Biology, Guangzchou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China ; Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou, China
| | - Yiren Xiao
- Key Laboratory of Regenerative Biology, Guangzchou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China ; Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou, China
| | - Zhixin Liu
- Key Laboratory of Regenerative Biology, Guangzchou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China ; Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou, China
| | - Pentao Liu
- Wellcome Trust Sanger Institute, Hinxton, Cambridge, CB10 1HH UK
| |
Collapse
|