1
|
Jia J, Zhang M, Cao Z, Hu X, Lei S, Zhang Y, Kang X. The rabbit model for spinal tuberculosis: An overview. J Orthop Surg (Hong Kong) 2024; 32:10225536241266703. [PMID: 39033332 DOI: 10.1177/10225536241266703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/23/2024] Open
Abstract
Mycobacterium tuberculosis infection has emerged as a global public health issue, predominantly manifesting as pulmonary tuberculosis. Bone and joint tuberculosis, with spinal tuberculosis accounting for approximately 50%, represents a significant form of extrapulmonary tuberculosis. Over the past years, there has been a rise in the incidence of spinal tuberculosis, and research concerning this area has gained significant attention. At present, animal models provide a means to investigate the pathogenesis, drug resistance, and novel treatment approaches for spinal tuberculosis. New Zealand rabbits, possessing a comparable anatomical structure to humans and capable of reproducing typical pathological features of human tuberculosis, are extensively employed in spinal tuberculosis research using animal models. This article comprehensively evaluates the strengths, considerations in strain selection, various modelling approaches, and practical applications of the rabbit model in studying spinal tuberculosis based on pertinent literature to guide fundamental research in this field by providing valuable insights into appropriate animal model selection.
Collapse
Affiliation(s)
- Jingwen Jia
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, P.R. China
| | - Mingtao Zhang
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, P.R. China
| | - Zhenyu Cao
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, P.R. China
| | - Xuchang Hu
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, P.R. China
| | - Shuanhu Lei
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, P.R. China
| | - Yizhi Zhang
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, P.R. China
| | - Xuewen Kang
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, P.R. China
| |
Collapse
|
2
|
Wang Y, Cho JW, Kastrunes G, Buck A, Razimbaud C, Culhane AC, Sun J, Braun DA, Choueiri TK, Wu CJ, Jones K, Nguyen QD, Zhu Z, Wei K, Zhu Q, Signoretti S, Freeman GJ, Hemberg M, Marasco WA. Immune-restoring CAR-T cells display antitumor activity and reverse immunosuppressive TME in a humanized ccRCC mouse model. iScience 2024; 27:108879. [PMID: 38327771 PMCID: PMC10847687 DOI: 10.1016/j.isci.2024.108879] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 12/01/2023] [Accepted: 01/08/2024] [Indexed: 02/09/2024] Open
Abstract
One of the major barriers that have restricted successful use of chimeric antigen receptor (CAR) T cells in the treatment of solid tumors is an unfavorable tumor microenvironment (TME). We engineered CAR-T cells targeting carbonic anhydrase IX (CAIX) to secrete anti-PD-L1 monoclonal antibody (mAb), termed immune-restoring (IR) CAR G36-PDL1. We tested CAR-T cells in a humanized clear cell renal cell carcinoma (ccRCC) orthotopic mouse model with reconstituted human leukocyte antigen (HLA) partially matched human leukocytes derived from fetal CD34+ hematopoietic stem cells (HSCs) and bearing human ccRCC skrc-59 cells under the kidney capsule. G36-PDL1 CAR-T cells, haploidentical to the tumor cells, had a potent antitumor effect compared to those without immune-restoring effect. Analysis of the TME revealed that G36-PDL1 CAR-T cells restored active antitumor immunity by promoting tumor-killing cytotoxicity, reducing immunosuppressive cell components such as M2 macrophages and exhausted CD8+ T cells, and enhancing T follicular helper (Tfh)-B cell crosstalk.
Collapse
Affiliation(s)
- Yufei Wang
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Harvard Medical School, Boston, MA 02215, USA
| | - Jae-Won Cho
- Harvard Medical School, Boston, MA 02215, USA
- Gene Lay Institute of Immunology and Inflammation, Brigham and Women’s Hospital, Boston, MA 02115, USA
| | - Gabriella Kastrunes
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Alicia Buck
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Cecile Razimbaud
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Aedin C. Culhane
- School of Medicine, University of Limerick, V94 T9PX Limerick, Ireland
| | - Jiusong Sun
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - David A. Braun
- Harvard Medical School, Boston, MA 02215, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Center of Molecular and Cellular Oncology, Yale Cancer Center, Yale School of Medicine, New Haven, CT 06525, USA
| | - Toni K. Choueiri
- Harvard Medical School, Boston, MA 02215, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Catherine J. Wu
- Harvard Medical School, Boston, MA 02215, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Kristen Jones
- Lurie Family Imaging Center, Center for Biomedical Imaging in Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Quang-De Nguyen
- Lurie Family Imaging Center, Center for Biomedical Imaging in Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Zhu Zhu
- Harvard Medical School, Boston, MA 02215, USA
- Division of Rheumatology, Inflammation and Immunity, Brigham and Women’s Hospital, Boston, MA 02115, USA
| | - Kevin Wei
- Harvard Medical School, Boston, MA 02215, USA
- Division of Rheumatology, Inflammation and Immunity, Brigham and Women’s Hospital, Boston, MA 02115, USA
| | - Quan Zhu
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Harvard Medical School, Boston, MA 02215, USA
| | - Sabina Signoretti
- Harvard Medical School, Boston, MA 02215, USA
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA 02115, USA
| | - Gordon J. Freeman
- Harvard Medical School, Boston, MA 02215, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Martin Hemberg
- Harvard Medical School, Boston, MA 02215, USA
- Gene Lay Institute of Immunology and Inflammation, Brigham and Women’s Hospital, Boston, MA 02115, USA
| | - Wayne A. Marasco
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Harvard Medical School, Boston, MA 02215, USA
| |
Collapse
|
3
|
Atamanyuk NI, Andreev SS, Peretykin AA, Styazhkina EV, Obvintseva NA, Tryapitsina GA, Pryakhin EA. Humanized Mice as a Model to Assess the Response of Human Hematopoietic Stem Cells to Irradiation. Bull Exp Biol Med 2023; 176:34-37. [PMID: 38091132 DOI: 10.1007/s10517-023-05962-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Indexed: 12/19/2023]
Abstract
NOD SCID mice were humanized by transplanting human hematopoietic cells isolated from umbilical cord blood. A dose-dependent death of hematopoietic cells and their subsequent recovery were shown after acute external γ-irradiation in the model of humanized mice. The proposed approach can be used for preclinical studies of radioprotective agents and for assessment of the impact of adverse factors on the survival rate and functional properties of human hematopoietic stem cells in vivo.
Collapse
Affiliation(s)
- N I Atamanyuk
- Urals Research Center for Radiation Medicine, Federal Medical-Biological Agency of Russia, Chelyabinsk, Russia.
| | - S S Andreev
- Urals Research Center for Radiation Medicine, Federal Medical-Biological Agency of Russia, Chelyabinsk, Russia
| | - A A Peretykin
- Urals Research Center for Radiation Medicine, Federal Medical-Biological Agency of Russia, Chelyabinsk, Russia
| | - E V Styazhkina
- Urals Research Center for Radiation Medicine, Federal Medical-Biological Agency of Russia, Chelyabinsk, Russia
- Chelyabinsk State University, Chelyabinsk, Russia
| | - N A Obvintseva
- Urals Research Center for Radiation Medicine, Federal Medical-Biological Agency of Russia, Chelyabinsk, Russia
| | - G A Tryapitsina
- Urals Research Center for Radiation Medicine, Federal Medical-Biological Agency of Russia, Chelyabinsk, Russia
- Chelyabinsk State University, Chelyabinsk, Russia
| | - E A Pryakhin
- Urals Research Center for Radiation Medicine, Federal Medical-Biological Agency of Russia, Chelyabinsk, Russia
| |
Collapse
|
4
|
Verhaeghe C, Talikka M, Sewer A, Sierro N, Auberson M, Peric D, Bornand D, Dulize R, Guedj E, Nef P, Tabruyn SP, Hoeng J, Peitsch MC, Lo Sasso G. Tobacco Alkaloid Assessment in a DSS-Induced Colitis Mouse Model with a Fully Humanized Immune System. Int J Mol Sci 2023; 24:ijms24076419. [PMID: 37047398 PMCID: PMC10095104 DOI: 10.3390/ijms24076419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 03/22/2023] [Accepted: 03/24/2023] [Indexed: 04/03/2023] Open
Abstract
Inflammatory bowel disease (IBD) refers to chronic intestinal immune-mediated diseases including two main disease manifestations: ulcerative colitis (UC) and Crohn’s disease (CD). Epidemiological, clinical, and preclinical evidence has highlighted the potential anti-inflammatory properties of naturally occurring alkaloids. In the present study, we investigated the potential anti-inflammatory activities of the tobacco alkaloids nicotine and anatabine in a dextran sulfate sodium (DSS)-induced UC mouse model with a fully humanized immune system. Our results show that nicotine significantly reduced all acute colitis symptoms and improved colitis-specific endpoints, including histopathologically assessed colon inflammation, tissue damage, and mononuclear cell infiltration. The tobacco alkaloid anatabine showed similar effectiveness trends, although they were generally weaker or not significant. Gene expression analysis in the context of biological network models of IBD further pinpointed a possible mechanism by which nicotine attenuated DSS-induced colitis in humanized mice. The current study enables further investigation of possible molecular mechanisms by which tobacco alkaloids attenuate UC symptoms.
Collapse
|
5
|
Bockorny B, Grossman JE, Hidalgo M. Facts and Hopes in Immunotherapy of Pancreatic Cancer. Clin Cancer Res 2022; 28:4606-4617. [PMID: 35775964 DOI: 10.1158/1078-0432.ccr-21-3452] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 04/26/2022] [Accepted: 06/14/2022] [Indexed: 01/24/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) remains one of the most challenging cancers to treat. For patients with advanced and metastatic disease, chemotherapy has yielded only modest incremental benefits, which are not durable. Immunotherapy has revolutionized the treatment of other solid tumors by leading to cures where none existed only a decade ago, yet it has made few inroads with PDAC. A host of trials with promising preclinical data have failed, except for in a small minority of patients with selected biomarkers. There is, however, a glimmer of hope, which we seek to cultivate. In this review, we discuss recent advances in the understanding of the uniquely immunosuppressive tumor microenvironment (TME) in PDAC, learnings from completed trials of checkpoint inhibitors, TME modifiers, cellular and vaccine therapies, oncolytic viruses, and other novel approaches. We go on to discuss our expectations for improved preclinical models of immunotherapy in PDAC, new approaches to modifying the TME including the myeloid compartment, and emerging biomarkers to better select patients who may benefit from immunotherapy. We also discuss improvements in clinical trial design specific to immunotherapy that will help us better measure success when we find it. Finally, we discuss the urgent imperative to better design and execute bold, but rational, combination trials of novel agents designed to cure patients with PDAC.
Collapse
Affiliation(s)
- Bruno Bockorny
- Division of Medical Oncology, Beth Israel Deaconess Medical Center, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | | | - Manuel Hidalgo
- Division of Hematology and Medical Oncology, Weill Cornell Medical College, New York, New York
- New York-Presbyterian Hospital, New York, New York
| |
Collapse
|
6
|
Sahu U, Barth RF, Otani Y, McCormack R, Kaur B. Rat and Mouse Brain Tumor Models for Experimental Neuro-Oncology Research. J Neuropathol Exp Neurol 2022; 81:312-329. [PMID: 35446393 PMCID: PMC9113334 DOI: 10.1093/jnen/nlac021] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Rodent brain tumor models have been useful for developing effective therapies for glioblastomas (GBMs). In this review, we first discuss the 3 most commonly used rat brain tumor models, the C6, 9L, and F98 gliomas, which are all induced by repeated injections of nitrosourea to adult rats. The C6 glioma arose in an outbred Wistar rat and its potential to evoke an alloimmune response is a serious limitation. The 9L gliosarcoma arose in a Fischer rat and is strongly immunogenic, which must be taken into consideration when using it for therapy studies. The F98 glioma may be the best of the 3 but it does not fully recapitulate human GBMs because it is weakly immunogenic. Next, we discuss a number of mouse models. The first are human patient-derived xenograft gliomas in immunodeficient mice. These have failed to reproduce the tumor-host interactions and microenvironment of human GBMs. Genetically engineered mouse models recapitulate the molecular alterations of GBMs in an immunocompetent environment and “humanized” mouse models repopulate with human immune cells. While the latter are rarely isogenic, expensive to produce, and challenging to use, they represent an important advance. The advantages and limitations of each of these brain tumor models are discussed. This information will assist investigators in selecting the most appropriate model for the specific focus of their research.
Collapse
Affiliation(s)
- Upasana Sahu
- From the Department of Neurosurgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Rolf F Barth
- Department of Pathology, The Ohio State University, Columbus, Ohio, USA
| | - Yoshihiro Otani
- From the Department of Neurosurgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Ryan McCormack
- From the Department of Neurosurgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Balveen Kaur
- From the Department of Neurosurgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas, USA
| |
Collapse
|
7
|
Current Status, Barriers, and Future Directions for Humanized Mouse Models to Evaluate Stem Cell–Based Islet Cell Transplant. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1387:89-106. [DOI: 10.1007/5584_2022_711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
8
|
Rojas C, García MP, Polanco AF, González-Osuna L, Sierra-Cristancho A, Melgar-Rodríguez S, Cafferata EA, Vernal R. Humanized Mouse Models for the Study of Periodontitis: An Opportunity to Elucidate Unresolved Aspects of Its Immunopathogenesis and Analyze New Immunotherapeutic Strategies. Front Immunol 2021; 12:663328. [PMID: 34220811 PMCID: PMC8248545 DOI: 10.3389/fimmu.2021.663328] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 05/10/2021] [Indexed: 12/17/2022] Open
Abstract
Periodontitis is an oral inflammatory disease in which the polymicrobial synergy and dysbiosis of the subgingival microbiota trigger a deregulated host immune response, that leads to the breakdown of tooth-supporting tissues and finally tooth loss. Periodontitis is characterized by the increased pathogenic activity of T helper type 17 (Th17) lymphocytes and defective immunoregulation mediated by phenotypically unstable T regulatory (Treg), lymphocytes, incapable of resolving the bone-resorbing inflammatory milieu. In this context, the complexity of the immune response orchestrated against the microbial challenge during periodontitis has made the study of its pathogenesis and therapy difficult and limited. Indeed, the ethical limitations that accompany human studies can lead to an insufficient etiopathogenic understanding of the disease and consequently, biased treatment decision-making. Alternatively, animal models allow us to manage these difficulties and give us the opportunity to partially emulate the etiopathogenesis of periodontitis by inoculating periodontopathogenic bacteria or by placing bacteria-accumulating ligatures around the teeth; however, these models still have limited translational application in humans. Accordingly, humanized animal models are able to emulate human-like complex networks of immune responses by engrafting human cells or tissues into specific strains of immunodeficient mice. Their characteristics enable a viable time window for the study of the establishment of a specific human immune response pattern in an in vivo setting and could be exploited for a wider study of the etiopathogenesis and/or treatment of periodontitis. For instance, the antigen-specific response of human dendritic cells against the periodontopathogen Porphyromonas gingivalis favoring the Th17/Treg response has already been tested in humanized mice models. Hypothetically, the proper emulation of periodontal dysbiosis in a humanized animal could give insights into the subtle molecular characteristics of a human-like local and systemic immune response during periodontitis and support the design of novel immunotherapeutic strategies. Therefore, the aims of this review are: To elucidate how the microbiota-elicited immunopathogenesis of periodontitis can be potentially emulated in humanized mouse models, to highlight their advantages and limitations in comparison with the already available experimental periodontitis non-humanized animal models, and to discuss the potential translational application of using these models for periodontitis immunotherapeutics.
Collapse
Affiliation(s)
- Carolina Rojas
- Periodontal Biology Laboratory, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
| | - Michelle P García
- Periodontal Biology Laboratory, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
| | - Alan F Polanco
- Periodontal Biology Laboratory, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
| | - Luis González-Osuna
- Periodontal Biology Laboratory, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
| | - Alfredo Sierra-Cristancho
- Periodontal Biology Laboratory, Faculty of Dentistry, Universidad de Chile, Santiago, Chile.,Faculty of Dentistry, Universidad Andres Bello, Santiago, Chile
| | - Samanta Melgar-Rodríguez
- Periodontal Biology Laboratory, Faculty of Dentistry, Universidad de Chile, Santiago, Chile.,Department of Conservative Dentistry, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
| | - Emilio A Cafferata
- Periodontal Biology Laboratory, Faculty of Dentistry, Universidad de Chile, Santiago, Chile.,Department of Periodontology, School of Dentistry, Universidad Científica del Sur, Lima, Perú
| | - Rolando Vernal
- Periodontal Biology Laboratory, Faculty of Dentistry, Universidad de Chile, Santiago, Chile.,Department of Conservative Dentistry, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
| |
Collapse
|
9
|
Okuno S, Adachi Y, Terakura S, Julamanee J, Sakai T, Umemura K, Miyao K, Goto T, Murase A, Shimada K, Nishida T, Murata M, Kiyoi H. Spacer Length Modification Facilitates Discrimination between Normal and Neoplastic Cells and Provides Clinically Relevant CD37 CAR T Cells. THE JOURNAL OF IMMUNOLOGY 2021; 206:2862-2874. [PMID: 34099546 DOI: 10.4049/jimmunol.2000768] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 04/14/2021] [Indexed: 11/19/2022]
Abstract
Despite the remarkable initial efficacy of CD19 chimeric Ag receptor T (CAR-T) cell therapy, a high incidence of relapse has been observed. To further increase treatment efficacy and reduce the rate of escape of Ag-negative cells, we need to develop CAR-T cells that target other Ags. Given its restricted expression pattern, CD37 was considered a preferred novel target for immunotherapy in hematopoietic malignancies. Therefore, we designed a CD37-targeting CAR-T (CD37CAR-T) using the single-chain variable fragment of a humanized anti-CD37 Ab, transmembrane and intracellular domains of CD28, and CD3ζ signaling domains. High levels of CD37 expression were confirmed in B cells from human peripheral blood and bone marrow B cell precursors at late developmental stages; by contrast, more limited expression of CD37 was observed in early precursor B cells. Furthermore, we found that human CD37CAR-T cells with longer spacer lengths exhibited high gene transduction efficacy but reduced capacity to proliferate; this may be due to overactivation and fratricide. Spacer length optimization resulted in a modest transduction efficiency together with robust capacity to proliferate. CD37CAR-T cells with optimized spacer length efficiently targeted various CD37+ human tumor cell lines but had no impact on normal leukocytes both in vitro and in vivo. CD37CAR-T cells effectively eradicated Raji cells in xenograft model. Collectively, these results suggested that spacer-optimized CD37CAR-T cells could target CD37-high neoplastic B cells both in vitro and in vivo, with only limited interactions with their normal leukocyte lineages, thereby providing an additional promising therapeutic intervention for patients with B cell malignancies.
Collapse
Affiliation(s)
- Shingo Okuno
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, Nagoya, Japan; and
| | - Yoshitaka Adachi
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, Nagoya, Japan; and
| | - Seitaro Terakura
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, Nagoya, Japan; and
| | - Jakrawadee Julamanee
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, Nagoya, Japan; and.,Division of Clinical Hematology, Faculty of Medicine, Prince of Songkla University, Songkhla, Thailand
| | - Toshiyasu Sakai
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, Nagoya, Japan; and
| | - Koji Umemura
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, Nagoya, Japan; and
| | - Kotaro Miyao
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, Nagoya, Japan; and
| | - Tatsunori Goto
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, Nagoya, Japan; and
| | - Atsushi Murase
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, Nagoya, Japan; and
| | - Kazuyuki Shimada
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, Nagoya, Japan; and
| | - Tetsuya Nishida
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, Nagoya, Japan; and
| | - Makoto Murata
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, Nagoya, Japan; and
| | - Hitoshi Kiyoi
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, Nagoya, Japan; and
| |
Collapse
|
10
|
Mansouri M, Leipzig ND. Advances in removing mass transport limitations for more physiologically relevant in vitro 3D cell constructs. BIOPHYSICS REVIEWS 2021; 2:021305. [PMID: 38505119 PMCID: PMC10903443 DOI: 10.1063/5.0048837] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 05/31/2021] [Indexed: 03/21/2024]
Abstract
Spheroids and organoids are promising models for biomedical applications ranging from human disease modeling to drug discovery. A main goal of these 3D cell-based platforms is to recapitulate important physiological parameters of their in vivo organ counterparts. One way to achieve improved biomimetic architectures and functions is to culture cells at higher density and larger total numbers. However, poor nutrient and waste transport lead to low stability, survival, and functionality over extended periods of time, presenting outstanding challenges in this field. Fortunately, important improvements in culture strategies have enhanced the survival and function of cells within engineered microtissues/organs. Here, we first discuss the challenges of growing large spheroids/organoids with a focus on mass transport limitations, then highlight recent tools and methodologies that are available for producing and sustaining functional 3D in vitro models. This information points toward the fact that there is a critical need for the continued development of novel cell culture strategies that address mass transport in a physiologically relevant human setting to generate long-lasting and large-sized spheroids/organoids.
Collapse
Affiliation(s)
- Mona Mansouri
- Department of Chemical, Biomolecular, and Corrosion Engineering, University of Akron, Akron, Ohio 44325, USA
| | - Nic D. Leipzig
- Department of Chemical, Biomolecular, and Corrosion Engineering, University of Akron, Akron, Ohio 44325, USA
| |
Collapse
|
11
|
Preclinical models and technologies to advance nanovaccine development. Adv Drug Deliv Rev 2021; 172:148-182. [PMID: 33711401 DOI: 10.1016/j.addr.2021.03.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 02/26/2021] [Accepted: 03/01/2021] [Indexed: 12/13/2022]
Abstract
The remarkable success of targeted immunotherapies is revolutionizing cancer treatment. However, tumor heterogeneity and low immunogenicity, in addition to several tumor-associated immunosuppression mechanisms are among the major factors that have precluded the success of cancer vaccines as targeted cancer immunotherapies. The exciting outcomes obtained in patients upon the injection of tumor-specific antigens and adjuvants intratumorally, reinvigorated interest in the use of nanotechnology to foster the delivery of vaccines to address cancer unmet needs. Thus, bridging nano-based vaccine platform development and predicted clinical outcomes the selection of the proper preclinical model will be fundamental. Preclinical models have revealed promising outcomes for cancer vaccines. However, only few cases were associated with clinical responses. This review addresses the major challenges related to the translation of cancer nano-based vaccines to the clinic, discussing the requirements for ex vivo and in vivo models of cancer to ensure the translation of preclinical success to patients.
Collapse
|
12
|
Humanization of Immunodeficient Animals for the Modeling of Transplantation, Graft Versus Host Disease, and Regenerative Medicine. Transplantation 2021; 104:2290-2306. [PMID: 32068660 PMCID: PMC7590965 DOI: 10.1097/tp.0000000000003177] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The humanization of animals is a powerful tool for the exploration of human disease pathogenesis in biomedical research, as well as for the development of therapeutic interventions with enhanced translational potential. Humanized models enable us to overcome biologic differences that exist between humans and other species, while giving us a platform to study human processes in vivo. To become humanized, an immune-deficient recipient is engrafted with cells, tissues, or organoids. The mouse is the most well studied of these hosts, with a variety of immunodeficient strains available for various specific uses. More recently, efforts have turned to the humanization of other animal species such as the rat, which offers some technical and immunologic advantages over mice. These advances, together with ongoing developments in the incorporation of human transgenes and additional mutations in humanized mouse models, have expanded our opportunities to replicate aspects of human allotransplantation and to assist in the development of immunotherapies. In this review, the immune and tissue humanization of various species is presented with an emphasis on their potential for use as models for allotransplantation, graft versus host disease, and regenerative medicine.
Collapse
|
13
|
Fisher E, Padula L, Podack K, O’Neill K, Seavey MM, Jayaraman P, Jasuja R, Strbo N. Induction of SARS-CoV-2 Protein S-Specific CD8+ T Cells in the Lungs of gp96-Ig-S Vaccinated Mice. Front Immunol 2021; 11:602254. [PMID: 33584668 PMCID: PMC7873992 DOI: 10.3389/fimmu.2020.602254] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 12/17/2020] [Indexed: 01/08/2023] Open
Abstract
Given the aggressive spread of COVID-19-related deaths, there is an urgent public health need to support the development of vaccine candidates to rapidly improve the available control measures against SARS-CoV-2. To meet this need, we are leveraging our existing vaccine platform to target SARS-CoV-2. Here, we generated cellular heat shock chaperone protein, glycoprotein 96 (gp96), to deliver SARS-CoV-2 protein S (spike) to the immune system and to induce cell-mediated immune responses. We showed that our vaccine platform effectively stimulates a robust cellular immune response against protein S. Moreover, we confirmed that gp96-Ig, secreted from allogeneic cells expressing full-length protein S, generates powerful, protein S polyepitope-specific CD4+ and CD8+ T cell responses in both lung interstitium and airways. These findings were further strengthened by the observation that protein-S -specific CD8+ T cells were induced in human leukocyte antigen HLA-A2.1 transgenic mice thus providing encouraging translational data that the vaccine is likely to work in humans, in the context of SARS-CoV-2 antigen presentation.
Collapse
Affiliation(s)
- Eva Fisher
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Laura Padula
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Kristin Podack
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Katelyn O’Neill
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL, United States
| | | | | | - Rahul Jasuja
- Heat Biologics, Inc., Morrisville, NC, United States
| | - Natasa Strbo
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL, United States
| |
Collapse
|
14
|
Pham TND, Shields MA, Spaulding C, Principe DR, Li B, Underwood PW, Trevino JG, Bentrem DJ, Munshi HG. Preclinical Models of Pancreatic Ductal Adenocarcinoma and Their Utility in Immunotherapy Studies. Cancers (Basel) 2021; 13:cancers13030440. [PMID: 33503832 PMCID: PMC7865443 DOI: 10.3390/cancers13030440] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Revised: 01/13/2021] [Accepted: 01/20/2021] [Indexed: 12/18/2022] Open
Abstract
Simple Summary Immune checkpoint blockade has provided durable clinical responses in a number of human malignancies, but not in patients with pancreatic cancer. Efforts to understand mechanisms of resistance and increase efficacy of immune checkpoint blockade in pancreatic cancer require the use of appropriate preclinical models in the laboratory. Here, we discuss the benefits, caveats, and potentials for improvement of the most commonly used models, including murine-based and patient-derived models. Abstract The advent of immunotherapy has transformed the treatment landscape for several human malignancies. Antibodies against immune checkpoints, such as anti-PD-1/PD-L1 and anti-CTLA-4, demonstrate durable clinical benefits in several cancer types. However, checkpoint blockade has failed to elicit effective anti-tumor responses in pancreatic ductal adenocarcinoma (PDAC), which remains one of the most lethal malignancies with a dismal prognosis. As a result, there are significant efforts to identify novel immune-based combination regimens for PDAC, which are typically first tested in preclinical models. Here, we discuss the utility and limitations of syngeneic and genetically-engineered mouse models that are currently available for testing immunotherapy regimens. We also discuss patient-derived xenograft mouse models, human PDAC organoids, and ex vivo slice cultures of human PDAC tumors that can complement murine models for a more comprehensive approach to predict response and resistance to immunotherapy regimens.
Collapse
Affiliation(s)
- Thao N. D. Pham
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; (M.A.S.); (C.S.)
- Jesse Brown VA Medical Center, Chicago, IL 60612, USA;
- Correspondence: (T.N.D.P.); (H.G.M.); Tel.: +1-312-503-0312 (T.N.D.P.); +1-312-503-2301 (H.G.M.)
| | - Mario A. Shields
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; (M.A.S.); (C.S.)
- Robert H. Lurie Comprehensive Cancer Center, Chicago, IL 60611, USA
| | - Christina Spaulding
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; (M.A.S.); (C.S.)
- Jesse Brown VA Medical Center, Chicago, IL 60612, USA;
| | - Daniel R. Principe
- Medical Scientist Training Program, University of Illinois, Chicago, IL 60612, USA;
| | - Bo Li
- Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA;
| | - Patrick W. Underwood
- Department of Surgery, University of Florida, Gainesville, FL 32611, USA; (P.W.U.); (J.G.T.)
| | - Jose G. Trevino
- Department of Surgery, University of Florida, Gainesville, FL 32611, USA; (P.W.U.); (J.G.T.)
| | - David J. Bentrem
- Jesse Brown VA Medical Center, Chicago, IL 60612, USA;
- Robert H. Lurie Comprehensive Cancer Center, Chicago, IL 60611, USA
- Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA;
| | - Hidayatullah G. Munshi
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; (M.A.S.); (C.S.)
- Jesse Brown VA Medical Center, Chicago, IL 60612, USA;
- Robert H. Lurie Comprehensive Cancer Center, Chicago, IL 60611, USA
- Correspondence: (T.N.D.P.); (H.G.M.); Tel.: +1-312-503-0312 (T.N.D.P.); +1-312-503-2301 (H.G.M.)
| |
Collapse
|
15
|
Chakravarty D, Huang L, Kahn M, Tewari AK. Immunotherapy for Metastatic Prostate Cancer: Current and Emerging Treatment Options. Urol Clin North Am 2020; 47:487-510. [PMID: 33008499 DOI: 10.1016/j.ucl.2020.07.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The advent of immunotherapy has revolutionized cancer treatment. Prostate cancer has an immunosuppressive microenvironment and a low tumor mutation burden, resulting in low neoantigen expression. The consensus was that immunotherapy would be less effective in prostate cancer. However, recent studies have reported that prostate cancer does have a high number of DNA damage and repair gene defects. Immunotherapies that have been tested in prostate cancer so far have been mainly vaccines and checkpoint inhibitors. A combination of genomically targeted therapies, with approaches to alleviate immune response and thereby make the tumor microenvironment immunologically hot, is promising.
Collapse
Affiliation(s)
- Dimple Chakravarty
- Department of Urology and the Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| | - Li Huang
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, New York, USA; Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Matthew Kahn
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Ashutosh K Tewari
- Department of Urology and the Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
16
|
Tay LS, Palmer N, Panwala R, Chew WL, Mali P. Translating CRISPR-Cas Therapeutics: Approaches and Challenges. CRISPR J 2020; 3:253-275. [PMID: 32833535 PMCID: PMC7469700 DOI: 10.1089/crispr.2020.0025] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
CRISPR-Cas clinical trials have begun, offering a first glimpse at how DNA and RNA targeting could enable therapies for many genetic and epigenetic human diseases. The speedy progress of CRISPR-Cas from discovery and adoption to clinical use is built on decades of traditional gene therapy research and belies the multiple challenges that could derail the successful translation of these new modalities. Here, we review how CRISPR-Cas therapeutics are translated from technological systems to therapeutic modalities, paying particular attention to the therapeutic cascade from cargo to delivery vector, manufacturing, administration, pipelines, safety, and therapeutic target profiles. We also explore potential solutions to some of the obstacles facing successful CRISPR-Cas translation. We hope to illuminate how CRISPR-Cas is brought from the academic bench toward use in the clinic.
Collapse
Affiliation(s)
- Lavina Sierra Tay
- Laboratory of Synthetic Biology and Genome Editing Therapeutics, Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore, Singapore
| | - Nathan Palmer
- Division of Biological Sciences, University of California San Diego, La Jolla, California, USA
| | - Rebecca Panwala
- Department of Bioengineering, University of California San Diego, La Jolla, California, USA
| | - Wei Leong Chew
- Laboratory of Synthetic Biology and Genome Editing Therapeutics, Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore, Singapore
| | - Prashant Mali
- Department of Bioengineering, University of California San Diego, La Jolla, California, USA
| |
Collapse
|
17
|
Kähkönen TE, Halleen JM, Bernoulli J. Immunotherapies and Metastatic Cancers: Understanding Utility and Predictivity of Human Immune Cell Engrafted Mice in Preclinical Drug Development. Cancers (Basel) 2020; 12:cancers12061615. [PMID: 32570871 PMCID: PMC7352707 DOI: 10.3390/cancers12061615] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 06/12/2020] [Accepted: 06/17/2020] [Indexed: 12/14/2022] Open
Abstract
Metastases cause high mortality in several cancers and immunotherapies are expected to be effective in the prevention and treatment of metastatic disease. However, only a minority of patients benefit from immunotherapies. This creates a need for novel therapies that are efficacious regardless of the cancer types and metastatic environments they are growing in. Preclinical immuno-oncology models for studying metastases have long been limited to syngeneic or carcinogenesis-inducible models that have murine cancer and immune cells. However, the translational power of these models has been questioned. Interactions between tumor and immune cells are often species-specific and regulated by different cytokines in mice and humans. For increased translational power, mice engrafted with functional parts of human immune system have been developed. These humanized mice are utilized to advance understanding the role of immune cells in the metastatic process, but increasingly also to study the efficacy and safety of novel immunotherapies. From these aspects, this review will discuss the role of immune cells in the metastatic process and the utility of humanized mouse models in immuno-oncology research for metastatic cancers, covering several models from the perspective of efficacy and safety of immunotherapies.
Collapse
Affiliation(s)
- Tiina E. Kähkönen
- OncoBone Ltd., Kalimenojankuja 3 C 4, FI-90810 Kiviniemi, Finland;
- Correspondence:
| | - Jussi M. Halleen
- OncoBone Ltd., Kalimenojankuja 3 C 4, FI-90810 Kiviniemi, Finland;
| | - Jenni Bernoulli
- Institute of Biomedicine, University of Turku, Kiinamyllynkatu 10, FI-20520 Turku, Finland;
| |
Collapse
|
18
|
Modeling the Efficacy of Oncolytic Adenoviruses In Vitro and In Vivo: Current and Future Perspectives. Cancers (Basel) 2020; 12:cancers12030619. [PMID: 32155969 PMCID: PMC7139921 DOI: 10.3390/cancers12030619] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Revised: 03/03/2020] [Accepted: 03/04/2020] [Indexed: 02/06/2023] Open
Abstract
Oncolytic adenoviruses (OAd) selectively target and lyse tumor cells and enhance anti- tumor immune responses. OAds have been used as promising cancer gene therapies for many years and there are a multitude of encouraging pre-clinical studies. However, translating OAd therapies to the clinic has had limited success, in part due to the lack of realistic pre-clinical models to rigorously test the efficacy of OAds. Solid tumors have a heterogenous and hostile microenvironment that provides many barriers to OAd treatment, including structural and immunosuppressive components that cannot be modeled in two-dimensional tissue culture. To replicate these characteristics and bridge the gap between pre-clinical and clinical success, studies must test OAd therapy in three-dimensional culture and animal models. This review focuses on current methods to test OAd efficacy in vitro and in vivo and the development of new model systems to test both oncolysis and immune stimulatory components of oncolytic adenovirotherapy.
Collapse
|
19
|
Curran M, Mairesse M, Matas-Céspedes A, Bareham B, Pellegrini G, Liaunardy A, Powell E, Sargeant R, Cuomo E, Stebbings R, Betts CJ, Saeb-Parsy K. Recent Advancements and Applications of Human Immune System Mice in Preclinical Immuno-Oncology. Toxicol Pathol 2019; 48:302-316. [PMID: 31847725 DOI: 10.1177/0192623319886304] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Significant advances in immunotherapies have resulted in the increasing need of predictive preclinical models to improve immunotherapeutic drug development, treatment combination, and to prevent or minimize toxicity in clinical trials. Immunodeficient mice reconstituted with human immune system (HIS), termed humanized mice or HIS mice, permit detailed analysis of human immune biology, development, and function. Although this model constitutes a great translational model, some aspects need to be improved as the incomplete engraftment of immune cells, graft versus host disease and the lack of human cytokines and growth factors. In this review, we discuss current HIS platforms, their pathology, and recent advances in their development to improve the quality of human immune cell reconstitution. We also highlight new technologies that can be used to better understand these models and how improved characterization is needed for their application in immuno-oncology safety, efficacy, and new modalities therapy development.
Collapse
Affiliation(s)
- Michelle Curran
- Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom.,Department of Surgery, University of Cambridge and NIHR Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Maelle Mairesse
- Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom
| | - Alba Matas-Céspedes
- Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom.,Department of Surgery, University of Cambridge and NIHR Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Bethany Bareham
- Department of Surgery, University of Cambridge and NIHR Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Giovanni Pellegrini
- Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Ardi Liaunardy
- Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom
| | - Edward Powell
- Department of Surgery, University of Cambridge and NIHR Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Rebecca Sargeant
- Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom
| | - Emanuela Cuomo
- Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom
| | - Richard Stebbings
- Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom
| | - Catherine J Betts
- Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom
| | - Kourosh Saeb-Parsy
- Department of Surgery, University of Cambridge and NIHR Cambridge Biomedical Campus, Cambridge, United Kingdom
| |
Collapse
|
20
|
Essential Role of Human T Cell Leukemia Virus Type 1 orf-I in Lethal Proliferation of CD4 + Cells in Humanized Mice. J Virol 2019; 93:JVI.00565-19. [PMID: 31315992 DOI: 10.1128/jvi.00565-19] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Accepted: 06/11/2019] [Indexed: 12/29/2022] Open
Abstract
Human T cell leukemia virus type 1 (HTLV-1) is the ethological agent of adult T cell leukemia/lymphoma (ATLL) and a number of lymphocyte-mediated inflammatory conditions, including HTLV-1-associated myelopathy/tropical spastic paraparesis. HTLV-1 orf-I encodes two proteins, p8 and p12, whose functions in humans are to counteract innate and adaptive responses and to support viral transmission. However, the in vivo requirements for orf-I expression vary in different animal models. In macaques, the ablation of orf-I expression by mutation of its ATG initiation codon abolishes the infectivity of the molecular clone HTLV-1p12KO In rabbits, HTLV-1p12KO is infective and persists efficiently. We used humanized mouse models to assess the infectivity of both wild-type HTLV-1 (HTLV-1WT) and HTLV-1p12KO We found that NOD/SCID/γC -/- c-kit+ mice engrafted with human tissues 1 day after birth (designated NSG-1d mice) were highly susceptible to infection by HTLV-1WT, with a syndrome characterized by the rapid polyclonal proliferation and infiltration of CD4+ CD25+ T cells into vital organs, weight loss, and death. HTLV-1 clonality studies revealed the presence of multiple clones of low abundance, confirming the polyclonal expansion of HTLV-1-infected cells in vivo HTLV-1p12KO infection in a bone marrow-liver-thymus (BLT) mouse model prone to graft-versus-host disease occurred only following reversion of the orf-I initiation codon mutation within weeks after exposure and was associated with high levels of HTLV-1 DNA in blood and the expansion of CD4+ CD25+ T cells. Thus, the incomplete reconstitution of the human immune system in BLT mice may provide a window of opportunity for HTLV-1 replication and the selection of viral variants with greater fitness.IMPORTANCE Humanized mice constitute a useful model for studying the HTLV-1-associated polyclonal proliferation of CD4+ T cells and viral integration sites in the human genome. The rapid death of infected animals, however, appears to preclude the clonal selection typically observed in human ATLL, which normally develops in 2 to 5% of individuals infected with HTLV-1. Nevertheless, the expansion of multiple clones of low abundance in these humanized mice mirrors the early phase of HTLV-1 infection in humans, providing a useful model to investigate approaches to inhibit virus-induced CD4+ T cell proliferation.
Collapse
|
21
|
Roberts RD, Lizardo MM, Reed DR, Hingorani P, Glover J, Allen-Rhoades W, Fan T, Khanna C, Sweet-Cordero EA, Cash T, Bishop MW, Hegde M, Sertil AR, Koelsche C, Mirabello L, Malkin D, Sorensen PH, Meltzer PS, Janeway KA, Gorlick R, Crompton BD. Provocative questions in osteosarcoma basic and translational biology: A report from the Children's Oncology Group. Cancer 2019; 125:3514-3525. [PMID: 31355930 PMCID: PMC6948723 DOI: 10.1002/cncr.32351] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 04/02/2019] [Accepted: 05/08/2019] [Indexed: 01/06/2023]
Abstract
Patients who are diagnosed with osteosarcoma (OS) today receive the same therapy that patients have received over the last 4 decades. Extensive efforts to identify more effective or less toxic regimens have proved disappointing. As we enter a postgenomic era in which we now recognize OS not as a cancer of mutations but as one defined by p53 loss, chromosomal complexity, copy number alteration, and profound heterogeneity, emerging threads of discovery leave many hopeful that an improving understanding of biology will drive discoveries that improve clinical care. Under the organization of the Bone Tumor Biology Committee of the Children's Oncology Group, a team of clinicians and scientists sought to define the state of the science and to identify questions that, if answered, have the greatest potential to drive fundamental clinical advances. Having discussed these questions in a series of meetings, each led by invited experts, we distilled these conversations into a series of seven Provocative Questions. These include questions about the molecular events that trigger oncogenesis, the genomic and epigenomic drivers of disease, the biology of lung metastasis, research models that best predict clinical outcomes, and processes for translating findings into clinical trials. Here, we briefly present each Provocative Question, review the current scientific evidence, note the immediate opportunities, and speculate on the impact that answered questions might have on the field. We do so with an intent to provide a framework around which investigators can build programs and collaborations to tackle the hardest problems and to establish research priorities for those developing policies and providing funding.
Collapse
Affiliation(s)
- Ryan D Roberts
- Center for Childhood Cancer, Nationwide Children's Hospital, The Ohio State University James Comprehensive Cancer Center, Columbus, Ohio
| | - Michael M Lizardo
- Department of Molecular Oncology, BC Cancer, Provincial Health Services Authority, Vancouver, British Columbia, Canada
| | - Damon R Reed
- Sarcoma Department, Chemical Biology and Molecular Medicine Program and Adolescent and Young Adult Oncology Program, Moffitt Cancer Center, Tampa, Florida
| | - Pooja Hingorani
- Center for Cancer and Blood Disorders, Phoenix Children's Hospital, Phoenix, Arizona
| | - Jason Glover
- Children's Cancer and Blood Disorders Program, Randall Children's Hospital, Portland, Oregon
| | - Wendy Allen-Rhoades
- Department of Pediatrics, Section of Hematology-Oncology, Baylor College of Medicine, Houston, Texas.,Texas Children's Hospital Cancer and Hematology Centers, Houston, Texas
| | - Timothy Fan
- Department of Veterinary Clinical Medicine, University of Illinois, Urbana-Champaign, Illinois
| | - Chand Khanna
- Ethos Vet Health, Woburn, Massachusetts.,Ethos Discovery (501c3), Washington, DC
| | - E Alejandro Sweet-Cordero
- Division of Hematology and Oncology, Department of Pediatrics, University of California San Francisco, San Francisco, California
| | - Thomas Cash
- Department of Pediatrics, Emory University, Children's Healthcare of Atlanta, Atlanta, Georgia
| | - Michael W Bishop
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Meenakshi Hegde
- Center for Cell and Gene Therapy, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas
| | - Aparna R Sertil
- Department of Basic Medical Sciences, College of Medicine Phoenix, University of Arizona, Phoenix, Arizona
| | - Christian Koelsche
- Department of General Pathology, Institute of Pathology, Ruprecht-Karls-University, Heidelberg, Germany
| | - Lisa Mirabello
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - David Malkin
- Hospital for Sick Children, Toronto, Ontario, Canada.,Department of Pediatrics, Division of Hematology/Oncology, University of Toronto, Toronto, Ontario, Canada
| | - Poul H Sorensen
- Department of Molecular Oncology, BC Cancer, Provincial Health Services Authority, Vancouver, British Columbia, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Paul S Meltzer
- Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Katherine A Janeway
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, Massachusetts
| | - Richard Gorlick
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Brian D Crompton
- Dana-Farber Cancer Institute, Boston, and Broad Institute of Harvard and MIT, Cambridge, Massachusetts
| |
Collapse
|
22
|
Villano JS, Vleck SE, Felt SA, Myers DD, Lester PA. Safety Considerations When Working with Humanized Animals. ILAR J 2018; 59:150-160. [PMID: 30541024 DOI: 10.1093/ilar/ily012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 08/31/2018] [Indexed: 01/05/2023] Open
Abstract
Research using laboratory animals has been revolutionized by the creation of humanized animal models, which are immunodeficient animals engrafted with human cells, tissues, or organs. These animal models provide the research community a unique and promising opportunity to mimic a wide variety of disease conditions in humans, from infectious disease to cancer. A vast majority of these models are humanized mice like those injected with human CD34+ hematopoietic stem cells and patient-derived xenografts. With this technology comes the need for the animal research enterprise to understand the inherent and potential risks, such as exposure to bloodborne pathogens, associated with the model development and research applications. Here, we review existing humanized animal models and provide recommendations for their safe use based on regulatory framework and literature. A risk assessment program-from handling the human material to its administration to animals and animal housing-is a necessary initial step in mitigating risks associated with the use of humanized animals in research. Ultimately, establishing institutional policies and guidelines to ensure personnel safety is a legal and ethical responsibility of the research institution as part of the occupational health and safety program and overall animal care and use program.
Collapse
Affiliation(s)
- Jason S Villano
- Unit for Laboratory Animal Medicine, University of Michigan Medical School in Ann Arbor, Michigan
| | - Susan E Vleck
- Department of Environmental Health and Safety at Stanford University in Stanford, California
| | - Stephen A Felt
- Department of Comparative Medicine, Stanford University, Stanford, California
| | - Daniel D Myers
- Unit for Laboratory Animal Medicine, University of Michigan Medical School in Ann Arbor, Michigan.,Department of Surgery, University of Michigan Medical School, University of Michigan, Ann Arbor, Michigan
| | - Patrick A Lester
- Unit for Laboratory Animal Medicine, University of Michigan Medical School in Ann Arbor, Michigan
| |
Collapse
|
23
|
Abstract
Tuberculosis (TB) remains a leading cause of death globally among infectious diseases that has killed more numbers of people than any other infectious diseases. Animal models have become the lynchpin for mimicking human infectious diseases. Research on TB could be facilitated by animal challenge models such as the guinea pig, mice, rabbit and non-human primates. No single model presents all aspects of disease pathogenesis due to considerable differences in disease resistance/susceptibility between these models. Availability of a wide range of animal strains, Mycobacterium tuberculosis strains, route of infection and doses affect the disease progression and intervention outcome. Different animal models have contributed significantly to the drug and vaccine development, identification of biomarkers, understanding of TB immunopathogenesis and host genetic influence on infection. In this review, the commonly used animal models in TB research are discussed along with their advantages and limitations.
Collapse
Affiliation(s)
- Amit Kumar Singh
- ICMR-National JALMA Institute of Leprosy & Other Mycobacterial Diseases, Agra, India
| | - Umesh D Gupta
- ICMR-National JALMA Institute of Leprosy & Other Mycobacterial Diseases, Agra, India
| |
Collapse
|
24
|
Kuryk L, Møller ASW, Jaderberg M. Combination of immunogenic oncolytic adenovirus ONCOS-102 with anti-PD-1 pembrolizumab exhibits synergistic antitumor effect in humanized A2058 melanoma huNOG mouse model. Oncoimmunology 2018; 8:e1532763. [PMID: 30713786 PMCID: PMC6343802 DOI: 10.1080/2162402x.2018.1532763] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 09/21/2018] [Accepted: 09/26/2018] [Indexed: 01/15/2023] Open
Abstract
Malignant melanoma is an aggressive type of skin cancer whose incidence is increasing globally. Although surgery is effective in early stage melanoma, patients with advanced melanoma only have a 20% 5-year survival rate. Hence, combinations of existing and new immunotherapy technologies and immunotherapeutic agents are being evaluated. ONCOS-102 is an oncolytic adenovirus armed with human GM-CSF and an Ad5/3 chimeric capsid. It has shown to be well tolerated in phase I study (NCT01598129) wherein it induced antitumor immunity, infiltration of CD8 + T cells to tumors, and up-regulation of PD-L1. We propose that ONCOS-102 could serve as an immunosensitizer in combination therapies with checkpoint inhibitors. In this preclinical study, we investigated the cytotoxicity of ONCOS-102 and pembrolizumab, an anti-PD-1 antibody, in four human melanoma cell lines, A375, A2058, SK-Mel-2 and SK-Mel-28. Humanized mice engrafted with A2058 melanoma cells showed significant tumor volume reduction after ONCOS-102 treatment. Combination of pembrolizumab with ONCOS-102 reduced tumor volume to an even greater extent, while pembrolizumab (200 µg, or 400 µg) did not show any therapeutic benefit by itself. Body weight loss, and metastasis were not significantly affected by any treatment. These data support the scientific rationale for the ongoing clinical study of combination therapy of ONCOS-102 and pembrolizumab for the treatment of melanoma (NCT03003676).
Collapse
Affiliation(s)
- Lukasz Kuryk
- Clinical Science, Targovax Oy, Helsinki, Finland.,Department of Virology, National Institute of Public Health - National Institute of Hygiene, Warsaw, Poland
| | | | | |
Collapse
|
25
|
McMillan JM, Cobb DA, Lin Z, Banoub MG, Dagur RS, Branch Woods AA, Wang W, Makarov E, Kocher T, Joshi PS, Quadros RM, Harms DW, Cohen SM, Gendelman HE, Gurumurthy CB, Gorantla S, Poluektova LY. Antiretroviral Drug Metabolism in Humanized PXR-CAR-CYP3A-NOG Mice. J Pharmacol Exp Ther 2018; 365:272-280. [PMID: 29476044 PMCID: PMC5878674 DOI: 10.1124/jpet.117.247288] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 02/22/2018] [Indexed: 12/16/2022] Open
Abstract
Antiretroviral drug (ARV) metabolism is linked largely to hepatic cytochrome P450 activity. One ARV drug class known to be metabolized by intestinal and hepatic CYP3A are the protease inhibitors (PIs). Plasma drug concentrations are boosted by CYP3A inhibitors such as cobisistat and ritonavir (RTV). Studies of such drug-drug interactions are limited since the enzyme pathways are human specific. While immune-deficient mice reconstituted with human cells are an excellent model to study ARVs during human immunodeficiency virus type 1 (HIV-1) infection, they cannot reflect human drug metabolism. Thus, we created a mouse strain with the human pregnane X receptor, constitutive androstane receptor, and CYP3A4/7 genes on a NOD.Cg-Prkdcscid Il2rgtm1Sug /JicTac background (hCYP3A-NOG) and used them to evaluate the impact of human CYP3A metabolism on ARV pharmacokinetics. In proof-of-concept studies we used nanoformulated atazanavir (nanoATV) with or without RTV. NOG and hCYP3A-NOG mice were treated weekly with 50 mg/kg nanoATV alone or boosted with nanoformulated ritonavir (nanoATV/r). Plasma was collected weekly and liver was collected at 28 days post-treatment. Plasma and liver atazanavir (ATV) concentrations in nanoATV/r-treated hCYP3A-NOG mice were 2- to 4-fold higher than in replicate NOG mice. RTV enhanced plasma and liver ATV concentrations 3-fold in hCYP3A-NOG mice and 1.7-fold in NOG mice. The results indicate that human CYP3A-mediated drug metabolism is reduced compared with mouse and that RTV differentially affects human gene activity. These differences can affect responses to PIs in humanized mouse models of HIV-1 infection. Importantly, hCYP3A-NOG mice reconstituted with human immune cells can be used for bench-to-bedside translation.
Collapse
Affiliation(s)
- JoEllyn M McMillan
- Department of Pharmacology and Experimental Neuroscience (J.M.M., D.A.C., M.G.B., R.S.D., A.A.B.W., W.W., E.M., T.K., P.S.J., H.E.G., S.G., L.Y.P.), Developmental Neuroscience, Munroe Meyer Institute for Genetics and Rehabilitation (C.B.G.), Department of Pharmaceutical Sciences (Z.L.), Mouse Genome Engineering Core Facility, Vice Chancellor for Research Office (R.M.Q., D.W.H., C.B.G.), and Department of Pathology and Microbiology (S.M.C.), University of Nebraska Medical Center, Omaha, Nebraska
| | - Denise A Cobb
- Department of Pharmacology and Experimental Neuroscience (J.M.M., D.A.C., M.G.B., R.S.D., A.A.B.W., W.W., E.M., T.K., P.S.J., H.E.G., S.G., L.Y.P.), Developmental Neuroscience, Munroe Meyer Institute for Genetics and Rehabilitation (C.B.G.), Department of Pharmaceutical Sciences (Z.L.), Mouse Genome Engineering Core Facility, Vice Chancellor for Research Office (R.M.Q., D.W.H., C.B.G.), and Department of Pathology and Microbiology (S.M.C.), University of Nebraska Medical Center, Omaha, Nebraska
| | - Zhiyi Lin
- Department of Pharmacology and Experimental Neuroscience (J.M.M., D.A.C., M.G.B., R.S.D., A.A.B.W., W.W., E.M., T.K., P.S.J., H.E.G., S.G., L.Y.P.), Developmental Neuroscience, Munroe Meyer Institute for Genetics and Rehabilitation (C.B.G.), Department of Pharmaceutical Sciences (Z.L.), Mouse Genome Engineering Core Facility, Vice Chancellor for Research Office (R.M.Q., D.W.H., C.B.G.), and Department of Pathology and Microbiology (S.M.C.), University of Nebraska Medical Center, Omaha, Nebraska
| | - Mary G Banoub
- Department of Pharmacology and Experimental Neuroscience (J.M.M., D.A.C., M.G.B., R.S.D., A.A.B.W., W.W., E.M., T.K., P.S.J., H.E.G., S.G., L.Y.P.), Developmental Neuroscience, Munroe Meyer Institute for Genetics and Rehabilitation (C.B.G.), Department of Pharmaceutical Sciences (Z.L.), Mouse Genome Engineering Core Facility, Vice Chancellor for Research Office (R.M.Q., D.W.H., C.B.G.), and Department of Pathology and Microbiology (S.M.C.), University of Nebraska Medical Center, Omaha, Nebraska
| | - Raghubendra S Dagur
- Department of Pharmacology and Experimental Neuroscience (J.M.M., D.A.C., M.G.B., R.S.D., A.A.B.W., W.W., E.M., T.K., P.S.J., H.E.G., S.G., L.Y.P.), Developmental Neuroscience, Munroe Meyer Institute for Genetics and Rehabilitation (C.B.G.), Department of Pharmaceutical Sciences (Z.L.), Mouse Genome Engineering Core Facility, Vice Chancellor for Research Office (R.M.Q., D.W.H., C.B.G.), and Department of Pathology and Microbiology (S.M.C.), University of Nebraska Medical Center, Omaha, Nebraska
| | - Amanda A Branch Woods
- Department of Pharmacology and Experimental Neuroscience (J.M.M., D.A.C., M.G.B., R.S.D., A.A.B.W., W.W., E.M., T.K., P.S.J., H.E.G., S.G., L.Y.P.), Developmental Neuroscience, Munroe Meyer Institute for Genetics and Rehabilitation (C.B.G.), Department of Pharmaceutical Sciences (Z.L.), Mouse Genome Engineering Core Facility, Vice Chancellor for Research Office (R.M.Q., D.W.H., C.B.G.), and Department of Pathology and Microbiology (S.M.C.), University of Nebraska Medical Center, Omaha, Nebraska
| | - Weimin Wang
- Department of Pharmacology and Experimental Neuroscience (J.M.M., D.A.C., M.G.B., R.S.D., A.A.B.W., W.W., E.M., T.K., P.S.J., H.E.G., S.G., L.Y.P.), Developmental Neuroscience, Munroe Meyer Institute for Genetics and Rehabilitation (C.B.G.), Department of Pharmaceutical Sciences (Z.L.), Mouse Genome Engineering Core Facility, Vice Chancellor for Research Office (R.M.Q., D.W.H., C.B.G.), and Department of Pathology and Microbiology (S.M.C.), University of Nebraska Medical Center, Omaha, Nebraska
| | - Edward Makarov
- Department of Pharmacology and Experimental Neuroscience (J.M.M., D.A.C., M.G.B., R.S.D., A.A.B.W., W.W., E.M., T.K., P.S.J., H.E.G., S.G., L.Y.P.), Developmental Neuroscience, Munroe Meyer Institute for Genetics and Rehabilitation (C.B.G.), Department of Pharmaceutical Sciences (Z.L.), Mouse Genome Engineering Core Facility, Vice Chancellor for Research Office (R.M.Q., D.W.H., C.B.G.), and Department of Pathology and Microbiology (S.M.C.), University of Nebraska Medical Center, Omaha, Nebraska
| | - Ted Kocher
- Department of Pharmacology and Experimental Neuroscience (J.M.M., D.A.C., M.G.B., R.S.D., A.A.B.W., W.W., E.M., T.K., P.S.J., H.E.G., S.G., L.Y.P.), Developmental Neuroscience, Munroe Meyer Institute for Genetics and Rehabilitation (C.B.G.), Department of Pharmaceutical Sciences (Z.L.), Mouse Genome Engineering Core Facility, Vice Chancellor for Research Office (R.M.Q., D.W.H., C.B.G.), and Department of Pathology and Microbiology (S.M.C.), University of Nebraska Medical Center, Omaha, Nebraska
| | - Poonam S Joshi
- Department of Pharmacology and Experimental Neuroscience (J.M.M., D.A.C., M.G.B., R.S.D., A.A.B.W., W.W., E.M., T.K., P.S.J., H.E.G., S.G., L.Y.P.), Developmental Neuroscience, Munroe Meyer Institute for Genetics and Rehabilitation (C.B.G.), Department of Pharmaceutical Sciences (Z.L.), Mouse Genome Engineering Core Facility, Vice Chancellor for Research Office (R.M.Q., D.W.H., C.B.G.), and Department of Pathology and Microbiology (S.M.C.), University of Nebraska Medical Center, Omaha, Nebraska
| | - Rolen M Quadros
- Department of Pharmacology and Experimental Neuroscience (J.M.M., D.A.C., M.G.B., R.S.D., A.A.B.W., W.W., E.M., T.K., P.S.J., H.E.G., S.G., L.Y.P.), Developmental Neuroscience, Munroe Meyer Institute for Genetics and Rehabilitation (C.B.G.), Department of Pharmaceutical Sciences (Z.L.), Mouse Genome Engineering Core Facility, Vice Chancellor for Research Office (R.M.Q., D.W.H., C.B.G.), and Department of Pathology and Microbiology (S.M.C.), University of Nebraska Medical Center, Omaha, Nebraska
| | - Donald W Harms
- Department of Pharmacology and Experimental Neuroscience (J.M.M., D.A.C., M.G.B., R.S.D., A.A.B.W., W.W., E.M., T.K., P.S.J., H.E.G., S.G., L.Y.P.), Developmental Neuroscience, Munroe Meyer Institute for Genetics and Rehabilitation (C.B.G.), Department of Pharmaceutical Sciences (Z.L.), Mouse Genome Engineering Core Facility, Vice Chancellor for Research Office (R.M.Q., D.W.H., C.B.G.), and Department of Pathology and Microbiology (S.M.C.), University of Nebraska Medical Center, Omaha, Nebraska
| | - Samuel M Cohen
- Department of Pharmacology and Experimental Neuroscience (J.M.M., D.A.C., M.G.B., R.S.D., A.A.B.W., W.W., E.M., T.K., P.S.J., H.E.G., S.G., L.Y.P.), Developmental Neuroscience, Munroe Meyer Institute for Genetics and Rehabilitation (C.B.G.), Department of Pharmaceutical Sciences (Z.L.), Mouse Genome Engineering Core Facility, Vice Chancellor for Research Office (R.M.Q., D.W.H., C.B.G.), and Department of Pathology and Microbiology (S.M.C.), University of Nebraska Medical Center, Omaha, Nebraska
| | - Howard E Gendelman
- Department of Pharmacology and Experimental Neuroscience (J.M.M., D.A.C., M.G.B., R.S.D., A.A.B.W., W.W., E.M., T.K., P.S.J., H.E.G., S.G., L.Y.P.), Developmental Neuroscience, Munroe Meyer Institute for Genetics and Rehabilitation (C.B.G.), Department of Pharmaceutical Sciences (Z.L.), Mouse Genome Engineering Core Facility, Vice Chancellor for Research Office (R.M.Q., D.W.H., C.B.G.), and Department of Pathology and Microbiology (S.M.C.), University of Nebraska Medical Center, Omaha, Nebraska
| | - Channabasavaiah B Gurumurthy
- Department of Pharmacology and Experimental Neuroscience (J.M.M., D.A.C., M.G.B., R.S.D., A.A.B.W., W.W., E.M., T.K., P.S.J., H.E.G., S.G., L.Y.P.), Developmental Neuroscience, Munroe Meyer Institute for Genetics and Rehabilitation (C.B.G.), Department of Pharmaceutical Sciences (Z.L.), Mouse Genome Engineering Core Facility, Vice Chancellor for Research Office (R.M.Q., D.W.H., C.B.G.), and Department of Pathology and Microbiology (S.M.C.), University of Nebraska Medical Center, Omaha, Nebraska
| | - Santhi Gorantla
- Department of Pharmacology and Experimental Neuroscience (J.M.M., D.A.C., M.G.B., R.S.D., A.A.B.W., W.W., E.M., T.K., P.S.J., H.E.G., S.G., L.Y.P.), Developmental Neuroscience, Munroe Meyer Institute for Genetics and Rehabilitation (C.B.G.), Department of Pharmaceutical Sciences (Z.L.), Mouse Genome Engineering Core Facility, Vice Chancellor for Research Office (R.M.Q., D.W.H., C.B.G.), and Department of Pathology and Microbiology (S.M.C.), University of Nebraska Medical Center, Omaha, Nebraska
| | - Larisa Y Poluektova
- Department of Pharmacology and Experimental Neuroscience (J.M.M., D.A.C., M.G.B., R.S.D., A.A.B.W., W.W., E.M., T.K., P.S.J., H.E.G., S.G., L.Y.P.), Developmental Neuroscience, Munroe Meyer Institute for Genetics and Rehabilitation (C.B.G.), Department of Pharmaceutical Sciences (Z.L.), Mouse Genome Engineering Core Facility, Vice Chancellor for Research Office (R.M.Q., D.W.H., C.B.G.), and Department of Pathology and Microbiology (S.M.C.), University of Nebraska Medical Center, Omaha, Nebraska
| |
Collapse
|
26
|
Abstract
CEA TCB is a novel T-cell-bispecific (TCB) antibody targeting the carcinoembryonic antigen (CEA) expressed on tumor cells and the CD3 epsilon chain (CD3e) present on T cells, which is currently in Phase 1 clinical trials (NCT02324257) for the treatment of CEA-positive solid tumors. Because the human CEA (hCEA) binder of CEA TCB does not cross-react with cynomolgus monkey and CEA is absent in rodents, alternative nonclinical safety evaluation approaches were considered. These included the development of a cynomolgus monkey cross-reactive homologous (surrogate) antibody (cyCEA TCB) for its evaluation in cynomolgus monkey and the development of double-transgenic mice, expressing hCEA and human CD3e (hCEA/hCD3e Tg), as a potential alternative species for nonclinical safety studies. However, a battery of nonclinical in vitro/ex vivo experiments demonstrated that neither of the previous approaches provided a suitable and pharmacologically relevant model to assess the safety of CEA TCB. Therefore, an alternative approach, a minimum anticipated biological effect level (MABEL), based on an in vitro tumor lysis assay was used to determine the starting dose for the first-in-human study. Using the most conservative approach to the MABEL assessment, a dose of 52 μg was selected as a safe starting dose for clinical study.
Collapse
|
27
|
Her Z, Yong KSM, Paramasivam K, Tan WWS, Chan XY, Tan SY, Liu M, Fan Y, Linn YC, Hui KM, Surana U, Chen Q. An improved pre-clinical patient-derived liquid xenograft mouse model for acute myeloid leukemia. J Hematol Oncol 2017; 10:162. [PMID: 28985760 PMCID: PMC5639594 DOI: 10.1186/s13045-017-0532-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 09/29/2017] [Indexed: 11/10/2022] Open
Abstract
Background Xenotransplantation of patient-derived AML (acute myeloid leukemia) cells in NOD-scid Il2rγnull (NSG) mice is the method of choice for evaluating this human hematologic malignancy. However, existing models constructed using intravenous injection in adult or newborn NSG mice have inferior engraftment efficiency, poor peripheral blood engraftment, or are difficult to construct. Methods Here, we describe an improved AML xenograft model where primary human AML cells were injected into NSG newborn pups intrahepatically. Results Introduction of primary cells from AML patients resulted in high levels of engraftment in peripheral blood, spleen, and bone marrow (BM) of recipient mice. The phenotype of engrafted AML cells remained unaltered during serial transplantation. The mice developed features that are consistent with human AML including spleen enlargement and infiltration of AML cells into multiple organs. Importantly, we demonstrated that although leukemic stem cell activity is enriched and mediated by CD34+CD117+ subpopulation, CD34+CD117− subpopulation can acquire CD34+CD117+ phenotype through de-differentiation. Lastly, we evaluated the therapeutic potential of Sorafenib and Regorafenib in this AML model and found that periphery and spleen AML cells are sensitive to these treatments, whereas BM provides a protective environment to AML. Conclusions Collectively, our improved model is robust, easy-to-construct, and reliable for pre-clinical AML studies. Electronic supplementary material The online version of this article (10.1186/s13045-017-0532-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Zhisheng Her
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Proteos, 61 Biopolis Drive, Singapore, 138673, Singapore
| | - Kylie Su Mei Yong
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Proteos, 61 Biopolis Drive, Singapore, 138673, Singapore
| | - Kathirvel Paramasivam
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Proteos, 61 Biopolis Drive, Singapore, 138673, Singapore
| | - Wilson Wei Sheng Tan
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Proteos, 61 Biopolis Drive, Singapore, 138673, Singapore
| | - Xue Ying Chan
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Proteos, 61 Biopolis Drive, Singapore, 138673, Singapore
| | - Sue Yee Tan
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Proteos, 61 Biopolis Drive, Singapore, 138673, Singapore
| | - Min Liu
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Proteos, 61 Biopolis Drive, Singapore, 138673, Singapore.,Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Yong Fan
- Key Laboratory for Major Obstetric Diseases of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
| | - Yeh Ching Linn
- Department of Haematology, Singapore General Hospital, Singapore, Singapore
| | - Kam Man Hui
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Proteos, 61 Biopolis Drive, Singapore, 138673, Singapore.,Division of Cellular and Molecular Research, National Cancer Centre, Singapore, Singapore
| | - Uttam Surana
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Proteos, 61 Biopolis Drive, Singapore, 138673, Singapore. .,Department of Pharmacology, National University of Singapore, Singapore, Singapore. .,Bioprocessing Technology Institute, Agency for Science, Technology and Research, Singapore, Singapore.
| | - Qingfeng Chen
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Proteos, 61 Biopolis Drive, Singapore, 138673, Singapore. .,Key Laboratory for Major Obstetric Diseases of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China. .,Division of Cellular and Molecular Research, National Cancer Centre, Singapore, Singapore. .,Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
28
|
Ehlerding EB, England CG, Jiang D, Graves SA, Kang L, Lacognata S, Barnhart TE, Cai W. CD38 as a PET Imaging Target in Lung Cancer. Mol Pharm 2017; 14:2400-2406. [PMID: 28573863 DOI: 10.1021/acs.molpharmaceut.7b00298] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Daratumumab (Darzalex, Janssen Biotech) is a clinically approved antibody targeting CD38 for the treatment of multiple myeloma. However, CD38 is also expressed by other cancer cell types, including lung cancer, where its expression or absence may offer prognostic value. We therefore developed a PET tracer based upon daratumumab for tracking CD38 expression, utilizing murine models of non-small cell lung cancer to verify its specificity. Daratumumab was prepared for radiolabeling with 89Zr (t1/2 = 78.4 h) through conjugation with desferrioxamine (Df). Western blot, flow cytometry, and saturation binding assays were utilized to characterize CD38 expression and binding of daratumumab to three non-small cell lung cancer cell lines: A549, H460, and H358. Murine xenograft models of the cell lines were also generated for further in vivo studies. Longitudinal PET imaging was performed following injection of 89Zr-Df-daratumumab out to 120 h postinjection, and nonspecific uptake was also evaluated through the injection of a radiolabeled control IgG antibody in A549 mice, 89Zr-Df-IgG. Ex vivo biodistribution and histological analyses were also performed after the terminal imaging time point at 120 h postinjection. Through cellular studies, A549 cells were found to express higher levels of CD38 than the H460 or H358 cell lines. PET imaging and ex vivo biodistribution studies verified in vitro trends, with A549 tumor uptake peaking at 8.1 ± 1.2%ID/g at 120 h postinjection according to PET analysis, and H460 and H358 at lower levels at the same time point (6.7 ± 0.7%ID/g and 5.1 ± 0.4%ID/g, respectively; n = 3 or 4). Injection of a nonspecific radiolabeled IgG into A549 tumor-bearing mice also demonstrated lower tracer uptake of 4.4 ± 1.3%ID/g at 120 h. Immunofluorescent staining of tumor tissues showed higher staining levels present in A549 tissues over H460 and H358. Thus, 89Zr-Df-daratumumab is able to image CD38-expressing tissues in vivo using PET, as verified through the exploration of non-small cell lung cancer models in this study. This agent therefore holds potential to image CD38 in other malignancies and aid in patient stratification and elucidation of the biodistribution of CD38.
Collapse
Affiliation(s)
- Emily B Ehlerding
- Department of Medical Physics, University of Wisconsin-Madison , Madison, Wisconsin 53705, United States
| | - Christopher G England
- Department of Medical Physics, University of Wisconsin-Madison , Madison, Wisconsin 53705, United States
| | - Dawei Jiang
- Department of Radiology, University of Wisconsin-Madison , Madison, Wisconsin 53705, United States
| | - Stephen A Graves
- Department of Radiology, University of Wisconsin-Madison , Madison, Wisconsin 53705, United States
| | - Lei Kang
- Department of Radiology, University of Wisconsin-Madison , Madison, Wisconsin 53705, United States
| | - Saige Lacognata
- Department of Radiology, University of Wisconsin-Madison , Madison, Wisconsin 53705, United States
| | - Todd E Barnhart
- Department of Medical Physics, University of Wisconsin-Madison , Madison, Wisconsin 53705, United States
| | - Weibo Cai
- Department of Medical Physics, University of Wisconsin-Madison , Madison, Wisconsin 53705, United States.,Department of Radiology, University of Wisconsin-Madison , Madison, Wisconsin 53705, United States.,Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health , Madison, Wisconsin 53705, United States
| |
Collapse
|
29
|
Creation of an immunodeficient HLA-transgenic mouse (HUMAMICE) and functional validation of human immunity after transfer of HLA-matched human cells. PLoS One 2017; 12:e0173754. [PMID: 28399128 PMCID: PMC5388326 DOI: 10.1371/journal.pone.0173754] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 02/27/2017] [Indexed: 11/19/2022] Open
Abstract
Research on human immunology has been hindered by the lack of optimal small animal models, given that the protective immune responses of human and non-human species show significant differences. However, due to ethical constraints[1] and the high cost of clinical trials, it is urgent to improve the current animal models that can mimic faithfully human physiology, particularly the human immune system (HIS). HIS mice had been generated recently by engrafting human hematopoietic stem cells (hHSCs) or human peripheral mononuclear cells (hPBMCs) into highly immuno-deficient mice such as NSG, NOG or NRG mice. However, a major experimental drawback for studies using these models is the rapid onset of Graft-versus-Host Disease (GvHD). In the present study, we overcome this limitation by generating new immuno-deficient mice named "HUMAMICE" (HLA-A2+/+/DR1+/+/H-2-β2m-/-/IAβ-/-/Rag2-/-/IL2rγ-/-/Perf-/- mice), which expressed human HLA molecules instead of mouse MHC molecules (H-2), and whose immuno-deficient status was reversed by transferring functional HLA-matched PBMCs thus producing mice with an immuno-competent status with a functional human immune system. We showed that in this HLA-matched context, the hPBMC-transfer led to high lymphocytes engraftment rates without GvHD over three months in this novel mouse model. Furthermore, to evaluate the utility of the hPBMC-HUMAMICE, we immunized them with commercial vaccine of Hepatitis B virus (HBsAg, Hepvac@) which resulted in robust and reproducible production of high levels of HBsAg-specific antibodies, implying that both transferred T and B lymphocytes were functional in HUMAMICE. These responses are comparable to those observed in human clinical trials with this identical vaccine. In conclusion, these findings indicated that the HLA-matched-hPBMC-HUMAMICE represents a promising model for dissecting human immune responses in various human diseases, including infectious diseases, cancers and tumors, and to facilitate the development of novel vaccines and cellular therapies.
Collapse
|
30
|
Mouse Models in Prostate Cancer Translational Research: From Xenograft to PDX. BIOMED RESEARCH INTERNATIONAL 2016; 2016:9750795. [PMID: 27294148 PMCID: PMC4887629 DOI: 10.1155/2016/9750795] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Accepted: 04/21/2016] [Indexed: 12/20/2022]
Abstract
Despite the advancement of clinical and preclinical research on PCa, which resulted in the last five years in a decrement of disease incidence by 3-4%, it remains the most frequent cancer in men and the second for mortality rate. Based on this evidence we present a brief dissertation on numerous preclinical models, comparing their advantages and disadvantages; among this we report the PDX mouse models that show greater fidelity to the disease, in terms of histopathologic features of implanted tumor, gene and miRNA expression, and metastatic pattern, well describing all tumor progression stages; this characteristic encourages the translation of preclinical results. These models become particularly useful in meeting the need of new treatments identification that eradicate PCa bone metastases growing, clarifying pathway of angiogenesis, identifying castration-resistant stem-like cells, and studying the antiandrogen therapies. Also of considerable interest are the studies of 3D cell cultures derived from PDX, which have the ability to maintain PDX cell viability with continued native androgen receptor expression, also showing a differential sensitivity to drugs. 3D PDX PCa may represent a diagnostic platform for the rapid assessment of drugs and push personalized medicine. Today the development of preclinical models in vitro and in vivo is necessary in order to obtain increasingly reliable answers before reaching phase III of the drug discovery.
Collapse
|
31
|
Burel JG, Apte SH, Doolan DL. Systems Approaches towards Molecular Profiling of Human Immunity. Trends Immunol 2016; 37:53-67. [DOI: 10.1016/j.it.2015.11.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Revised: 11/14/2015] [Accepted: 11/15/2015] [Indexed: 12/12/2022]
|
32
|
Halvorsen EC, Mahmoud SM, Bennewith KL. Emerging roles of regulatory T cells in tumour progression and metastasis. Cancer Metastasis Rev 2015; 33:1025-41. [PMID: 25359584 DOI: 10.1007/s10555-014-9529-x] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The metastasis of cancer is a complex and life-threatening process that is only partially understood. Immune suppressive cells are recognized as important contributors to tumour progression and may also promote the development and growth of tumour metastases. Specifically, regulatory T cells (Tregs) have been found to promote primary tumour progression, and emerging pre-clinical data suggests that Tregs may promote metastasis and metastatic tumour growth. While the precise role that Tregs play in metastatic progression is understudied, recent findings have indicated that by suppressing innate and adaptive anti-tumour immunity, Tregs may shield tumour cells from immune detection, and thereby allow tumour cells to survive, proliferate and acquire characteristics that facilitate dissemination. This review will highlight our current understanding of Tregs in metastasis, including an overview of pre-clinical findings and discussion of clinical data regarding Tregs and therapeutic outcome. Evolving strategies to directly ablate Tregs or to inhibit their function will also be discussed. Improving our understanding of how Tregs may influence tumour metastasis may lead to novel treatments for metastatic cancer.
Collapse
Affiliation(s)
- Elizabeth C Halvorsen
- Department of Integrative Oncology, British Columbia Cancer Agency, 9-202, 675 West 10th Avenue, Vancouver, British Columbia, V5Z 1L3, Canada
| | | | | |
Collapse
|
33
|
Roth MD, Harui A. Human tumor infiltrating lymphocytes cooperatively regulate prostate tumor growth in a humanized mouse model. J Immunother Cancer 2015; 3:12. [PMID: 25901284 PMCID: PMC4404579 DOI: 10.1186/s40425-015-0056-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Accepted: 03/06/2015] [Indexed: 12/17/2022] Open
Abstract
Background The complex interactions that occur between human tumors, tumor infiltrating lymphocytes (TIL) and the systemic immune system are likely to define critical factors in the host response to cancer. While conventional animal models have identified an array of potential anti-tumor therapies, mouse models often fail to translate into effective human treatments. Our goal is to establish a humanized tumor model as a more effective pre-clinical platform for understanding and manipulating TIL. Methods The immune system in NOD/SCID/IL-2Rγnull (NSG) mice was reconstituted by the co-administration of human peripheral blood lymphocytes (PBL) or subsets (CD4+ or CD8+) and autologous human dendritic cells (DC), and animals simultaneously challenged by implanting human prostate cancer cells (PC3 line). Tumor growth was evaluated over time and the phenotype of recovered splenocytes and TIL characterized by flow cytometry and immunohistochemistry (IHC). Serum levels of circulating cytokines and chemokines were also assessed. Results A tumor-bearing huPBL-NSG model was established in which human leukocytes reconstituted secondary lymphoid organs and promoted the accumulation of TIL. These TIL exhibited a unique phenotype when compared to splenocytes with a predominance of CD8+ T cells that exhibited increased expression of CD69, CD56, and an effector memory phenotype. TIL from huPBL-NSG animals closely matched the features of TIL recovered from primary human prostate cancers. Human cytokines were readily detectible in the serum and exhibited a different profile in animals implanted with PBL alone, tumor alone, and those reconstituted with both. Immune reconstitution slowed but could not eliminate tumor growth and this effect required the presence of CD4+ T cell help. Conclusions Simultaneous implantation of human PBL, DC and tumor results in a huPBL-NSG model that recapitulates the development of human TIL and allows an assessment of tumor and immune system interaction that cannot be carried out in humans. Furthermore, the capacity to manipulate individual features and cell populations provides an opportunity for hypothesis testing and outcome monitoring in a humanized system that may be more relevant than conventional mouse models.
Collapse
Affiliation(s)
- Michael D Roth
- Division of Pulmonary & Critical Care, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-1690 USA
| | - Airi Harui
- Division of Pulmonary & Critical Care, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-1690 USA
| |
Collapse
|
34
|
Wang Y, Yuan L, Yang XM, Wei D, Wang B, Sun XX, Feng F, Nan G, Wang Y, Chen ZN, Bian H. A chimeric antibody targeting CD147 inhibits hepatocellular carcinoma cell motility via FAK-PI3K-Akt-Girdin signaling pathway. Clin Exp Metastasis 2014; 32:39-53. [PMID: 25424030 DOI: 10.1007/s10585-014-9689-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2014] [Accepted: 11/19/2014] [Indexed: 01/11/2023]
Abstract
CD147 is expressed at low levels in normal tissues but frequently highly expressed in a wide range of tumor types such as lung, breast, and liver and therefore it is a potentially unique therapeutic target for these diverse tumor types. We previously generated a murine antibody HAb18 which suppresses matrix met al.loproteinase-2 and matrix metalloproteinase-9 secretion, attenuates cell invasion by blocking the CD147 molecule in tumor cells. Here, we generated a chimeric antibody containing the variable heavy and variable light chains of murine HAb18 and the constant regions of human IgG1γ1 and human κ chain as a potential therapeutic agent (designated cHAb18). Quantitative measurement of cHAb18 antibody affinity for antigen CD147 with surface plasmon resonance showed the equilibrium dissociation constant KD was 2.66 × 10(-10) mol/L, similar to that of KD 2.73 × 10(-10) mol/L for murine HAb18. cHAb18 induced antibody-dependent cell-mediated cytotoxicity in two hepatocellular carcinoma cell lines, SMMC-7721 and Huh-7 cells. It inhibited cancer invasion and migration in hepatocellular carcinoma cells by specifically blocking CD147. Except for the depression of matrix metalloproteinase-2 and matrix metalloproteinase-9 expressions, cHAb18 antibody suppressed cell motility by rearrangement of actin cytoskeleton, which was probably induced by decreasing the phosphorylation of focal adhesion kinase, phosphatidylinositide-3 kinase (PI3K), Akt, and Girdin in the integrin signaling pathway. In an orthotopic model of hepatocellular carcinoma in BALB/c nude mice, cHAb18 treatment effectively reduced the tumor metastasis in liver and prolonged the survival. These findings reveal new therapeutic potential for cHAb18 antibody targeting CD147 on tumor therapy.
Collapse
Affiliation(s)
- Yuan Wang
- State Key Laboratory of Cancer Biology, Department of Cell Biology and Cell Engineering Research Center, Fourth Military Medical University, Xi'an, 710032, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|