1
|
Crouigneau R, Li YF, Auxillos J, Goncalves-Alves E, Marie R, Sandelin A, Pedersen SF. Mimicking and analyzing the tumor microenvironment. CELL REPORTS METHODS 2024; 4:100866. [PMID: 39353424 DOI: 10.1016/j.crmeth.2024.100866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 07/22/2024] [Accepted: 09/09/2024] [Indexed: 10/04/2024]
Abstract
The tumor microenvironment (TME) is increasingly appreciated to play a decisive role in cancer development and response to therapy in all solid tumors. Hypoxia, acidosis, high interstitial pressure, nutrient-poor conditions, and high cellular heterogeneity of the TME arise from interactions between cancer cells and their environment. These properties, in turn, play key roles in the aggressiveness and therapy resistance of the disease, through complex reciprocal interactions between the cancer cell genotype and phenotype, and the physicochemical and cellular environment. Understanding this complexity requires the combination of sophisticated cancer models and high-resolution analysis tools. Models must allow both control and analysis of cellular and acellular TME properties, and analyses must be able to capture the complexity at high depth and spatial resolution. Here, we review the advantages and limitations of key models and methods in order to guide further TME research and outline future challenges.
Collapse
Affiliation(s)
- Roxane Crouigneau
- Section for Cell Biology and Physiology, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Yan-Fang Li
- Department of Health Technology, Technical University of Denmark, 2800 Kongens Lyngby, Denmark
| | - Jamie Auxillos
- Section for Computational and RNA Biology, Department of Biology, University of Copenhagen, Copenhagen, Denmark; Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen, Denmark
| | - Eliana Goncalves-Alves
- Section for Cell Biology and Physiology, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Rodolphe Marie
- Department of Health Technology, Technical University of Denmark, 2800 Kongens Lyngby, Denmark.
| | - Albin Sandelin
- Section for Computational and RNA Biology, Department of Biology, University of Copenhagen, Copenhagen, Denmark; Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen, Denmark.
| | - Stine Falsig Pedersen
- Section for Cell Biology and Physiology, Department of Biology, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
2
|
Panisello C, Aschero R, Martinez-Moreno A, Roca Ho H, Falgas A, González-Navarro EA, Carabelli J, Pradenas E, Lázaro-Díez M, Prado JG, Blanco J, Carrillo J, Juan M, Carcaboso ÁM, Bueno C, Menendez P. NSGS mice humanized with cord blood mononuclear cells show sustained and functional myeloid-lymphoid representation with limited graft-versus-host disease. J Immunother Cancer 2024; 12:e009198. [PMID: 39379097 PMCID: PMC11459296 DOI: 10.1136/jitc-2024-009198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/16/2024] [Indexed: 10/10/2024] Open
Abstract
Humanized immunodeficient mice serve as critical models for investigating the functional interplay between transplanted human cells and a pre-reconstituted human immune system. These models facilitate the study of molecular and cellular pathogenic mechanisms and enable the evaluation of the efficacy and toxicity of immunotherapies, thereby accelerating their preclinical and clinical development. Current strategies rely on inefficient, long-term/delayed hematopoietic reconstitution by CD34+ hematopoietic progenitors or short-term reconstitution with peripheral blood mononuclear cells (PB-MNCs) associated with high rates of graft-versus-host disease (GvHD) and an inefficient representation of immune cell populations. Here, we hypothesized that immunologically naïve cord blood mononuclear cells (CB-MNCs) could serve as a superior alternative, providing long-lasting and functionally effective immune reconstitution. We conducted a comprehensive comparison between the non-obese diabetic (NOD).Cg-Prkdc∧ˆscid-IL2rg∧ˆtm1Wjl/SzJ (NSG) and NSG-Tg(CMV-IL3,CSF2,KITLG)∧ˆ1Eav/MloySzJ (NSGS) immunodeficient mouse models following humanization with either PB-MNCs or CB-MNCs. We assessed the engraftment dynamics of various human immune cells over time and monitored the development of GvHD in both models. For the most promising model, we extensively evaluated immune cell functionality in vitro and in vivo using sarcoma and leukemia xenografts. Humanizing NSGS mice with CB-MNCs results in a rapid, robust, and sustained representation of a diverse range of functional human lymphoid and myeloid cell populations while minimizing GvHD incidence. In this model, human immune cell populations significantly impair the growth and engraftment of sarcoma and B-cell acute lymphoblastic leukemia cells, with a significant inverse correlation between immune cell levels and tumor growth. This study establishes a fast, efficient, and reliable in vivo platform for various applications in cancer immunotherapy, particularly for exploring the complex interactions between cancer cells, immune cells, and the tumor microenvironment in vivo, prior to clinical development.
Collapse
Affiliation(s)
- Carla Panisello
- Josep Carreras Leukaemia Research Institute (IJC), Barcelona, Spain
- Red Española de Terapias Avanzadas (TERAV), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Germans Trias i Pujol Research Institute (IGTP), Badalona, Spain
| | - Rosario Aschero
- Paediatric Cancer Treatment, Sant Joan de Deu Research Institute, Barcelona, Spain
| | - Alba Martinez-Moreno
- Josep Carreras Leukaemia Research Institute (IJC), Barcelona, Spain
- Red Española de Terapias Avanzadas (TERAV), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Heleia Roca Ho
- Josep Carreras Leukaemia Research Institute (IJC), Barcelona, Spain
- Red Española de Terapias Avanzadas (TERAV), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Aida Falgas
- Josep Carreras Leukaemia Research Institute (IJC), Barcelona, Spain
- Red Española de Terapias Avanzadas (TERAV), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Europa Azucena González-Navarro
- Department of Immunology and Immunotherapy, Hospital Clínic Barcelona, Barcelona, Spain
- August Pi i Sunyer Institute of Biomedical Research (IDIBAPS), Barcelona, Spain
| | | | | | | | - Julia G Prado
- Germans Trias i Pujol Research Institute (IGTP), Badalona, Spain
- IrsiCaixa AIDS Research Institute, Badalona, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Julià Blanco
- Germans Trias i Pujol Research Institute (IGTP), Badalona, Spain
- IrsiCaixa AIDS Research Institute, Badalona, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Department of Microbiology, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Jorge Carrillo
- Germans Trias i Pujol Research Institute (IGTP), Badalona, Spain
- IrsiCaixa AIDS Research Institute, Badalona, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Manel Juan
- Red Española de Terapias Avanzadas (TERAV), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Department of Immunology and Immunotherapy, Hospital Clínic Barcelona, Barcelona, Spain
- August Pi i Sunyer Institute of Biomedical Research (IDIBAPS), Barcelona, Spain
| | - Ángel M Carcaboso
- Paediatric Cancer Treatment, Sant Joan de Deu Research Institute, Barcelona, Spain
| | - Clara Bueno
- Josep Carreras Leukaemia Research Institute (IJC), Barcelona, Spain
- Red Española de Terapias Avanzadas (TERAV), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Centro Investigación Biomédica en Red en Oncología (CIBERONC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Pablo Menendez
- Josep Carreras Leukaemia Research Institute (IJC), Barcelona, Spain
- Red Española de Terapias Avanzadas (TERAV), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Centro Investigación Biomédica en Red en Oncología (CIBERONC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
- Departament of Biomedicine, School of Medicine, Universitat de Barcelona, Barcelona, Spain
| |
Collapse
|
3
|
Ménoret S, Renart-Depontieu F, Martin G, Thiam K, Anegon I. Efficient generation of human immune system rats using human CD34 + cells. Stem Cell Reports 2024; 19:1255-1263. [PMID: 39151431 PMCID: PMC11411320 DOI: 10.1016/j.stemcr.2024.07.005] [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: 10/02/2023] [Revised: 07/20/2024] [Accepted: 07/21/2024] [Indexed: 08/19/2024] Open
Abstract
Human immune system (HIS) mice generated using human CD34+ hematopoietic stem cells serve as a pivotal model for the in vivo evaluation of immunotherapies for humans. Yet, HIS mice possess certain limitations. Rats, due to their size and comprehensive immune system, hold promise for translational experiments. Here, we describe an efficacious method for long-term immune humanization, through intrahepatic injection of hCD34+ cells in newborn immunodeficient rats expressing human SIRPα. In contrast to HIS mice and similar to humans, HIS rats showed in blood a predominance of T cells, followed by B cells. Immune humanization was also high in central and secondary lymphoid organs. HIS rats treated with the anti-human CD3 antibody were depleted of human T cells, and human cytokines were detected in sera. We describe for the first time a method to efficiently generate HIS rats. HIS rats have the potential to be a useful model for translational immunology.
Collapse
Affiliation(s)
- Séverine Ménoret
- Nantes Université, CHU Nantes, Inserm, CNRS, SFR Santé, Inserm UMS 016, CNRS UMS 3556, F-44000 Nantes, France; INSERM, Centre de Recherche en Transplantation et Immunologie UMR1064, Nantes Université, Nantes, France.
| | | | | | | | - Ignacio Anegon
- INSERM, Centre de Recherche en Transplantation et Immunologie UMR1064, Nantes Université, Nantes, France.
| |
Collapse
|
4
|
Kim KH, Lee SW, Baek IJ, Song HY, Jo SJ, Ryu JW, Ryu SH, Seo JH, Kim JC, Heo SH. CD47;Rag2;IL-2rγ triple knock-out mice pre-conditioning with busulfan could be a novel platform for generating hematopoietic stem cells engrafted humanized mice. Front Immunol 2024; 15:1365946. [PMID: 39131155 PMCID: PMC11310007 DOI: 10.3389/fimmu.2024.1365946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 07/08/2024] [Indexed: 08/13/2024] Open
Abstract
Introduction Humanized mouse models to recapitulate human biological systems still have limitations, such as the onset of lethal graft-versus-host disease (GvHD), a variable success rate, and the low accessibility of total body irradiation (TBI). Recently, mice modified with the CD47-SIRPA axis have been studied to improve humanized mouse models. However, such trials have been rarely applied in NOD mice. In this study, we created a novel mouse strain, NOD-CD47nullRag2nullIL-2rγnull (RTKO) mice, and applied it to generate humanized mice. Methods Four-week-old female NOD-Rag2nullIL-2rγnull (RID) and RTKO mice pre-conditioned with TBI or busulfan (BSF) injection were used for generating human CD34+ hematopoietic stem cell (HSC) engrafted humanized mice. Clinical signs were observed twice a week, and body weight was measured once a week. Flow cytometry for human leukocyte antigens was performed at intervals of four weeks or two weeks, and mice were sacrificed at 48 weeks after HSC injection. Results For a long period from 16 to 40 weeks post transplantation, the percentage of hCD45 was mostly maintained above 25% in all groups, and it was sustained the longest and highest in the RTKO BSF group. Reconstruction of human leukocytes, including hCD3, was also most prominent in the RTKO BSF group. Only two mice died before 40 weeks post transplantation in all groups, and there were no life-threatening GvHD lesions except in the dead mice. The occurrence of GvHD has been identified as mainly due to human T cells infiltrating tissues and their related cytokines. Discussion Humanized mouse models under all conditions applied in this study are considered suitable models for long-term experiments based on the improvement of human leukocytes reconstruction and the stable animal health. Especially, RTKO mice pretreated with BSF are expected to be a valuable platform not only for generating humanized mice but also for various immune research fields.
Collapse
Affiliation(s)
- Kang-Hyun Kim
- Convergence Medicine Research Center, Asan Medical Center, Seoul, Republic of Korea
- College of Veterinary Medicine, Chonnam National University, Gwangju, Republic of Korea
| | - Sang-wook Lee
- Department of Radiation Oncology, Asan Medical Center, Seoul, Republic of Korea
| | - In-Jeoung Baek
- Convergence Medicine Research Center, Asan Medical Center, Seoul, Republic of Korea
- Asan Institute for Lifesciences, Asan Medical Center, Seoul, Republic of Korea
| | - Hye-Young Song
- Asan Institute for Lifesciences, Asan Medical Center, Seoul, Republic of Korea
| | - Seon-Ju Jo
- Asan Institute for Lifesciences, Asan Medical Center, Seoul, Republic of Korea
| | - Je-Won Ryu
- Convergence Medicine Research Center, Asan Medical Center, Seoul, Republic of Korea
| | - Seung-Hee Ryu
- Asan Institute for Lifesciences, Asan Medical Center, Seoul, Republic of Korea
| | - Jin-Hee Seo
- Korea Radioisotope Center for Pharmaceuticals, Korea Institute of Radiological and Medical Sciences, Seoul, Republic of Korea
| | - Jong-Choon Kim
- College of Veterinary Medicine, Chonnam National University, Gwangju, Republic of Korea
| | - Seung-Ho Heo
- Convergence Medicine Research Center, Asan Medical Center, Seoul, Republic of Korea
- Asan Institute for Lifesciences, Asan Medical Center, Seoul, Republic of Korea
| |
Collapse
|
5
|
Li W, Xia C, Wang K, Xue L, Wang Y, Yang JY, Zhang M, Yin M, Ju C, Miao Z, Li Y, Zhao X, Yang Z, Tang R, Yang W. Technical considerations and strategies for generating and optimizing humanized mouse tumor models in immuno-oncology research. Int Immunopharmacol 2024; 139:112722. [PMID: 39033663 DOI: 10.1016/j.intimp.2024.112722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 07/15/2024] [Accepted: 07/16/2024] [Indexed: 07/23/2024]
Abstract
The field of cancer immunotherapy has experienced significant progress, resulting in the emergence of numerous biological drug candidates requiring in vivo efficacy testing and a better understanding of their mechanism of action (MOA). Humanized immune system (HIS) models are valuable tools in this regard. However, there is a lack of systematic guidance on HIS modeling. To address this issue, the present study aimed to establish and optimize a variety of HIS models for immune-oncology (IO) study, including genetically engineered mouse models and HIS models with human immune components reconstituted in severely immunocompromised mice. The efficacy and utility of these models were tested with several marketed or investigational IO drugs according to their MOA, followed by immunophenotypic analysis and efficacy evaluation. The results of the present study demonstrated that the HIS models responded to various IO drugs as expected and that each model had unique niches, utilities and limitations. Researchers should carefully choose the appropriate models based on the MOA and the targeted immune cell populations of the investigational drug. The present study provides valuable methodologies and actionable technical guidance on designing, generating or utilizing appropriate HIS models to address specific questions in translational IO.
Collapse
Affiliation(s)
- Wenjing Li
- State Key Laboratory of Neurology and Oncology Drug Development, Nanjing 210000, China; Simcere Zaiming Pharmaceutical Co, Ltd., Shanghai 200120, China
| | - Chunlei Xia
- State Key Laboratory of Neurology and Oncology Drug Development, Nanjing 210000, China; Simcere Zaiming Pharmaceutical Co, Ltd., Shanghai 200120, China
| | - Kun Wang
- State Key Laboratory of Neurology and Oncology Drug Development, Nanjing 210000, China; Simcere Zaiming Pharmaceutical Co, Ltd., Shanghai 200120, China
| | - Liting Xue
- State Key Laboratory of Neurology and Oncology Drug Development, Nanjing 210000, China; Simcere Zaiming Pharmaceutical Co, Ltd., Shanghai 200120, China
| | - Yan Wang
- State Key Laboratory of Neurology and Oncology Drug Development, Nanjing 210000, China; Simcere Zaiming Pharmaceutical Co, Ltd., Shanghai 200120, China
| | | | | | - Ming Yin
- Beijing Vitalstar Biotechnology Co., Ltd., Beijing 100000, China
| | - Cunxiang Ju
- Gempharmatech Co., Ltd., Nanjing 210000, China
| | - Zhenchuan Miao
- Beijing Vitalstar Biotechnology Co., Ltd., Beijing 100000, China
| | - Ying Li
- Gempharmatech Co., Ltd., Nanjing 210000, China
| | - Xiaofeng Zhao
- State Key Laboratory of Neurology and Oncology Drug Development, Nanjing 210000, China; Jiangsu Simcere Pharmaceutical Co, Ltd., Nanjing 210000, China
| | - Zhijian Yang
- ClinBridge Biotech Co., Ltd., Nanjing 210000, China
| | - Renhong Tang
- State Key Laboratory of Neurology and Oncology Drug Development, Nanjing 210000, China; Simcere Zaiming Pharmaceutical Co, Ltd., Shanghai 200120, China.
| | - WenQing Yang
- State Key Laboratory of Neurology and Oncology Drug Development, Nanjing 210000, China; Simcere Zaiming Pharmaceutical Co, Ltd., Shanghai 200120, China.
| |
Collapse
|
6
|
Meric-Bernstam F, Lloyd MW, Koc S, Evrard YA, McShane LM, Lewis MT, Evans KW, Li D, Rubinstein L, Welm A, Dean DA, Srivastava A, Grover JW, Ha MJ, Chen H, Huang X, Varadarajan K, Wang J, Roth JA, Welm B, Govinden R, Ding L, Kaochar S, Mitsiades N, Carvajal-Carmona L, Herylyn M, Davies MA, Shapiro GI, Fields R, Trevino JG, Harrell JC, Doroshow JH, Chuang JH, Moscow JA. Assessment of Patient-Derived Xenograft Growth and Antitumor Activity: The NCI PDXNet Consensus Recommendations. Mol Cancer Ther 2024; 23:924-938. [PMID: 38641411 PMCID: PMC11217730 DOI: 10.1158/1535-7163.mct-23-0471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 12/08/2023] [Accepted: 03/29/2024] [Indexed: 04/21/2024]
Abstract
Although patient-derived xenografts (PDX) are commonly used for preclinical modeling in cancer research, a standard approach to in vivo tumor growth analysis and assessment of antitumor activity is lacking, complicating the comparison of different studies and determination of whether a PDX experiment has produced evidence needed to consider a new therapy promising. We present consensus recommendations for assessment of PDX growth and antitumor activity, providing public access to a suite of tools for in vivo growth analyses. We expect that harmonizing PDX study design and analysis and assessing a suite of analytical tools will enhance information exchange and facilitate identification of promising novel therapies and biomarkers for guiding cancer therapy.
Collapse
Affiliation(s)
- Funda Meric-Bernstam
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | | | - Soner Koc
- Seven Bridges Genomics, Charlestown, Massachusetts.
| | - Yvonne A. Evrard
- Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland.
| | - Lisa M. McShane
- Biometric Research Program, DCTD, National Cancer Institute, National Institutes of Health, Bethesda, Maryland.
| | - Michael T. Lewis
- Departments of Molecular and Cellular Biology and Radiology, Lester and Sue Smith Breast Center, Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas.
| | - Kurt W. Evans
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Dali Li
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Lawrence Rubinstein
- Biometric Research Program, DCTD, National Cancer Institute, National Institutes of Health, Bethesda, Maryland.
| | - Alana Welm
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah.
| | | | - Anuj Srivastava
- The Jackson Laboratory for Genomic Medicine, Farmington, Connecticut.
| | | | - Min J. Ha
- Department of Biostatistics, Graduate School of Public Health, Yonsei University, Seoul, Republic of Korea.
| | - Huiqin Chen
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Xuelin Huang
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Kaushik Varadarajan
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Jing Wang
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Jack A. Roth
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Bryan Welm
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah.
| | - Ramaswamy Govinden
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri.
| | - Li Ding
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri.
| | - Salma Kaochar
- Department of Medicine, Baylor College of Medicine, Houston, Texas.
| | - Nicholas Mitsiades
- Department of Molecular Cellular Biology, Baylor College of Medicine, Houston, Texas.
| | - Luis Carvajal-Carmona
- Department of Biochemistry and Molecular Medicine, University of California, Davis, California.
| | | | - Michael A. Davies
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Geoffrey I. Shapiro
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.
| | - Ryan Fields
- Department of Surgery, Washington University School of Medicine, St. Louis, Missouri.
| | - Jose G. Trevino
- Department of Surgery, Virginia Commonwealth University, Richmond, Virginia.
| | - Joshua C. Harrell
- Department of Pathology, Virginia Commonwealth University, Richmond, Virginia.
| | | | - James H. Doroshow
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Bethesda, Maryland.
| | - Jeffrey H. Chuang
- The Jackson Laboratory for Genomic Medicine, Farmington, Connecticut.
| | - Jeffrey A. Moscow
- Investigational Drug Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland.
| |
Collapse
|
7
|
Willis E, Verrelle J, Banerjee E, Assenmacher CA, Tarrant JC, Skuli N, Jacobson ML, O’Rouke DM, Binder ZA, Radaelli E. Humanization with CD34-positive hematopoietic stem cells in NOG-EXL mice results in improved long-term survival and less severe myeloid cell hyperactivation phenotype relative to NSG-SGM3 mice. Vet Pathol 2024; 61:664-674. [PMID: 38197423 PMCID: PMC11264550 DOI: 10.1177/03009858231222216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2024]
Abstract
NSG-SGM3 and NOG-EXL mice combine severe immunodeficiency with transgenic expression of human myeloid stimulatory cytokines, resulting in marked expansion of myeloid populations upon humanization with CD34+ hematopoietic stem cells (HSCs). Humanized NSG-SGM3 mice typically develop a lethal macrophage activation syndrome and mast cell hyperplasia that limit their use in long-term studies (e.g., humanization followed by tumor xenotransplantation). It is currently unclear to what extent humanized NOG-EXL mice suffer from the same condition observed in humanized NSG-SGM3 mice. We compared the effects of human CD34+ HSC engraftment in these two strains in an orthotopic patient-derived glioblastoma model. NSG-SGM3 mice humanized in-house were compared to NOG-EXL mice humanized in-house and commercially available humanized NOG-EXL mice. Mice were euthanized at humane or study endpoints, and complete pathological assessments were performed. A semiquantitative multiparametric clinicopathological scoring system was developed to characterize chimeric myeloid cell hyperactivation (MCH) syndrome. NSG-SGM3 mice were euthanized at 16 weeks after humanization because of severe deterioration of clinical conditions. Humanized NOG-EXL mice survived to the study endpoint at 22 weeks after humanization and showed less-severe MCH phenotypes than NSG-SGM3 mice. Major differences included the lack of mast cell expansion and limited tissue/organ involvement in NOG-EXL mice compared to NSG-SGM3 mice. Engraftment of human lymphocytes, assessed by immunohistochemistry, was similar in the two strains. The longer survival and decreased MCH phenotype severity in NOG-EXL mice enabled their use in a tumor xenotransplantation study. The NOG-EXL model is better suited than the NSG-SGM3 model for immuno-oncology studies requiring long-term survival after humanization.
Collapse
Affiliation(s)
- Elinor Willis
- Comparative Pathology Core, Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA
| | - Jillian Verrelle
- Comparative Pathology Core, Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA
| | - Esha Banerjee
- Comparative Pathology Core, Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA
| | - Charles-Antoine Assenmacher
- Comparative Pathology Core, Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA
| | | | - Nicholas Skuli
- Stem Cell and Xenograft Core, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | | | - Donald M. O’Rouke
- Department of Neurosurgery, Perelman School of Medicine, GBM Translational Center of Excellence, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA
| | - Zev A. Binder
- Department of Neurosurgery, Perelman School of Medicine, GBM Translational Center of Excellence, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Enrico Radaelli
- Comparative Pathology Core, Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
8
|
Celhar T, Li X, Zhao Y, Tay HC, Lee A, Liew HH, Shepherdson EK, Rajarethinam R, Fan Y, Mak A, Chan JKY, Singhal A, Takahashi T. Fetal liver CD34 + contain human immune and endothelial progenitors and mediate solid tumor rejection in NOG mice. Stem Cell Res Ther 2024; 15:164. [PMID: 38853275 PMCID: PMC11163708 DOI: 10.1186/s13287-024-03756-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 05/07/2024] [Indexed: 06/11/2024] Open
Abstract
BACKGROUND Transplantation of CD34+ hematopoietic stem and progenitor cells (HSPC) into immunodeficient mice is an established method to generate humanized mice harbouring a human immune system. Different sources and methods for CD34+ isolation have been employed by various research groups, resulting in customized models that are difficult to compare. A more detailed characterization of CD34+ isolates is needed for a better understanding of engraftable hematopoietic and potentially non-hematopoietic cells. Here we have performed a direct comparison of CD34+ isolated from cord blood (CB-CD34+) or fetal liver (FL-CD34+ and FL-CD34+CD14-) and their engraftment into immunocompromised NOD/Shi-scid Il2rgnull (NOG) mice. METHODS NOG mice were transplanted with either CB-CD34+, FL-CD34+ or FL-CD34+CD14- to generate CB-NOG, FL-NOG and FL-CD14--NOG, respectively. After 15-20 weeks, the mice were sacrificed and human immune cell reconstitution was assessed in blood and several organs. Liver sections were pathologically assessed upon Haematoxylin and Eosin staining. To assess the capability of allogenic tumor rejection in CB- vs. FL-reconstituted mice, animals were subcutaneously engrafted with an HLA-mismatched melanoma cell line. Tumor growth was assessed by calliper measurements and a Luminex-based assay was used to compare the cytokine/chemokine profiles. RESULTS We show that CB-CD34+ are a uniform population of HSPC that reconstitute NOG mice more rapidly than FL-CD34+ due to faster B cell development. However, upon long-term engraftment, FL-NOG display increased numbers of neutrophils, dendritic cells and macrophages in multiple tissues. In addition to HSPC, FL-CD34+ isolates contain non-hematopoietic CD14+ endothelial cells that enhance the engraftment of the human immune system in FL-NOG mice. We demonstrate that these CD14+CD34+ cells are capable of reconstituting Factor VIII-producing liver sinusoidal endothelial cells (LSEC) in FL-NOG. However, CD14+CD34+ also contribute to hepatic sinusoidal dilatation and immune cell infiltration, which may culminate in a graft-versus-host disease (GVHD) pathology upon long-term engraftment. Finally, using an HLA-A mismatched CDX melanoma model, we show that FL-NOG, but not CB-NOG, can mount a graft-versus-tumor (GVT) response resulting in tumor rejection. CONCLUSION Our results highlight important phenotypical and functional differences between CB- and FL-NOG and reveal FL-NOG as a potential model to study hepatic sinusoidal dilatation and mechanisms of GVT.
Collapse
Affiliation(s)
- Teja Celhar
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, Immunos #04-06, Singapore, 138648, Republic of Singapore.
- Central Institute for Experimental Animals (CIEA), 3-25-12 Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa, 210-0821, Japan.
- A*STAR Infectious Diseases Labs (A*STAR ID Labs), Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, Immunos #05-13, Singapore, 138648, Republic of Singapore.
| | - Xinyi Li
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, Immunos #04-06, Singapore, 138648, Republic of Singapore
- Interdisciplinary Life Sciences, Purdue University, West Lafayette, IN, 47907, USA
| | - Yunqian Zhao
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, Immunos #04-06, Singapore, 138648, Republic of Singapore
| | - Hui Chien Tay
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, Immunos #04-06, Singapore, 138648, Republic of Singapore
| | - Andrea Lee
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, Immunos #04-06, Singapore, 138648, Republic of Singapore
- A*STAR Infectious Diseases Labs (A*STAR ID Labs), Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, Immunos #05-13, Singapore, 138648, Republic of Singapore
| | - Hui Hua Liew
- Department of Reproductive Medicine, KK Women's and Children's Hospital, Singapore, 229899, Republic of Singapore
| | - Edwin Kunxiang Shepherdson
- Department of Reproductive Medicine, KK Women's and Children's Hospital, Singapore, 229899, Republic of Singapore
| | - Ravisankar Rajarethinam
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Republic of Singapore
| | - Yiping Fan
- Department of Reproductive Medicine, KK Women's and Children's Hospital, Singapore, 229899, Republic of Singapore
- Obstetrics and Gynaecology Academic Clinical Programme, Duke-NUS Medical School, Singapore, 169857, Republic of Singapore
- Experimental Fetal Medicine Group, Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University Health System, Singapore, 117597, Republic of Singapore
| | - Anselm Mak
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Division of Rheumatology, University Medicine Cluster, National University Health System, Singapore, Republic of Singapore
| | - Jerry Kok Yen Chan
- Department of Reproductive Medicine, KK Women's and Children's Hospital, Singapore, 229899, Republic of Singapore
- Obstetrics and Gynaecology Academic Clinical Programme, Duke-NUS Medical School, Singapore, 169857, Republic of Singapore
- Experimental Fetal Medicine Group, Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University Health System, Singapore, 117597, Republic of Singapore
| | - Amit Singhal
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, Immunos #04-06, Singapore, 138648, Republic of Singapore
- A*STAR Infectious Diseases Labs (A*STAR ID Labs), Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, Immunos #05-13, Singapore, 138648, Republic of Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 636921, Republic of Singapore
| | - Takeshi Takahashi
- Central Institute for Experimental Animals (CIEA), 3-25-12 Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa, 210-0821, Japan
| |
Collapse
|
9
|
Xu Y, Shan W, Luo Q, Zhang M, Huo D, Chen Y, Li H, Ye Y, Yu X, Luo Y, Huang H. Establishment of a humanized mouse model using steady-state peripheral blood-derived hematopoietic stem and progenitor cells facilitates screening of cancer-targeted T-cell repertoires. CANCER INNOVATION 2024; 3:e118. [PMID: 38947755 PMCID: PMC11212321 DOI: 10.1002/cai2.118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 12/19/2023] [Accepted: 01/19/2024] [Indexed: 07/02/2024]
Abstract
Background Cancer-targeted T-cell receptor T (TCR-T) cells hold promise in treating cancers such as hematological malignancies and breast cancers. However, approaches to obtain cancer-reactive TCR-T cells have been unsuccessful. Methods Here, we developed a novel strategy to screen for cancer-targeted TCR-T cells using a special humanized mouse model with person-specific immune fingerprints. Rare steady-state circulating hematopoietic stem and progenitor cells were expanded via three-dimensional culture of steady-state peripheral blood mononuclear cells, and then the expanded cells were applied to establish humanized mice. The human immune system was evaluated according to the kinetics of dendritic cells, monocytes, T-cell subsets, and cytokines. To fully stimulate the immune response and to obtain B-cell precursor NAML-6- and triple-negative breast cancer MDA-MB-231-targeted TCR-T cells, we used the inactivated cells above to treat humanized mice twice a day every 7 days. Then, human T cells were processed for TCR β-chain (TRB) sequencing analysis. After the repertoires had been constructed, features such as the fraction, diversity, and immune signature were investigated. Results The results demonstrated an increase in diversity and clonality of T cells after treatment. The preferential usage and features of TRBV, TRBJ, and the V-J combination were also changed. The stress also induced highly clonal expansion. Tumor burden and survival analysis demonstrated that stress induction could significantly inhibit the growth of subsequently transfused live tumor cells and prolong the survival of the humanized mice. Conclusions We constructed a personalized humanized mouse model to screen cancer-targeted TCR-T pools. Our platform provides an effective source of cancer-targeted TCR-T cells and allows for the design of patient-specific engineered T cells. It therefore has the potential to greatly benefit cancer treatment.
Collapse
Affiliation(s)
- Yulin Xu
- Bone Marrow Transplantation Center, The First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
- Liangzhu LaboratoryZhejiang UniversityHangzhouChina
- Institute of HematologyZhejiang UniversityHangzhouChina
- Zhejiang Province Engineering Research Center for Stem Cell and Immunity TherapyHangzhouChina
- School of MedicineZhejiang UniversityHangzhouChina
| | - Wei Shan
- Bone Marrow Transplantation Center, The First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
- Liangzhu LaboratoryZhejiang UniversityHangzhouChina
- Institute of HematologyZhejiang UniversityHangzhouChina
- Zhejiang Province Engineering Research Center for Stem Cell and Immunity TherapyHangzhouChina
- School of MedicineZhejiang UniversityHangzhouChina
| | - Qian Luo
- Bone Marrow Transplantation Center, The First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
- Liangzhu LaboratoryZhejiang UniversityHangzhouChina
- Institute of HematologyZhejiang UniversityHangzhouChina
- Zhejiang Province Engineering Research Center for Stem Cell and Immunity TherapyHangzhouChina
- School of MedicineZhejiang UniversityHangzhouChina
| | - Meng Zhang
- Bone Marrow Transplantation Center, The First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
- Liangzhu LaboratoryZhejiang UniversityHangzhouChina
- Institute of HematologyZhejiang UniversityHangzhouChina
- Zhejiang Province Engineering Research Center for Stem Cell and Immunity TherapyHangzhouChina
- School of MedicineZhejiang UniversityHangzhouChina
| | - Dawei Huo
- Bone Marrow Transplantation Center, The First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
- Liangzhu LaboratoryZhejiang UniversityHangzhouChina
- Institute of HematologyZhejiang UniversityHangzhouChina
- Zhejiang Province Engineering Research Center for Stem Cell and Immunity TherapyHangzhouChina
- School of MedicineZhejiang UniversityHangzhouChina
| | - Yijin Chen
- Bone Marrow Transplantation Center, The First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
- Liangzhu LaboratoryZhejiang UniversityHangzhouChina
- Institute of HematologyZhejiang UniversityHangzhouChina
- Zhejiang Province Engineering Research Center for Stem Cell and Immunity TherapyHangzhouChina
- School of MedicineZhejiang UniversityHangzhouChina
| | - Honghu Li
- Bone Marrow Transplantation Center, The First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
- Liangzhu LaboratoryZhejiang UniversityHangzhouChina
- Institute of HematologyZhejiang UniversityHangzhouChina
- Zhejiang Province Engineering Research Center for Stem Cell and Immunity TherapyHangzhouChina
- School of MedicineZhejiang UniversityHangzhouChina
| | - Yishan Ye
- Bone Marrow Transplantation Center, The First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
- Liangzhu LaboratoryZhejiang UniversityHangzhouChina
- Institute of HematologyZhejiang UniversityHangzhouChina
- Zhejiang Province Engineering Research Center for Stem Cell and Immunity TherapyHangzhouChina
- School of MedicineZhejiang UniversityHangzhouChina
| | - Xiaohong Yu
- Liangzhu LaboratoryZhejiang UniversityHangzhouChina
- Institute of HematologyZhejiang UniversityHangzhouChina
- Zhejiang Province Engineering Research Center for Stem Cell and Immunity TherapyHangzhouChina
- School of MedicineZhejiang UniversityHangzhouChina
| | - Yi Luo
- Bone Marrow Transplantation Center, The First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
- Liangzhu LaboratoryZhejiang UniversityHangzhouChina
- Institute of HematologyZhejiang UniversityHangzhouChina
- Zhejiang Province Engineering Research Center for Stem Cell and Immunity TherapyHangzhouChina
- School of MedicineZhejiang UniversityHangzhouChina
| | - He Huang
- Bone Marrow Transplantation Center, The First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
- Liangzhu LaboratoryZhejiang UniversityHangzhouChina
- Institute of HematologyZhejiang UniversityHangzhouChina
- Zhejiang Province Engineering Research Center for Stem Cell and Immunity TherapyHangzhouChina
- School of MedicineZhejiang UniversityHangzhouChina
| |
Collapse
|
10
|
Tian J, Ashique AM, Weeks S, Lan T, Yang H, Chen HIH, Song C, Koyano K, Mondal K, Tsai D, Cheung I, Moshrefi M, Kekatpure A, Fan B, Li B, Qurashi S, Rocha L, Aguayo J, Rodgers C, Meza M, Heeke D, Medfisch SM, Chu C, Starck S, Basak NP, Sankaran S, Malhotra M, Crawley S, Tran TT, Duey DY, Ho C, Mikaelian I, Liu W, Rivera LB, Huang J, Paavola KJ, O'Hollaren K, Blum LK, Lin VY, Chen P, Iyer A, He S, Roda JM, Wang Y, Sissons J, Kutach AK, Kaplan DD, Stone GW. ILT2 and ILT4 Drive Myeloid Suppression via Both Overlapping and Distinct Mechanisms. Cancer Immunol Res 2024; 12:592-613. [PMID: 38393969 DOI: 10.1158/2326-6066.cir-23-0568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Revised: 10/28/2023] [Accepted: 02/20/2024] [Indexed: 02/25/2024]
Abstract
Solid tumors are dense three-dimensional (3D) multicellular structures that enable efficient receptor-ligand trans interactions via close cell-cell contact. Immunoglobulin-like transcript (ILT)2 and ILT4 are related immune-suppressive receptors that play a role in the inhibition of myeloid cells within the tumor microenvironment. The relative contribution of ILT2 and ILT4 to immune inhibition in the context of solid tumor tissue has not been fully explored. We present evidence that both ILT2 and ILT4 contribute to myeloid inhibition. We found that although ILT2 inhibits myeloid cell activation in the context of trans-engagement by MHC-I, ILT4 efficiently inhibits myeloid cells in the presence of either cis- or trans-engagement. In a 3D spheroid tumor model, dual ILT2/ILT4 blockade was required for the optimal activation of myeloid cells, including the secretion of CXCL9 and CCL5, upregulation of CD86 on dendritic cells, and downregulation of CD163 on macrophages. Humanized mouse tumor models showed increased immune activation and cytolytic T-cell activity with combined ILT2 and ILT4 blockade, including evidence of the generation of immune niches, which have been shown to correlate with clinical response to immune-checkpoint blockade. In a human tumor explant histoculture system, dual ILT2/ILT4 blockade increased CXCL9 secretion, downregulated CD163 expression, and increased the expression of M1 macrophage, IFNγ, and cytolytic T-cell gene signatures. Thus, we have revealed distinct contributions of ILT2 and ILT4 to myeloid cell biology and provide proof-of-concept data supporting the combined blockade of ILT2 and ILT4 to therapeutically induce optimal myeloid cell reprogramming in the tumor microenvironment.
Collapse
Affiliation(s)
- Jane Tian
- NGM Biopharmaceuticals, South San Francisco, California
| | | | - Sabrina Weeks
- NGM Biopharmaceuticals, South San Francisco, California
| | - Tian Lan
- NGM Biopharmaceuticals, South San Francisco, California
| | - Hong Yang
- NGM Biopharmaceuticals, South San Francisco, California
| | | | | | - Kikuye Koyano
- NGM Biopharmaceuticals, South San Francisco, California
| | | | - Daniel Tsai
- NGM Biopharmaceuticals, South San Francisco, California
| | - Isla Cheung
- NGM Biopharmaceuticals, South San Francisco, California
| | | | | | - Bin Fan
- NGM Biopharmaceuticals, South San Francisco, California
| | - Betty Li
- NGM Biopharmaceuticals, South San Francisco, California
| | - Samir Qurashi
- NGM Biopharmaceuticals, South San Francisco, California
| | - Lauren Rocha
- NGM Biopharmaceuticals, South San Francisco, California
| | | | - Col Rodgers
- NGM Biopharmaceuticals, South San Francisco, California
| | | | - Darren Heeke
- NGM Biopharmaceuticals, South San Francisco, California
| | | | - Chun Chu
- NGM Biopharmaceuticals, South San Francisco, California
| | | | | | | | | | | | | | - Dana Y Duey
- NGM Biopharmaceuticals, South San Francisco, California
| | - Carmence Ho
- NGM Biopharmaceuticals, South San Francisco, California
| | | | - Wenhui Liu
- NGM Biopharmaceuticals, South San Francisco, California
| | - Lee B Rivera
- NGM Biopharmaceuticals, South San Francisco, California
| | - Jiawei Huang
- NGM Biopharmaceuticals, South San Francisco, California
| | | | | | - Lisa K Blum
- NGM Biopharmaceuticals, South San Francisco, California
| | - Vicky Y Lin
- NGM Biopharmaceuticals, South San Francisco, California
| | - Peirong Chen
- NGM Biopharmaceuticals, South San Francisco, California
| | | | - Sisi He
- NGM Biopharmaceuticals, South San Francisco, California
| | - Julie M Roda
- NGM Biopharmaceuticals, South San Francisco, California
| | - Yan Wang
- NGM Biopharmaceuticals, South San Francisco, California
| | - James Sissons
- NGM Biopharmaceuticals, South San Francisco, California
| | - Alan K Kutach
- NGM Biopharmaceuticals, South San Francisco, California
| | | | | |
Collapse
|
11
|
Chen L, Chen Y, Ge L, Zhang Q, Meng J. Recent advances in patient-derived tumor organoids for reconstructing TME of head and neck cancer. J Oral Pathol Med 2024; 53:238-245. [PMID: 38561906 DOI: 10.1111/jop.13532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Revised: 03/13/2024] [Accepted: 03/17/2024] [Indexed: 04/04/2024]
Abstract
BACKGROUND The differences between existing preclinical models and the tumor microenvironment in vivo are one of the significant challenges hindering cancer therapy development. Patient-derived tumor organoids (PDTO) can highly retain tumor heterogeneity. Thus, it provides a more reliable platform for research in tumor biology, new drug screening, and precision medicine. METHODS We conducted a systematic review to summarise the characteristics of the existing preclinical models, the advantages of patient-derived tumor organoids in reconstructing the tumor microenvironment, and the latest research progress. Moreover, this study deciphers organoid culture technology in the clinical precision treatment of head and neck cancer to achieve better transformation. Studies were identified through a comprehensive search of Ovid MEDLINE (Wolters Kluwer), PubMed (National Library of Medicine), web of Science (Thomson Reuters) and, Scopus (Elsevier) databases, without publication date or language restrictions. RESULTS In tumor development, the interaction between cellular and non-cellular components in the tumor microenvironment (TME) has a crucial role. Co-culture, Air-liquid interface culture, microfluidics, and decellularized matrix have depicted great potential in reconstructing the tumor microenvironment and simulating tumor genesis, development, and metastasis. CONCLUSION An accurate determination of stromal cells, immune cells, and extracellular matrix can be achieved by reconstructing the head and neck cancer tumor microenvironment using the PDTO model. Moreover, the interaction between head and neck cancer cells can also play an essential role in implementing the individualized precision treatment of head and neck cancer.
Collapse
Affiliation(s)
- Lin Chen
- Department of Stomatology, Xuzhou Central Hospital, Xuzhou, Jiangsu, China
- School of Stomatology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yinyu Chen
- Department of Stomatology, Xuzhou Central Hospital, Xuzhou, Jiangsu, China
- School of Stomatology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Liangyu Ge
- Department of Stomatology, Xuzhou Central Hospital, Xuzhou, Jiangsu, China
| | - Qian Zhang
- Department of Stomatology, Xuzhou Central Hospital, Xuzhou, Jiangsu, China
| | - Jian Meng
- Department of Stomatology, Xuzhou Central Hospital, Xuzhou, Jiangsu, China
- School of Stomatology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| |
Collapse
|
12
|
Zhou X, Geyer FK, Happel D, Takimoto J, Kolmar H, Rabinovich B. Using protein geometry to optimize cytotoxicity and the cytokine window of a ROR1 specific T cell engager. Front Immunol 2024; 15:1323049. [PMID: 38455046 PMCID: PMC10917902 DOI: 10.3389/fimmu.2024.1323049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 02/05/2024] [Indexed: 03/09/2024] Open
Abstract
T cell engaging bispecific antibodies have shown clinical proof of concept for hematologic malignancies. Still, cytokine release syndrome, neurotoxicity, and on-target-off-tumor toxicity, especially in the solid tumor setting, represent major obstacles. Second generation TCEs have been described that decouple cytotoxicity from cytokine release by reducing the apparent binding affinity for CD3 and/or the TAA but the results of such engineering have generally led only to reduced maximum induction of cytokine release and often at the expense of maximum cytotoxicity. Using ROR1 as our model TAA and highly modular camelid nanobodies, we describe the engineering of a next generation decoupled TCE that incorporates a "cytokine window" defined as a dose range in which maximal killing is reached but cytokine release may be modulated from very low for safety to nearly that induced by first generation TCEs. This latter attribute supports pro-inflammatory anti-tumor activity including bystander killing and can potentially be used by clinicians to safely titrate patient dose to that which mediates maximum efficacy that is postulated as greater than that possible using standard second generation approaches. We used a combined method of optimizing TCE mediated synaptic distance and apparent affinity tuning of the TAA binding arms to generate a relatively long but persistent synapse that supports a wide cytokine window, potent killing and a reduced propensity towards immune exhaustion. Importantly, this next generation TCE induced significant tumor growth inhibition in vivo but unlike a first-generation non-decoupled benchmark TCE that induced lethal CRS, no signs of adverse events were observed.
Collapse
Affiliation(s)
- Xueyuan Zhou
- Drug Discovery and Development, Fuse Biotherapeutics, Woburn, MA, United States
| | - Felix Klaus Geyer
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Darmstadt, Germany
| | - Dominic Happel
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Darmstadt, Germany
| | - Jeffrey Takimoto
- Drug Discovery and Development, Fuse Biotherapeutics, Woburn, MA, United States
| | - Harald Kolmar
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Darmstadt, Germany
- Centre for Synthetic Biology, Technical University of Darmstadt, Darmstadt, Germany
| | - Brian Rabinovich
- Drug Discovery and Development, Fuse Biotherapeutics, Woburn, MA, United States
| |
Collapse
|
13
|
Huang T, Leung B, Huang Y, Price L, Gui J, Lau BW. A murine model to evaluate immunotherapy effectiveness for human Fanconi anemia-mutated acute myeloid leukemia. PLoS One 2024; 19:e0292375. [PMID: 38289944 PMCID: PMC10826936 DOI: 10.1371/journal.pone.0292375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 09/19/2023] [Indexed: 02/01/2024] Open
Abstract
Fanconi anemia (FA)-mutated acute myeloid leukemia (AML) is a secondary AML with very poor prognosis and limited therapeutic options due to increased sensitivity to DNA-damaging agents. PD-1 immune checkpoint inhibitors upregulate T-cell killing of cancer cells and is a class of promising treatment for FA-AML. Here, we developed a novel FA-AML murine model that allows the study of human AML with a humanized immune system in order to investigate immunotherapeutic treatments in vivo. FA-AML1 cells and non-FA-mutated Kasumi-1 cells were injected into 8-10 week old NSG mice. Once leukemic engraftment was confirmed by HLA-DR expression in the peripheral blood, human peripheral blood mononuclear cells (hPBMCs) were injected into the mice. One week post-hPBMCs injection, Nivolumab (PD-1 inhibitor) or PBS vehicle control was administered to the mice bi-weekly. In our Nivolumab treated mice, FA-AML1, but not Kasumi-1-engrafted mice, had significantly prolonged overall survival. Both FA-AML1 and Kasumi-1 engrafted mice had decreased spleen weights. Higher leukemic infiltration into vital organs was observed in FA-AML1 engrafted mice compared to Kasumi-1 engrafted mice. In conclusion, our novel humanized murine model of FA-mutated AML is an attractive tool for supporting further studies and clinical trials using PD-1 inhibitors to treat FA-mutated AML.
Collapse
Affiliation(s)
- Tingting Huang
- Dartmouth Health Cancer Center, Department of Pediatrics, Geisel School of Medicine at Dartmouth College, Lebanon, NH, United States of America
| | - Bernice Leung
- Dartmouth Health Cancer Center, Department of Pediatrics, Geisel School of Medicine at Dartmouth College, Lebanon, NH, United States of America
| | - Yuyang Huang
- Dartmouth Health Cancer Center, Department of Pediatrics, Geisel School of Medicine at Dartmouth College, Lebanon, NH, United States of America
| | - Laura Price
- Dartmouth Health Cancer Center, Department of Pediatrics, Geisel School of Medicine at Dartmouth College, Lebanon, NH, United States of America
| | - Jiang Gui
- Department of Biomedical Data Science, Geisel School of Medicine, Lebanon, NH, United States of America
| | - Bonnie W. Lau
- Dartmouth Health Cancer Center, Department of Pediatrics, Geisel School of Medicine at Dartmouth College, Lebanon, NH, United States of America
| |
Collapse
|
14
|
Jia B, Zhao C, Minagawa K, Shike H, Claxton DF, Ehmann WC, Rybka WB, Mineishi S, Wang M, Schell TD, Prabhu KS, Paulson RF, Zhang Y, Shultz LD, Zheng H. Acute Myeloid Leukemia Causes T Cell Exhaustion and Depletion in a Humanized Graft-versus-Leukemia Model. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 211:1426-1437. [PMID: 37712758 DOI: 10.4049/jimmunol.2300111] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 08/21/2023] [Indexed: 09/16/2023]
Abstract
Allogeneic hematopoietic stem cell transplantation (alloSCT) is, in many clinical settings, the only curative treatment for acute myeloid leukemia (AML). The clinical benefit of alloSCT greatly relies on the graft-versus-leukemia (GVL) effect. However, AML relapse remains the top cause of posttransplant death; this highlights the urgent need to enhance GVL. Studies of human GVL have been hindered by the lack of optimal clinically relevant models. In this article, we report, the successful establishment of a novel (to our knowledge) humanized GVL model system by transplanting clinically paired donor PBMCs and patient AML into MHC class I/II knockout NSG mice. We observed significantly reduced leukemia growth in humanized mice compared with mice that received AML alone, demonstrating a functional GVL effect. Using this model system, we studied human GVL responses against human AML cells in vivo and discovered that AML induced T cell depletion, likely because of increased T cell apoptosis. In addition, AML caused T cell exhaustion manifested by upregulation of inhibitory receptors, increased expression of exhaustion-related transcription factors, and decreased T cell function. Importantly, combined blockade of human T cell-inhibitory pathways effectively reduced leukemia burden and reinvigorated CD8 T cell function in this model system. These data, generated in a highly clinically relevant humanized GVL model, not only demonstrate AML-induced inhibition of alloreactive T cells but also identify promising therapeutic strategies targeting T cell depletion and exhaustion for overcoming GVL failure and treating AML relapse after alloSCT.
Collapse
Affiliation(s)
- Bei Jia
- Penn State Cancer Institute, Penn State University College of Medicine, Hershey, PA
| | - Chenchen Zhao
- Penn State Cancer Institute, Penn State University College of Medicine, Hershey, PA
| | - Kentaro Minagawa
- Penn State Cancer Institute, Penn State University College of Medicine, Hershey, PA
| | - Hiroko Shike
- Department of Pathology, Penn State University College of Medicine, Hershey, PA
| | - David F Claxton
- Penn State Cancer Institute, Penn State University College of Medicine, Hershey, PA
| | - W Christopher Ehmann
- Penn State Cancer Institute, Penn State University College of Medicine, Hershey, PA
| | - Witold B Rybka
- Penn State Cancer Institute, Penn State University College of Medicine, Hershey, PA
| | - Shin Mineishi
- Penn State Cancer Institute, Penn State University College of Medicine, Hershey, PA
| | - Ming Wang
- Department of Population and Quantitative Health Sciences, School of Medicine, Case Western Reserve University, Cleveland, OH
| | - Todd D Schell
- Penn State Cancer Institute, Penn State University College of Medicine, Hershey, PA
- Department of Microbiology and Immunology, Penn State University College of Medicine, Hershey, PA
| | - K Sandeep Prabhu
- Department of Veterinary and Biomedical Sciences, Penn State University, University Park, PA
| | - Robert F Paulson
- Department of Veterinary and Biomedical Sciences, Penn State University, University Park, PA
| | - Yi Zhang
- Center for Discovery and Innovation, Hackensack Meridian Health, Edison, NJ
| | - Leonard D Shultz
- Department of Immunology, The Jackson Laboratory, Bar Harbor, ME
| | - Hong Zheng
- Penn State Cancer Institute, Penn State University College of Medicine, Hershey, PA
- Department of Microbiology and Immunology, Penn State University College of Medicine, Hershey, PA
| |
Collapse
|
15
|
Chan WK, Williams J, Sorathia K, Pray B, Abusaleh K, Bian Z, Sharma A, Hout I, Nishat S, Hanel W, Sloan SL, Yasin A, Denlinger N, Zhang X, Muthusamy N, Vasu S, de Lima M, Yang Y, Baiocchi R, Alinari L. A novel CAR-T cell product targeting CD74 is an effective therapeutic approach in preclinical mantle cell lymphoma models. Exp Hematol Oncol 2023; 12:79. [PMID: 37740214 PMCID: PMC10517521 DOI: 10.1186/s40164-023-00437-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 08/18/2023] [Indexed: 09/24/2023] Open
Abstract
BACKGROUND Mantle cell lymphoma (MCL) is a rare B-cell non-Hodgkin lymphoma subtype which remains incurable despite multimodal approach including chemoimmunotherapy followed by stem cell transplant, targeted approaches such as the BTK inhibitor ibrutinib, and CD19 chimeric antigen receptor (CAR) T cells. CD74 is a nonpolymorphic type II integral membrane glycoprotein identified as an MHC class II chaperone and a receptor for macrophage migration inhibitory factor. Our group previously reported on CD74's abundant expression in MCL and its ability to increase via pharmacological inhibition of autophagosomal degradation. Milatuzumab, a fully humanized anti-CD74 monoclonal antibody, demonstrated significant activity in preclinical lymphoma models but failed to provide meaningful benefits in clinical trials mainly due to its short half-life. We hypothesized that targeting CD74 using a CAR-T cell would provide potent and durable anti-MCL activity. METHODS We engineered a second generation anti-CD74 CAR with 4-1BB and CD3ζ signaling domains (74bbz). Through in silico and rational mutagenesis on the scFV domain, the 74bbz CAR was functionally optimized for superior antigen binding affinity, proliferative signaling, and cytotoxic activity against MCL cells in vitro and in vivo. RESULTS Functionally optimized 74bbz CAR-T cells (clone 42105) induced significant killing of MCL cell lines, and primary MCL patient samples including one relapse after commercial CD19 CAR-T cell therapy with direct correlation between antigen density and cytotoxicity. It significantly prolonged the survival of an animal model established in NOD-SCIDγc-/- (NSG) mice engrafted with a human MCL cell line Mino subcutaneously compared to controls. Finally, while CD74 is also expressed on normal immune cell subsets, treatment with 74bbz CAR-T cells resulted in minimal cytotoxicity against these cells both in vitro and in vivo. CONCLUSIONS Targeting CD74 with 74bbz CAR-T cells represents a new cell therapy to provide a potent and durable and anti-MCL activity.
Collapse
Affiliation(s)
- Wing Keung Chan
- Department of Internal Medicine, Division of Hematology, College of Medicine, The Ohio State University, 400 W. 12th Ave, 481D Wiseman Hall, Columbus, OH, 43210, USA
| | - Jessica Williams
- Department of Internal Medicine, Division of Hematology, College of Medicine, The Ohio State University, 400 W. 12th Ave, 481D Wiseman Hall, Columbus, OH, 43210, USA
| | - Kinnari Sorathia
- Department of Internal Medicine, Division of Hematology, College of Medicine, The Ohio State University, 400 W. 12th Ave, 481D Wiseman Hall, Columbus, OH, 43210, USA
| | - Betsy Pray
- Department of Internal Medicine, Division of Hematology, College of Medicine, The Ohio State University, 400 W. 12th Ave, 481D Wiseman Hall, Columbus, OH, 43210, USA
| | - Kaled Abusaleh
- Department of Internal Medicine, Division of Hematology, College of Medicine, The Ohio State University, 400 W. 12th Ave, 481D Wiseman Hall, Columbus, OH, 43210, USA
| | - Zehua Bian
- Department of Internal Medicine, Division of Hematology, College of Medicine, The Ohio State University, 400 W. 12th Ave, 481D Wiseman Hall, Columbus, OH, 43210, USA
| | - Archisha Sharma
- Department of Internal Medicine, Division of Hematology, College of Medicine, The Ohio State University, 400 W. 12th Ave, 481D Wiseman Hall, Columbus, OH, 43210, USA
| | - Ian Hout
- Department of Internal Medicine, Division of Hematology, College of Medicine, The Ohio State University, 400 W. 12th Ave, 481D Wiseman Hall, Columbus, OH, 43210, USA
| | - Shamama Nishat
- Department of Internal Medicine, Division of Hematology, College of Medicine, The Ohio State University, 400 W. 12th Ave, 481D Wiseman Hall, Columbus, OH, 43210, USA
| | - Walter Hanel
- Department of Internal Medicine, Division of Hematology, College of Medicine, The Ohio State University, 400 W. 12th Ave, 481D Wiseman Hall, Columbus, OH, 43210, USA
| | - Shelby L Sloan
- Department of Internal Medicine, Division of Hematology, College of Medicine, The Ohio State University, 400 W. 12th Ave, 481D Wiseman Hall, Columbus, OH, 43210, USA
| | - Aneeq Yasin
- Department of Internal Medicine, Division of Hematology, College of Medicine, The Ohio State University, 400 W. 12th Ave, 481D Wiseman Hall, Columbus, OH, 43210, USA
| | - Nathan Denlinger
- Department of Internal Medicine, Division of Hematology, College of Medicine, The Ohio State University, 400 W. 12th Ave, 481D Wiseman Hall, Columbus, OH, 43210, USA
| | - Xiaoli Zhang
- Department of Biomedical Informatics/Center for Biostatistics, The Ohio State University, Columbus, OH, 43210, USA
| | - Natarajan Muthusamy
- Department of Internal Medicine, Division of Hematology, College of Medicine, The Ohio State University, 400 W. 12th Ave, 481D Wiseman Hall, Columbus, OH, 43210, USA
| | - Sumithira Vasu
- Department of Internal Medicine, Division of Hematology, College of Medicine, The Ohio State University, 400 W. 12th Ave, 481D Wiseman Hall, Columbus, OH, 43210, USA
| | - Marcos de Lima
- Department of Internal Medicine, Division of Hematology, College of Medicine, The Ohio State University, 400 W. 12th Ave, 481D Wiseman Hall, Columbus, OH, 43210, USA
| | - Yiping Yang
- Department of Internal Medicine, Division of Hematology, College of Medicine, The Ohio State University, 400 W. 12th Ave, 481D Wiseman Hall, Columbus, OH, 43210, USA
| | - Robert Baiocchi
- Department of Internal Medicine, Division of Hematology, College of Medicine, The Ohio State University, 400 W. 12th Ave, 481D Wiseman Hall, Columbus, OH, 43210, USA
| | - Lapo Alinari
- Department of Internal Medicine, Division of Hematology, College of Medicine, The Ohio State University, 400 W. 12th Ave, 481D Wiseman Hall, Columbus, OH, 43210, USA.
| |
Collapse
|
16
|
Baietti MF, Leucci E. Humanized mouse models for anti-cancer therapy. Methods Cell Biol 2023; 183:317-333. [PMID: 38548416 DOI: 10.1016/bs.mcb.2023.06.002] [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] [Indexed: 04/02/2024]
Abstract
Patient-derived xenograft (PDX) models are the golden standard for preclinical oncology as they can recapitulate the genotypic and phenotypic complexity of human tumors, thus enabling the development of effective therapeutic strategies. PDX models are typically established in immunocompromised animals that allow efficient growth of the xenografted tumor. Given the recent success of immune therapies in different tumors however, the establishment of humanized PDX models is critical to evaluate immune oncology drugs and/or combinations thereof. Here, we describe the detailed methods to obtain humanized PDX models for anti-cancer therapy testing.
Collapse
Affiliation(s)
- Maria Francesca Baietti
- TRACE PDX Platform, LKI Leuven Cancer Institute, Leuven, Belgium; Laboratory of RNA Cancer Biology, Department of Oncology, LKI Leuven Cancer Institute, Leuven, Belgium
| | - Eleonora Leucci
- TRACE PDX Platform, LKI Leuven Cancer Institute, Leuven, Belgium; Laboratory of RNA Cancer Biology, Department of Oncology, LKI Leuven Cancer Institute, Leuven, Belgium.
| |
Collapse
|
17
|
Yan C, Nebhan CA, Saleh N, Shattuck-Brandt R, Chen SC, Ayers GD, Weiss V, Richmond A, Vilgelm AE. Generation of Orthotopic Patient-Derived Xenografts in Humanized Mice for Evaluation of Emerging Targeted Therapies and Immunotherapy Combinations for Melanoma. Cancers (Basel) 2023; 15:3695. [PMID: 37509357 PMCID: PMC10377652 DOI: 10.3390/cancers15143695] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 07/07/2023] [Accepted: 07/13/2023] [Indexed: 07/30/2023] Open
Abstract
Current methodologies for developing PDX in humanized mice in preclinical trials with immune-based therapies are limited by GVHD. Here, we compared two approaches for establishing PDX tumors in humanized mice: (1) PDX are first established in immune-deficient mice; or (2) PDX are initially established in humanized mice; then established PDX are transplanted to a larger cohort of humanized mice for preclinical trials. With the first approach, there was rapid wasting of PDX-bearing humanized mice with high levels of activated T cells in the circulation and organs, indicating immune-mediated toxicity. In contrast, with the second approach, toxicity was less of an issue and long-term human melanoma tumor growth and maintenance of human chimerism was achieved. Preclinical trials from the second approach revealed that rigosertib, but not anti-PD-1, increased CD8/CD4 T cell ratios in spleen and blood and inhibited PDX tumor growth. Resistance to anti-PD-1 was associated with PDX tumors established from tumors with limited CD8+ T cell content. Our findings suggest that it is essential to carefully manage immune editing by first establishing PDX tumors in humanized mice before expanding PDX tumors into a larger cohort of humanized mice to evaluate therapy response.
Collapse
Affiliation(s)
- Chi Yan
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA; (C.Y.); (N.S.); (R.S.-B.)
- Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, TN 37232, USA;
| | - Caroline A. Nebhan
- Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, TN 37232, USA;
- Division of Hematology & Oncology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Nabil Saleh
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA; (C.Y.); (N.S.); (R.S.-B.)
| | - Rebecca Shattuck-Brandt
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA; (C.Y.); (N.S.); (R.S.-B.)
- Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, TN 37232, USA;
| | - Sheau-Chiann Chen
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN 37232, USA; (S.-C.C.); (G.D.A.)
| | - Gregory D. Ayers
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN 37232, USA; (S.-C.C.); (G.D.A.)
| | - Vivian Weiss
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA;
| | - Ann Richmond
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA; (C.Y.); (N.S.); (R.S.-B.)
- Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, TN 37232, USA;
| | - Anna E. Vilgelm
- Department of Pathology, Ohio State University, Columbus, OH 43210, USA
- Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center—Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, Columbus, OH 43210, USA
| |
Collapse
|
18
|
Foss A, Pathania M. Pediatric Glioma Models Provide Insights into Tumor Development and Future Therapeutic Strategies. Dev Neurosci 2023; 46:22-43. [PMID: 37231843 DOI: 10.1159/000531040] [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: 02/20/2023] [Accepted: 05/09/2023] [Indexed: 05/27/2023] Open
Abstract
In depth study of pediatric gliomas has been hampered due to difficulties in accessing patient tissue and a lack of clinically representative tumor models. Over the last decade, however, profiling of carefully curated cohorts of pediatric tumors has identified genetic drivers that molecularly segregate pediatric gliomas from adult gliomas. This information has inspired the development of a new set of powerful in vitro and in vivo tumor models that can aid in identifying pediatric-specific oncogenic mechanisms and tumor microenvironment interactions. Single-cell analyses of both human tumors and these newly developed models have revealed that pediatric gliomas arise from spatiotemporally discrete neural progenitor populations in which developmental programs have become dysregulated. Pediatric high-grade gliomas also harbor distinct sets of co-segregating genetic and epigenetic alterations, often accompanied by unique features within the tumor microenvironment. The development of these novel tools and data resources has led to insights into the biology and heterogeneity of these tumors, including identification of distinctive sets of driver mutations, developmentally restricted cells of origin, recognizable patterns of tumor progression, characteristic immune environments, and tumor hijacking of normal microenvironmental and neural programs. As concerted efforts have broadened our understanding of these tumors, new therapeutic vulnerabilities have been identified, and for the first time, promising new strategies are being evaluated in the preclinical and clinical settings. Even so, dedicated and sustained collaborative efforts are necessary to refine our knowledge and bring these new strategies into general clinical use. In this review, we will discuss the range of currently available glioma models, the way in which they have each contributed to recent developments in the field, their benefits and drawbacks for addressing specific research questions, and their future utility in advancing biological understanding and treatment of pediatric glioma.
Collapse
Affiliation(s)
- Amelia Foss
- Department of Oncology and the Milner Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
- CRUK Children's Brain Tumour Centre of Excellence, University of Cambridge, Cambridge, UK
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Manav Pathania
- Department of Oncology and the Milner Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
- CRUK Children's Brain Tumour Centre of Excellence, University of Cambridge, Cambridge, UK
| |
Collapse
|
19
|
Suvilesh KN, Manjunath Y, Pantel K, Kaifi JT. Preclinical models to study patient-derived circulating tumor cells and metastasis. Trends Cancer 2023; 9:355-371. [PMID: 36759267 DOI: 10.1016/j.trecan.2023.01.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 01/17/2023] [Accepted: 01/18/2023] [Indexed: 02/10/2023]
Abstract
Circulating tumor cells (CTCs) that are detached from the tumor can be precursors of metastasis. The majority of studies focus on enumeration of CTCs from patient blood to predict recurrence and therapy outcomes. Very few studies have managed to expand CTCs to investigate their functional dynamics with respect to genetic changes, tumorigenic potential, and response to drug treatment. A growing amount of evidence based on successful CTC expansion has revealed novel therapeutic targets that are associated with the process of metastasis. In this review, we summarize the successes, challenges, and limitations that collectively contribute to the better understanding of metastasis using patient-derived CTCs as blood-borne seeds of metastasis. The roadblocks and future avenues to move CTC-based scientific discoveries forward are also discussed.
Collapse
Affiliation(s)
- Kanve N Suvilesh
- Hugh E. Stephenson Jr., MD, Department of Surgery, Ellis Fischel Cancer Center, University of Missouri, Columbia, MO, USA.
| | - Yariswamy Manjunath
- Hugh E. Stephenson Jr., MD, Department of Surgery, Ellis Fischel Cancer Center, University of Missouri, Columbia, MO, USA; Harry S. Truman Memorial Veterans' Hospital, Columbia, MO, USA
| | - Klaus Pantel
- Institute for Tumor Biology, University of Hamburg, Hamburg, Germany
| | - Jussuf T Kaifi
- Hugh E. Stephenson Jr., MD, Department of Surgery, Ellis Fischel Cancer Center, University of Missouri, Columbia, MO, USA; Harry S. Truman Memorial Veterans' Hospital, Columbia, MO, USA; Siteman Cancer Center, St. Louis, MO, USA.
| |
Collapse
|
20
|
Chuprin J, Buettner H, Seedhom MO, Greiner DL, Keck JG, Ishikawa F, Shultz LD, Brehm MA. Humanized mouse models for immuno-oncology research. Nat Rev Clin Oncol 2023; 20:192-206. [PMID: 36635480 PMCID: PMC10593256 DOI: 10.1038/s41571-022-00721-2] [Citation(s) in RCA: 96] [Impact Index Per Article: 96.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/12/2022] [Indexed: 01/14/2023]
Abstract
Immunotherapy has emerged as a promising treatment paradigm for many malignancies and is transforming the drug development landscape. Although immunotherapeutic agents have demonstrated clinical efficacy, they are associated with variable clinical responses, and substantial gaps remain in our understanding of their mechanisms of action and specific biomarkers of response. Currently, the number of preclinical models that faithfully recapitulate interactions between the human immune system and tumours and enable evaluation of human-specific immunotherapies in vivo is limited. Humanized mice, a term that refers to immunodeficient mice co-engrafted with human tumours and immune components, provide several advantages for immuno-oncology research. In this Review, we discuss the benefits and challenges of the currently available humanized mice, including specific interactions between engrafted human tumours and immune components, the development and survival of human innate immune populations in these mice, and approaches to study mice engrafted with matched patient tumours and immune cells. We highlight the latest advances in the generation of humanized mouse models, with the aim of providing a guide for their application to immuno-oncology studies with potential for clinical translation.
Collapse
Affiliation(s)
- Jane Chuprin
- Program in Molecular Medicine, The University of Massachusetts Chan Medical School, Worcester, MA, USA
- Department of Molecular, Cell and Cancer Biology, The University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Hannah Buettner
- Program in Molecular Medicine, The University of Massachusetts Chan Medical School, Worcester, MA, USA
- Department of Surgery, The University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Mina O Seedhom
- Program in Molecular Medicine, The University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Dale L Greiner
- Program in Molecular Medicine, The University of Massachusetts Chan Medical School, Worcester, MA, USA
| | | | | | | | - Michael A Brehm
- Program in Molecular Medicine, The University of Massachusetts Chan Medical School, Worcester, MA, USA.
| |
Collapse
|
21
|
Han Y, Fan X, Fan L, Wu Y, Zhou Z, Wang G, Guo L, Gao W, Chen Y, Gao Q. Liujunzi decoction exerts potent antitumor activity in oesophageal squamous cell carcinoma by inhibiting miR-34a/STAT3/IL-6R feedback loop, and modifies antitumor immunity. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 111:154672. [PMID: 36701994 DOI: 10.1016/j.phymed.2023.154672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 12/30/2022] [Accepted: 01/12/2023] [Indexed: 06/17/2023]
Abstract
BACKGROUND Liujunzi decoction (LJZD), a traditional herbal formula and one of the most commonly used adjuvant medications for the treatment of oesophageal squamous cell carcinoma (ESCC), exerts good antitumor and immunomodulatory activity. However, its specific mechanism of action remains largely unclear. PURPOSE In order to examine the potential primary and adjuvant antitumor mechanisms of LJZD, both in vitro and in vivo. METHODS IL-6 and miR-34a inhibitors were used to activate the miR-34a/STAT3/IL-6R feedback loop to observe the effects of LJZD. A humanised mouse model with a functional human immune system was constructed to evaluate the antitumor efficacy of LJZD in vivo on xenograft tumours, which was compared to that of the positive control drug anti-PD-1 monoclonal antibodies (mAb). Finally, a co-culture system of peripheral blood mononuclear and tumour cells in vitro was used to analyse the cytotoxic activity of LJZD on T cells. RESULTS LJZD significantly interfered with IL-6-induced activation of the miR-34a/STAT3/IL-6R feedback loop in ESCC by restoring the expression of the tumour suppressor miR-34a, and inhibited the proliferation of EC109 oesophageal cancer cells in a dose-dependant manner. Furthermore, LJZD effectively suppressed oesophageal tumour growth in vivo and alleviated organ injury and visceral index. Furthermore, LJZD boosted antitumor immunity by increasing IFN-γ expression and CD8+tumour-infiltrating lymphocytes (TILs) infiltration in the peripheral blood and tumour tissues, respectively, which may be related to a decrease in PD-1, but not PD-L1 expression. Finally, we confirmed that LJZD strengthens the killing ability of T cells by suppressing PD-1 expression in a co-culture system in vitro. CONCLUSION LJZD exerts excellent antitumor effect by interfering with the miR-34a/STAT3/IL-6R feedback loop and augmenting antitumor immune responses. Which provides new insights into mechanisms for LJZD and sheds light on the multifaceted role of phytomedicine in cancer.
Collapse
Affiliation(s)
- Yicun Han
- Henan International Joint Laboratory of TCM Syndrome and Prescription in Signaling, Henan University of Chinese Medicine, Zhengzhou, Henan Province 450046, China; Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan Province 450046, China; The Tumor Hospital Affiliated to Zhengzhou University, Zhengzhou, Henan Province 450008, China
| | - Xiuqi Fan
- Henan International Joint Laboratory of TCM Syndrome and Prescription in Signaling, Henan University of Chinese Medicine, Zhengzhou, Henan Province 450046, China; Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan Province 450046, China; The Tumor Hospital Affiliated to Zhengzhou University, Zhengzhou, Henan Province 450008, China
| | - Liyan Fan
- Henan International Joint Laboratory of TCM Syndrome and Prescription in Signaling, Henan University of Chinese Medicine, Zhengzhou, Henan Province 450046, China; Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan Province 450046, China
| | - Yaosong Wu
- Henan International Joint Laboratory of TCM Syndrome and Prescription in Signaling, Henan University of Chinese Medicine, Zhengzhou, Henan Province 450046, China; Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan Province 450046, China
| | - Zhexu Zhou
- Henan International Joint Laboratory of TCM Syndrome and Prescription in Signaling, Henan University of Chinese Medicine, Zhengzhou, Henan Province 450046, China; Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan Province 450046, China
| | - Ge Wang
- The Tumor Hospital Affiliated to Zhengzhou University, Zhengzhou, Henan Province 450008, China
| | - Lanwei Guo
- The Tumor Hospital Affiliated to Zhengzhou University, Zhengzhou, Henan Province 450008, China
| | - Wendong Gao
- Henan International Joint Laboratory of TCM Syndrome and Prescription in Signaling, Henan University of Chinese Medicine, Zhengzhou, Henan Province 450046, China; Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan Province 450046, China
| | - Yulong Chen
- Henan International Joint Laboratory of TCM Syndrome and Prescription in Signaling, Henan University of Chinese Medicine, Zhengzhou, Henan Province 450046, China; Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan Province 450046, China.
| | - Qilong Gao
- The Tumor Hospital Affiliated to Zhengzhou University, Zhengzhou, Henan Province 450008, China.
| |
Collapse
|
22
|
TGF-β2 antisense oligonucleotide enhances T-cell mediated anti-tumor activities by IL-2 via attenuation of fibrotic reaction in a humanized mouse model of pancreatic ductal adenocarcinoma. Biomed Pharmacother 2023; 159:114212. [PMID: 36610224 DOI: 10.1016/j.biopha.2022.114212] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 12/20/2022] [Accepted: 12/31/2022] [Indexed: 01/07/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most aggressive cancers, with high mortality and recurrence rate. In this study, we generated a human immune system mouse model by transplanting human peripheral blood mononuclear cells into NSG-B2m mice followed by xenografting AsPC-1 cells, after which we assessed the role of transforming growth factor-β2 (TGF-β2) in T-cell-mediated anti-tumor immunity. We observed that inhibiting the TGF-β2 production by TGF-β2 antisense oligonucleotide (TASO) combined with IL-2 delays pancreatic cancer growth. Co-treatment of TASO and IL-2 had little effect on the SMAD-dependent pathway, but significantly inhibited the Akt phosphorylation and sequentially activated GSK-3β. Activation of GSK-3β by TASO subsequently suppressed β-catenin and α-SMA expression and resulted in attenuated fibrotic reactions, facilitating the infiltration of CD8 + cytotoxic T lymphocytes (CTLs) into the tumor. TGF-β2 inhibition suppressed the Foxp3 + regulatory T-cells in peripheral blood and tumors, thereby enhancing the tumoricidal effects of CTLs associated with increased granzyme B and cleaved caspase-3. Moreover, changes in the T-cell composition in peripheral blood and at the tumor site by TASO and IL-2 induced the increase of pro-inflammatory cytokines such as IFN-γ and TNF-α and the decrease of anti-inflammatory cytokines such as TGF-βs. These results indicate that the TGF-β2 inhibition by TASO combined with IL-2 enhances the T-cell mediated anti-tumor immunity against SMAD4-mutated PDAC by modulating the tumor-associated fibrosis, suggesting that TASO in combination with IL-2 may be a promising immunotherapeutic intervention for PDAC.
Collapse
|
23
|
Ito R, Katano I, Kwok IWH, Ng LG, Ida-Tanaka M, Ohno Y, Mu Y, Morita H, Nishinaka E, Nishime C, Mochizuki M, Kawai K, Chien TH, Yunqian Z, Yiping F, Hua LH, Celhar T, Yen Chan JK, Takahashi T, Goto M, Ogura T, Takahashi R, Ito M. Efficient differentiation of human neutrophils with recapitulation of emergency granulopoiesis in human G-CSF knockin humanized mice. Cell Rep 2022; 41:111841. [PMID: 36543125 DOI: 10.1016/j.celrep.2022.111841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 09/28/2022] [Accepted: 11/24/2022] [Indexed: 12/24/2022] Open
Abstract
Neutrophils are critical mediators during the early stages of innate inflammation in response to bacterial or fungal infections. A human hematopoietic system reconstituted in humanized mice aids in the study of human hematology and immunology. However, the poor development of human neutrophils is a well-known limitation of humanized mice. Here, we generate a human granulocyte colony-stimulating factor (hG-CSF) knockin (KI) NOD/Shi-scid-IL2rgnull (NOG) mouse in which hG-CSF is systemically expressed while the mouse G-CSF receptor is disrupted. These mice generate high numbers of mature human neutrophils, which can be readily mobilized into the periphery, compared with conventional NOG mice. Moreover, these neutrophils exhibit infection-mediated emergency granulopoiesis and are capable of efficient phagocytosis and reactive oxygen species production. Thus, hG-CSF KI mice provide a useful model for studying the development of human neutrophils, emergency granulopoiesis, and a potential therapeutic model for sepsis.
Collapse
Affiliation(s)
- Ryoji Ito
- Central Institute for Experimental Animals, 3-25-12 Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa 210-0821, Japan.
| | - Ikumi Katano
- Central Institute for Experimental Animals, 3-25-12 Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa 210-0821, Japan
| | - Immanuel W H Kwok
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A(∗)STAR), Singapore 138648, Singapore
| | - Lai Guan Ng
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A(∗)STAR), Singapore 138648, Singapore
| | - Miyuki Ida-Tanaka
- Central Institute for Experimental Animals, 3-25-12 Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa 210-0821, Japan
| | - Yusuke Ohno
- Central Institute for Experimental Animals, 3-25-12 Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa 210-0821, Japan
| | - Yunmei Mu
- Central Institute for Experimental Animals, 3-25-12 Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa 210-0821, Japan
| | - Hanako Morita
- Central Institute for Experimental Animals, 3-25-12 Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa 210-0821, Japan
| | - Eiko Nishinaka
- Central Institute for Experimental Animals, 3-25-12 Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa 210-0821, Japan
| | - Chiyoko Nishime
- Central Institute for Experimental Animals, 3-25-12 Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa 210-0821, Japan
| | - Misa Mochizuki
- Central Institute for Experimental Animals, 3-25-12 Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa 210-0821, Japan
| | - Kenji Kawai
- Central Institute for Experimental Animals, 3-25-12 Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa 210-0821, Japan
| | - Tay Hui Chien
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A(∗)STAR), Singapore 138648, Singapore
| | - Zhao Yunqian
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A(∗)STAR), Singapore 138648, Singapore
| | - Fan Yiping
- Department of Reproductive Medicine, KK Women's and Children's Hospital, Singapore 229899, Singapore
| | - Liew Hui Hua
- Department of Reproductive Medicine, KK Women's and Children's Hospital, Singapore 229899, Singapore
| | - Teja Celhar
- Central Institute for Experimental Animals, 3-25-12 Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa 210-0821, Japan; Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A(∗)STAR), Singapore 138648, Singapore
| | - Jerry Kok Yen Chan
- Department of Reproductive Medicine, KK Women's and Children's Hospital, Singapore 229899, Singapore
| | - Takeshi Takahashi
- Central Institute for Experimental Animals, 3-25-12 Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa 210-0821, Japan
| | - Motohito Goto
- Central Institute for Experimental Animals, 3-25-12 Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa 210-0821, Japan
| | - Tomoyuki Ogura
- Central Institute for Experimental Animals, 3-25-12 Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa 210-0821, Japan
| | - Riichi Takahashi
- Central Institute for Experimental Animals, 3-25-12 Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa 210-0821, Japan
| | - Mamoru Ito
- Central Institute for Experimental Animals, 3-25-12 Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa 210-0821, Japan
| |
Collapse
|
24
|
Manjili MH. The adaptation model of immunity: Is the goal of central tolerance to eliminate defective T cells or self-reactive T cells? Scand J Immunol 2022; 96:e13209. [PMID: 36239215 PMCID: PMC9539632 DOI: 10.1111/sji.13209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 06/09/2022] [Accepted: 08/01/2022] [Indexed: 11/29/2022]
Abstract
The self-non-self model and the danger model are designed to understand how an immune response is induced. These models are not meant to predict if an immune response may succeed or fail in destroying/controlling its target. However, these immunological models rely on either self-antigens or self-dendritic cells for understanding of central tolerance, which have been discussed by Fuchs and Matzinger in response to Al-Yassin. In an attempt to address some questions that these models are facing when it comes to understanding central tolerance, I propose that the goal of negative selection in the thymus is to eliminate defective T cells but not self-reactive T cells. Therefore, any escape from negative selection could increase lymphopenia because of the depletion of defective naïve T cells outside the thymus, as seen in the elderly.
Collapse
Affiliation(s)
- Masoud H. Manjili
- Department of Microbiology & Immunology, VCU School of MedicineVCU Massey Cancer CenterRichmondVirginiaUSA
| |
Collapse
|
25
|
Kanikarla Marie P, Sorokin AV, Bitner LA, Aden R, Lam M, Manyam G, Woods MN, Anderson A, Capasso A, Fowlkes N, Overman MJ, Menter DG, Kopetz S. Autologous humanized mouse models to study combination and single-agent immunotherapy for colorectal cancer patient-derived xenografts. Front Oncol 2022; 12:994333. [PMID: 36212401 PMCID: PMC9532947 DOI: 10.3389/fonc.2022.994333] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 09/02/2022] [Indexed: 11/24/2022] Open
Abstract
Designing studies of immunotherapy is limited due to a lack of pre-clinical models that reliably predict effective immunotherapy responses. To address this gap, we developed humanized mouse models of colorectal cancer (CRC) incorporating patient-derived xenografts (PDX) with human peripheral blood mononuclear cells (PBMC). Humanized mice with CRC PDXs were generated via engraftment of autologous (isolated from the same patients as the PDXs) or allogeneic (isolated from healthy donors) PBMCs. Human T cells were detected in mouse blood, tissues, and infiltrated the implanted PDXs. The inclusion of anti-PD-1 therapy revealed that tumor responses in autologous but not allogeneic models were more comparable to that of patients. An overall non-specific graft-vs-tumor effect occurred in allogeneic models and negatively correlated with that seen in patients. In contrast, autologous humanized mice more accurately correlated with treatment outcomes by engaging pre-existing tumor specific T-cell populations. As autologous T cells appear to be the major drivers of tumor response thus, autologous humanized mice may serve as models at predicting treatment outcomes in pre-clinical settings for therapies reliant on pre-existing tumor specific T-cell populations.
Collapse
Affiliation(s)
- Preeti Kanikarla Marie
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Alexey V. Sorokin
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Lea A. Bitner
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Rebecca Aden
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Michael Lam
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Ganiraju Manyam
- Department of Bioinformatics & Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Melanie N. Woods
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Amanda Anderson
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Anna Capasso
- Department of Oncology, The University of Texas Health Austin, Austin, TX, United States
| | - Natalie Fowlkes
- Department of Veterinary Medicine & Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Michael J. Overman
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - David G. Menter
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Scott Kopetz
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
- *Correspondence: Scott Kopetz,
| |
Collapse
|
26
|
Chen C, Jing W, Chen Y, Wang G, Abdalla M, Gao L, Han M, Shi C, Li A, Sun P, Jiang X, Yang Z, Zhang S, Zhang J, Tang C, Liu Y, Zhang R, Xu F, Dong B, Li X, Liu M, Qiang B, Sun Y, Wei X, Li J, Hu Q, Jiang X. Intracavity generation of glioma stem cell-specific CAR macrophages primes locoregional immunity for postoperative glioblastoma therapy. Sci Transl Med 2022; 14:eabn1128. [PMID: 35921473 DOI: 10.1126/scitranslmed.abn1128] [Citation(s) in RCA: 88] [Impact Index Per Article: 44.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Glioblastoma multiforme (GBM) remains incurable despite aggressive implementation of multimodal treatments after surgical debulking. Almost all patients with GBM relapse within a narrow margin around the initial resected lesion due to postsurgery residual glioma stem cells (GSCs). Tracking and eradicating postsurgery residual GSCs is critical for preventing postoperative relapse of this devastating disease, yet effective strategies remain elusive. Here, we report a cavity-injectable nanoporter-hydrogel superstructure that creates GSC-specific chimeric antigen receptor (CAR) macrophages/microglia (MΦs) surrounding the cavity to prevent GBM relapse. Specifically, we demonstrate that the CAR gene-laden nanoporter in the hydrogel can introduce GSC-targeted CAR genes into MΦ nuclei after intracavity delivery to generate CAR-MΦs in mouse models of GBM. These CAR-MΦs were able to seek and engulf GSCs and clear residual GSCs by stimulating an adaptive antitumor immune response in the tumor microenvironment and prevented postoperative glioma relapse by inducing long-term antitumor immunity in mice. In an orthotopic patient-derived glioblastoma humanized mouse model, the combined treatment with nanoporter-hydrogel superstructure and CD47 antibody increased the frequency of positive immune responding cells and suppressed the negative immune regulating cells, conferring a robust tumoricidal immunity surrounding the postsurgical cavity and inhibiting postoperative glioblastoma relapse. Therefore, our work establishes a locoregional treatment strategy for priming cancer stem cell-specific tumoricidal immunity with broad application in patients suffering from recurrent malignancies.
Collapse
Affiliation(s)
- Chen Chen
- Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Cultural West Road, Jinan, Shandong Province 250012, China.,NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, Shandong University, 44 Cultural West Road, Jinan, Shandong Province 250012, China
| | - Weiqiang Jing
- Department of Urology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, 107 Cultural West Road, Jinan, Shandong Province 250012, China
| | - Yu Chen
- Pharmaceutical Sciences Division, Wisconsin Center for NanoBioSystems, School of Pharmacy, University of Wisconsin-Madison (UW-Madison), Madison, WI 53705, USA
| | - Ganyu Wang
- Department of Pediatric Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province 250012, China
| | - Mohnad Abdalla
- Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Cultural West Road, Jinan, Shandong Province 250012, China
| | - Lin Gao
- Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Cultural West Road, Jinan, Shandong Province 250012, China
| | - Maosen Han
- Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Cultural West Road, Jinan, Shandong Province 250012, China
| | - Chongdeng Shi
- Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Cultural West Road, Jinan, Shandong Province 250012, China
| | - Anning Li
- Department of Radiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, 107 Cultural West Road, Jinan, Shandong Province 250012, China
| | - Peng Sun
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province 250355, China
| | - Xin Jiang
- Department of Urology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, 107 Cultural West Road, Jinan, Shandong Province 250012, China
| | - Zhenmei Yang
- Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Cultural West Road, Jinan, Shandong Province 250012, China
| | - Shengchang Zhang
- Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Cultural West Road, Jinan, Shandong Province 250012, China
| | - Jing Zhang
- Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Cultural West Road, Jinan, Shandong Province 250012, China
| | - Chunwei Tang
- Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Cultural West Road, Jinan, Shandong Province 250012, China
| | - Ying Liu
- Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Cultural West Road, Jinan, Shandong Province 250012, China
| | - Rui Zhang
- Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Cultural West Road, Jinan, Shandong Province 250012, China
| | - Fengbo Xu
- Yinfeng Biological Group Co. Ltd., 1109 Gangxing San Lu, High-tech Zone, Jinan, Shandong Province 250102, China
| | - Baixiang Dong
- Yinfeng Biological Group Co. Ltd., 1109 Gangxing San Lu, High-tech Zone, Jinan, Shandong Province 250102, China
| | - Xueen Li
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, 107 Cultural West Road, Jinan, Shandong Province 250012, China
| | - Minglu Liu
- Bellastem Biotechnology Limited, High-Tech incubator, Intersection of Liquan Street and Gaoxin Er Road, Gaomi City, Shandong Province 261500, China
| | - Bangming Qiang
- Bellastem Biotechnology Limited, High-Tech incubator, Intersection of Liquan Street and Gaoxin Er Road, Gaomi City, Shandong Province 261500, China
| | - Yanhua Sun
- Shandong Provincial Key Laboratory of Microparticles Drug Delivery Technology, Qilu Pharmaceutical Co. Ltd., Jinan, Shandong Province 250100, China
| | - Xia Wei
- Department of Pharmacology and Toxicology, Shandong Institute for Food and Drug Control, Jinan, Shandong Province 250101, China
| | - Jun Li
- Department of Pharmacology and Toxicology, Shandong Institute for Food and Drug Control, Jinan, Shandong Province 250101, China
| | - Quanyin Hu
- Pharmaceutical Sciences Division, Wisconsin Center for NanoBioSystems, School of Pharmacy, University of Wisconsin-Madison (UW-Madison), Madison, WI 53705, USA
| | - Xinyi Jiang
- Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Cultural West Road, Jinan, Shandong Province 250012, China.,NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, Shandong University, 44 Cultural West Road, Jinan, Shandong Province 250012, China
| |
Collapse
|
27
|
Connolly KA, Fitzgerald B, Damo M, Joshi NS. Novel Mouse Models for Cancer Immunology. ANNUAL REVIEW OF CANCER BIOLOGY 2022; 6:269-291. [PMID: 36875867 PMCID: PMC9979244 DOI: 10.1146/annurev-cancerbio-070620-105523] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Mouse models for the study of cancer immunology provide excellent systems in which to test biological mechanisms of the immune response against cancer. Historically, these models have been designed to have different strengths based on the current major research questions at the time. As such, many mouse models of immunology used today were not originally developed to study questions currently plaguing the relatively new field of cancer immunology, but instead have been adapted for such purposes. In this review, we discuss various mouse model of cancer immunology in a historical context as a means to provide a fuller perspective of each model's strengths. From this outlook, we discuss the current state of the art and strategies for tackling future modeling challenges.
Collapse
Affiliation(s)
- Kelli A. Connolly
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06519, USA
| | - Brittany Fitzgerald
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06519, USA
| | - Martina Damo
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06519, USA
| | - Nikhil S. Joshi
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06519, USA
| |
Collapse
|
28
|
Cogels MM, Rouas R, Ghanem GE, Martinive P, Awada A, Van Gestel D, Krayem M. Humanized Mice as a Valuable Pre-Clinical Model for Cancer Immunotherapy Research. Front Oncol 2021; 11:784947. [PMID: 34869042 PMCID: PMC8636317 DOI: 10.3389/fonc.2021.784947] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 10/29/2021] [Indexed: 01/31/2023] Open
Abstract
Immunotherapy with checkpoint inhibitors opened new horizons in cancer treatment. Clinical trials for novel immunotherapies or unexplored combination regimens either need years of development or are simply impossible to perform like is the case in cancer patients with limited life expectancy. Thus, the need for preclinical models that rapidly and safely allow for a better understanding of underlying mechanisms, drug kinetics and toxicity leading to the selection of the best regimen to be translated into the clinic, is of high importance. Humanized mice that can bear both human immune system and human tumors, are increasingly used in recent preclinical immunotherapy studies and represent a remarkably unprecedented tool in this field. In this review, we describe, summarize, and discuss the recent advances of humanized mouse models used for cancer immunotherapy research and the challenges faced during their establishment. We also highlight the lack of preclinical studies using this model for radiotherapy-based research and argue that it can be a great asset to understand and answer many open questions around radiation therapy such as its presumed associated "abscopal effect".
Collapse
Affiliation(s)
- Morgane M. Cogels
- Department of Radiation Oncology, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
- Laboratory of Clinical and Experimental Oncology (LOCE), Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Redouane Rouas
- Laboratory of Cellular Therapy (UTCH), Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Ghanem E. Ghanem
- Laboratory of Clinical and Experimental Oncology (LOCE), Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Philippe Martinive
- Department of Radiation Oncology, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Ahmad Awada
- Laboratory of Clinical and Experimental Oncology (LOCE), Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
- Department of Medical Oncology, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Dirk Van Gestel
- Department of Radiation Oncology, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Mohammad Krayem
- Department of Radiation Oncology, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
- Laboratory of Clinical and Experimental Oncology (LOCE), Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| |
Collapse
|
29
|
Romualdo GR, Leroy K, Costa CJS, Prata GB, Vanderborght B, da Silva TC, Barbisan LF, Andraus W, Devisscher L, Câmara NOS, Vinken M, Cogliati B. In Vivo and In Vitro Models of Hepatocellular Carcinoma: Current Strategies for Translational Modeling. Cancers (Basel) 2021; 13:5583. [PMID: 34771745 PMCID: PMC8582701 DOI: 10.3390/cancers13215583] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 11/02/2021] [Accepted: 11/04/2021] [Indexed: 12/24/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the sixth most common cancer worldwide and the third leading cause of cancer-related death globally. HCC is a complex multistep disease and usually emerges in the setting of chronic liver diseases. The molecular pathogenesis of HCC varies according to the etiology, mainly caused by chronic hepatitis B and C virus infections, chronic alcohol consumption, aflatoxin-contaminated food, and non-alcoholic fatty liver disease associated with metabolic syndrome or diabetes mellitus. The establishment of HCC models has become essential for both basic and translational research to improve our understanding of the pathophysiology and unravel new molecular drivers of this disease. The ideal model should recapitulate key events observed during hepatocarcinogenesis and HCC progression in view of establishing effective diagnostic and therapeutic strategies to be translated into clinical practice. Despite considerable efforts currently devoted to liver cancer research, only a few anti-HCC drugs are available, and patient prognosis and survival are still poor. The present paper provides a state-of-the-art overview of in vivo and in vitro models used for translational modeling of HCC with a specific focus on their key molecular hallmarks.
Collapse
Affiliation(s)
- Guilherme Ribeiro Romualdo
- Department of Pathology, School of Veterinary Medicine and Animal Science, University of São Paulo (USP), São Paulo 05508-270, Brazil; (G.R.R.); (C.J.S.C.); (T.C.d.S.)
- Department of Structural and Functional Biology, Biosciences Institute, São Paulo State University (UNESP), Botucatu 18618-689, Brazil; (G.B.P.); (L.F.B.)
- Department of Pathology, Botucatu Medical School, São Paulo State University (UNESP), Botucatu 18618-687, Brazil
| | - Kaat Leroy
- Department of Pharmaceutical and Pharmacological Sciences, Vrije Universiteit Brussel, 1090 Brussels, Belgium; (K.L.); (M.V.)
| | - Cícero Júlio Silva Costa
- Department of Pathology, School of Veterinary Medicine and Animal Science, University of São Paulo (USP), São Paulo 05508-270, Brazil; (G.R.R.); (C.J.S.C.); (T.C.d.S.)
| | - Gabriel Bacil Prata
- Department of Structural and Functional Biology, Biosciences Institute, São Paulo State University (UNESP), Botucatu 18618-689, Brazil; (G.B.P.); (L.F.B.)
- Department of Pathology, Botucatu Medical School, São Paulo State University (UNESP), Botucatu 18618-687, Brazil
| | - Bart Vanderborght
- Gut-Liver Immunopharmacology Unit, Basic and Applied Medical Sciences, Liver Research Center Ghent, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium;
- Hepatology Research Unit, Internal Medicine and Paediatrics, Liver Research Center Ghent, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium;
| | - Tereza Cristina da Silva
- Department of Pathology, School of Veterinary Medicine and Animal Science, University of São Paulo (USP), São Paulo 05508-270, Brazil; (G.R.R.); (C.J.S.C.); (T.C.d.S.)
| | - Luís Fernando Barbisan
- Department of Structural and Functional Biology, Biosciences Institute, São Paulo State University (UNESP), Botucatu 18618-689, Brazil; (G.B.P.); (L.F.B.)
| | - Wellington Andraus
- Department of Gastroenterology, Clinics Hospital, School of Medicine, University of São Paulo (HC-FMUSP), São Paulo 05403-000, Brazil;
| | - Lindsey Devisscher
- Hepatology Research Unit, Internal Medicine and Paediatrics, Liver Research Center Ghent, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium;
| | - Niels Olsen Saraiva Câmara
- Department of Immunology, Institute of Biomedical Sciences IV, University of São Paulo (USP), São Paulo 05508-000, Brazil;
| | - Mathieu Vinken
- Department of Pharmaceutical and Pharmacological Sciences, Vrije Universiteit Brussel, 1090 Brussels, Belgium; (K.L.); (M.V.)
| | - Bruno Cogliati
- Department of Pathology, School of Veterinary Medicine and Animal Science, University of São Paulo (USP), São Paulo 05508-270, Brazil; (G.R.R.); (C.J.S.C.); (T.C.d.S.)
| |
Collapse
|
30
|
Peng S, Hu P, Xiao YT, Lu W, Guo D, Hu S, Xie J, Wang M, Yu W, Yang J, Chen H, Zhang X, Zhu Y, Wang Y, Yang Y, Zhu G, Chen S, Wang J, Zhang B, Chen W, Wu H, Sun Z, Ding T, Zhang H, Yi Z, Liu M, Ren S. Single-cell analysis reveals EP4 as a target for restoring T cell infiltration and sensitizing prostate cancer to immunotherapy. Clin Cancer Res 2021; 28:552-567. [PMID: 34740924 DOI: 10.1158/1078-0432.ccr-21-0299] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 04/22/2021] [Accepted: 10/29/2021] [Indexed: 12/09/2022]
Abstract
PURPOSE Immunotherapies targeting immune checkpoint molecules have shown promising treatment for a subset of cancers; however, many "cold" tumors, such as prostate cancer, remain unresponsive. We aimed to identify a potential targetable marker relevant to prostate cancer and develop novel immunotherapy. EXPERIMENTAL DESIGN Analysis of transcriptomic profiles at single-cell resolution was performed in clinical patients' samples, along with integrated analysis of multiple RNA-seq datasets. The antitumor activity of YY001, a novel EP4 antagonist, combined with anti-programmed cell death protein 1 (PD-1) antibody was evaluated both in vitro and in vivo Results: We identified EP4 (PTGER4) as expressed in epithelial cells and various immune cells and involved in modulating the prostate cancer immune microenvironment. YY001, a novel EP4 antagonist, inhibited the differentiation, maturation, and immunosuppressive function of myeloid-derived suppressor cells (MDSCs) while enhancing the proliferation and anticancer functions of T cells. Furthermore, it reversed the infiltration levels of MDSCs and T cells in the tumor microenvironment by overturning the chemokine profile of tumor cells in vitro and in vivo The combined immunotherapy demonstrated a robust antitumor immune response as indicated by the robust accumulation and activation of CD8+ cytotoxic T cells, with a significantly decreased MDSC ratio and reduced MDSC immunosuppression function. CONCLUSIONS Our study identified EP4 as a specific target for prostate cancer immunotherapy and demonstrated that YY001 inhibited the growth of prostate tumors by regulating the immune microenvironment and strongly synergized with anti-PD-1 antibodies to convert completely unresponsive prostate cancers into responsive cancers, resulting in marked tumor regression, long-term survival, and lasting immunologic memory.
Collapse
Affiliation(s)
- Shihong Peng
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University
| | - Pan Hu
- East China Normal University
| | - Yu-Tian Xiao
- Department of Urology, Shanghai Changhai Hospital
| | - Weiqiang Lu
- East China Normal University, Institute of Biomedical Sciences and School of Life Sciences
| | - Dandan Guo
- Biomedical Sciences and School of Life Sciences, East China Normal University
| | - Shixiu Hu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University
| | | | | | - Weiwei Yu
- School of Life Sciences, Institute of Biomedical Sciences
| | - Junjie Yang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University
| | | | | | - Yasheng Zhu
- Department of Urology, Second Military Medical University
| | | | - Yue Yang
- Department of Urology, Shanghai Changhai Hospital, Second Military Medical University
| | | | | | | | | | | | - Huangan Wu
- Shanghai University of Traditional Chinese Medicine
| | - Zhenliang Sun
- Shanghai University of Medicine & Health Sciences Affiliated Sixth People's Hospital South Campus
| | - Tao Ding
- Urology, Shanghai Putuo Hospital, Shanghai Traditional Chinese Medicine University
| | - Hankun Zhang
- East China Normal University, Institute of Biomedical Sciences and School of Life Sciences
| | - Zhengfang Yi
- Institute of Biomedical Sciences and School of Life Sciences, East China Normal University
| | - Mingyao Liu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University
| | | |
Collapse
|
31
|
Scherer SD, Riggio AI, Haroun F, DeRose YS, Ekiz HA, Fujita M, Toner J, Zhao L, Li Z, Oesterreich S, Samatar AA, Welm AL. An immune-humanized patient-derived xenograft model of estrogen-independent, hormone receptor positive metastatic breast cancer. Breast Cancer Res 2021; 23:100. [PMID: 34717714 PMCID: PMC8556932 DOI: 10.1186/s13058-021-01476-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 10/11/2021] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Metastatic breast cancer (MBC) is incurable, with a 5-year survival rate of 28%. In the USA, more than 42,000 patients die from MBC every year. The most common type of breast cancer is estrogen receptor-positive (ER+), and more patients die from ER+ breast cancer than from any other subtype. ER+ tumors can be successfully treated with hormone therapy, but many tumors acquire endocrine resistance, at which point treatment options are limited. There is an urgent need for model systems that better represent human ER+ MBC in vivo, where tumors can metastasize. Patient-derived xenografts (PDX) made from MBC spontaneously metastasize, but the immunodeficient host is a caveat, given the known role of the immune system in tumor progression and response to therapy. Thus, we attempted to develop an immune-humanized PDX model of ER+ MBC. METHODS NSG-SGM3 mice were immune-humanized with CD34+ hematopoietic stem cells, followed by engraftment of human ER+ endocrine resistant MBC tumor fragments. Strategies for exogenous estrogen supplementation were compared, and immune-humanization in blood, bone marrow, spleen, and tumors was assessed by flow cytometry and tissue immunostaining. Characterization of the new model includes assessment of the human tumor microenvironment performed by immunostaining. RESULTS We describe the development of an immune-humanized PDX model of estrogen-independent endocrine resistant ER+ MBC. Importantly, our model harbors a naturally occurring ESR1 mutation, and immune-humanization recapitulates the lymphocyte-excluded and myeloid-rich tumor microenvironment of human ER+ breast tumors. CONCLUSION This model sets the stage for development of other clinically relevant models of human breast cancer and should allow future studies on mechanisms of endocrine resistance and tumor-immune interactions in an immune-humanized in vivo setting.
Collapse
Affiliation(s)
- Sandra D Scherer
- Department of Oncological Sciences, University of Utah, 2000 Circle of Hope, Salt Lake City, UT, 84112, USA
- Huntsman Cancer Institute, University of Utah, 2000 Circle of Hope, Salt Lake City, UT, 84112, USA
| | - Alessandra I Riggio
- Department of Oncological Sciences, University of Utah, 2000 Circle of Hope, Salt Lake City, UT, 84112, USA
- Huntsman Cancer Institute, University of Utah, 2000 Circle of Hope, Salt Lake City, UT, 84112, USA
| | - Fadi Haroun
- Department of Oncological Sciences, University of Utah, 2000 Circle of Hope, Salt Lake City, UT, 84112, USA
- Huntsman Cancer Institute, University of Utah, 2000 Circle of Hope, Salt Lake City, UT, 84112, USA
| | - Yoko S DeRose
- Department of Oncological Sciences, University of Utah, 2000 Circle of Hope, Salt Lake City, UT, 84112, USA
- Huntsman Cancer Institute, University of Utah, 2000 Circle of Hope, Salt Lake City, UT, 84112, USA
| | - H Atakan Ekiz
- Department of Oncological Sciences, University of Utah, 2000 Circle of Hope, Salt Lake City, UT, 84112, USA
- Huntsman Cancer Institute, University of Utah, 2000 Circle of Hope, Salt Lake City, UT, 84112, USA
| | - Maihi Fujita
- Department of Oncological Sciences, University of Utah, 2000 Circle of Hope, Salt Lake City, UT, 84112, USA
- Huntsman Cancer Institute, University of Utah, 2000 Circle of Hope, Salt Lake City, UT, 84112, USA
| | - Jennifer Toner
- Department of Oncological Sciences, University of Utah, 2000 Circle of Hope, Salt Lake City, UT, 84112, USA
- Huntsman Cancer Institute, University of Utah, 2000 Circle of Hope, Salt Lake City, UT, 84112, USA
| | - Ling Zhao
- Department of Oncological Sciences, University of Utah, 2000 Circle of Hope, Salt Lake City, UT, 84112, USA
- Huntsman Cancer Institute, University of Utah, 2000 Circle of Hope, Salt Lake City, UT, 84112, USA
| | - Zheqi Li
- Department of Pharmacology and Chemical Biology, UPMC Hillman Cancer Center, Magee Women's Research Institute, University of Pittsburgh, 204 Craft Avenue, Pittsburgh, PA, 15213, USA
| | - Steffi Oesterreich
- Department of Pharmacology and Chemical Biology, UPMC Hillman Cancer Center, Magee Women's Research Institute, University of Pittsburgh, 204 Craft Avenue, Pittsburgh, PA, 15213, USA
| | - Ahmed A Samatar
- Zentalis Pharmaceuticals, Inc., 10835 Road to the Cure, Suite 205, San Diego, CA, 92121, USA
| | - Alana L Welm
- Department of Oncological Sciences, University of Utah, 2000 Circle of Hope, Salt Lake City, UT, 84112, USA.
- Huntsman Cancer Institute, University of Utah, 2000 Circle of Hope, Salt Lake City, UT, 84112, USA.
| |
Collapse
|
32
|
AAV-mediated in vivo CAR gene therapy for targeting human T-cell leukemia. Blood Cancer J 2021; 11:119. [PMID: 34162832 PMCID: PMC8222347 DOI: 10.1038/s41408-021-00508-1] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 05/19/2021] [Accepted: 06/01/2021] [Indexed: 12/12/2022] Open
Abstract
Chimeric antigen receptor (CAR) T-cell therapy is the most active field in immuno-oncology and brings substantial benefit to patients with B cell malignancies. However, the complex procedure for CAR T-cell generation hampers its widespread applications. Here, we describe a novel approach in which human CAR T cells can be generated within the host upon injecting an Adeno-associated virus (AAV) vector carrying the CAR gene, which we call AAV delivering CAR gene therapy (ACG). Upon single infusion into a humanized NOD.Cg-Prkdcscid Il2rgem26/Nju tumor mouse model of human T-cell leukemia, AAV generates sufficient numbers of potent in vivo CAR cells, resulting in tumor regression; these in vivo-generated CAR cells produce antitumor immunological characteristics. This instantaneous generation of in vivo CAR T cells may bypass the need for patient lymphodepletion, as well as the β processes of traditional CAR T-cell production, which may make CAR therapy simpler and less expensive. It may allow the development of intricate, individualized treatments in the form of on-demand and diverse therapies.
Collapse
|
33
|
Guo W, Zhang C, Qiao T, Zhao J, Shi C. Strategies for the Construction of Mouse Models With Humanized Immune System and Evaluation of Tumor Immune Checkpoint Inhibitor Therapy. Front Oncol 2021; 11:673199. [PMID: 33996603 PMCID: PMC8117211 DOI: 10.3389/fonc.2021.673199] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Accepted: 04/13/2021] [Indexed: 12/15/2022] Open
Abstract
Immunotherapy has been used as a first-line treatment for a variety of advanced tumors, allowing remarkable progress to be made in cancer treatment. Nonetheless, only a small number of patients can benefit from immune checkpoint inhibitor monotherapy. To improve the effect of immunotherapy, the underlying mechanism of combination therapy was investigated in the context of an intact human tumor immune microenvironment using mice with a human immune system (HIS) bearing human tumors. Herein, we summarize and discuss strategies for the development and use of HIS mice models in tumor immunotherapies. Most importantly, this review proposes a method of t11umor identification and classification in HIS mice based on the tumor-infiltrating lymphocytes and PD-L1 expression, and according to this classification, we propose different combination treatment strategies that can be utilized to enhance the effect of immunotherapy. Thus, we provide effective experimental schemes for tumor immunotherapy in HIS mice models.
Collapse
Affiliation(s)
- Wenwen Guo
- Division of Cancer Biology, Laboratory Animal Center, Fourth Military Medical University, Xi'an, China.,School of Basic Medical Sciences, Medical College of Yan'an University, Yanan, China
| | - Caiqin Zhang
- Division of Cancer Biology, Laboratory Animal Center, Fourth Military Medical University, Xi'an, China
| | - Tianyun Qiao
- Department of Thoracic Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Jumei Zhao
- School of Basic Medical Sciences, Medical College of Yan'an University, Yanan, China
| | - Changhong Shi
- Division of Cancer Biology, Laboratory Animal Center, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
34
|
Lala PK, Nandi P, Hadi A, Halari C. A crossroad between placental and tumor biology: What have we learnt? Placenta 2021; 116:12-30. [PMID: 33958236 DOI: 10.1016/j.placenta.2021.03.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Revised: 02/27/2021] [Accepted: 03/02/2021] [Indexed: 01/06/2023]
Abstract
Placenta in certain species including the human has evolved as a highly invasive tumor-like organ invading the uterus aned its vasculature to derive oxygen and nutrients for the fetus and exchange waste products. While several excellent reviews have been written comparing hemochorial placentation with tumors, no comprehensive review is available dealing with mechanistic insights into what makes them different, and what tumor biologists can learn from placental biologists, and vice versa. In this review, we analyze the structure-function relationship of the human placenta, emphasizing the functional need of the spatio-temporally orchestrated trophoblast invasiveness for fetal development and growth, and pathological consequences of aberrant invasiveness for fetal and maternal health. We then analyze similarities and differences between the placenta and invasive tumors in terms of hallmarks of cancer, some key molecules regulating their invasive functions, and how placental cancers (choriocarcinomas) or other cancers become refractory or even addicted to these invasion-restraining molecules. We cite in vitro models of human trophoblast and choriocarcinoma cell lines utilized to study mechanisms in normal placental development as well as those responsible for tumor progression. We discuss the pathobiology of hyper-invasive placentas and show thattrophoblastic neoplasias are a unique and heterogeneous class of tumors. We delve into the questions as to why metastasis from other organs rarely occurs at the placental site and whether pregnancy makes the mother more or less vulnerable to cancer-related morbidity/mortality. We attempt to compare trophoblast stem cells and cancer stem cells. Finally, we leave the readers with some thoughts as foods of future investigations.
Collapse
Affiliation(s)
- Peeyush K Lala
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Westernat Ontario, London, Ontario, N6A5C1, Canada; Associate Scientist, Children's Health Research Institute, University of Western Ontario, London, Ontario, Canada N6C2V5.
| | - Pinki Nandi
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Westernat Ontario, London, Ontario, N6A5C1, Canada.
| | - Ali Hadi
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Westernat Ontario, London, Ontario, N6A5C1, Canada.
| | - Chidambra Halari
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Westernat Ontario, London, Ontario, N6A5C1, Canada.
| |
Collapse
|
35
|
Martínez Bedoya D, Dutoit V, Migliorini D. Allogeneic CAR T Cells: An Alternative to Overcome Challenges of CAR T Cell Therapy in Glioblastoma. Front Immunol 2021; 12:640082. [PMID: 33746981 PMCID: PMC7966522 DOI: 10.3389/fimmu.2021.640082] [Citation(s) in RCA: 77] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 02/08/2021] [Indexed: 12/18/2022] Open
Abstract
Chimeric antigen receptor (CAR) T cell therapy has emerged as one of the major breakthroughs in cancer immunotherapy in the last decade. Outstanding results in hematological malignancies and encouraging pre-clinical anti-tumor activity against a wide range of solid tumors have made CAR T cells one of the most promising fields for cancer therapies. CAR T cell therapy is currently being investigated in solid tumors including glioblastoma (GBM), a tumor for which survival has only modestly improved over the past decades. CAR T cells targeting EGFRvIII, Her2, or IL-13Rα2 have been tested in GBM, but the first clinical trials have shown modest results, potentially due to GBM heterogeneity and to the presence of an immunosuppressive microenvironment. Until now, the use of autologous T cells to manufacture CAR products has been the norm, but this approach has several disadvantages regarding production time, cost, manufacturing delay and dependence on functional fitness of patient T cells, often reduced by the disease or previous therapies. Universal “off-the-shelf,” or allogeneic, CAR T cells is an alternative that can potentially overcome these issues, and allow for multiple modifications and CAR combinations to target multiple tumor antigens and avoid tumor escape. Advances in genome editing tools, especially via CRISPR/Cas9, might allow overcoming the two main limitations of allogeneic CAR T cells product, i.e., graft-vs.-host disease and host allorejection. Here, we will discuss how allogeneic CAR T cells could allow for multivalent approaches and alteration of the tumor microenvironment, potentially allowing the development of next generation therapies for the treatment of patients with GBM.
Collapse
Affiliation(s)
- Darel Martínez Bedoya
- Center for Translational Research in Onco-Hematology, University of Geneva, Geneva, Switzerland.,Swiss Cancer Center Léman, Lausanne, Switzerland.,Brain Tumor and Immune Cell Engineering Group, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Valérie Dutoit
- Center for Translational Research in Onco-Hematology, University of Geneva, Geneva, Switzerland.,Swiss Cancer Center Léman, Lausanne, Switzerland.,Brain Tumor and Immune Cell Engineering Group, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Denis Migliorini
- Center for Translational Research in Onco-Hematology, University of Geneva, Geneva, Switzerland.,Swiss Cancer Center Léman, Lausanne, Switzerland.,Brain Tumor and Immune Cell Engineering Group, Faculty of Medicine, University of Geneva, Geneva, Switzerland.,Department of Oncology, Geneva University Hospitals (HUG), Geneva, Switzerland
| |
Collapse
|
36
|
Park N, Pandey K, Chang SK, Kwon AY, Cho YB, Hur J, Katwal NB, Kim SK, Lee SA, Son GW, Jo JM, Ahn HJ, Moon YW. Preclinical platform for long-term evaluation of immuno-oncology drugs using hCD34+ humanized mouse model. J Immunother Cancer 2020; 8:jitc-2020-001513. [PMID: 33239416 PMCID: PMC7689593 DOI: 10.1136/jitc-2020-001513] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/09/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Well-characterized preclinical models are essential for immune-oncology research. We investigated the feasibility of our humanized mouse model for evaluating the long-term efficacy of immunotherapy and biomarkers. METHODS Humanized mice were generated by injecting human fetal cord blood-derived CD34+ hematopoietic stem cells to NOD-scid IL2rγnull (NSG) mice myeloablated with irradiation or busulfan. The humanization success was defined as a 25% or higher ratio of human CD45+ cells to mice peripheral blood mononuclear cells. RESULTS Busulfan was ultimately selected as the appropriate myeloablative method because it provided a higher success rate of humanization (approximately 80%) and longer survival time (45 weeks). We proved the development of functional T cells by demonstrating the anticancer effect of the programmed cell death-1 (PD-1) inhibitor in our humanized mice but not in non-humanized NSG mice. After confirming the long-lasting humanization state (45 weeks), we further investigated the response durability of the PD-1 inhibitor and biomarkers in our humanized mice. Early increase in serum tumor necrosis factor α levels, late increase in serum interleukin 6 levels and increase in tumor-infiltrating CD8+ T lymphocytes correlated more with a durable response over 60 days than with a non-durable response. CONCLUSIONS Our CD34+ humanized mouse model is the first in vivo platform for testing the long-term efficacy of anticancer immunotherapies and biomarkers, given that none of the preclinical models has ever been evaluated for such a long duration.
Collapse
Affiliation(s)
- Nahee Park
- Hematology and Oncology, Department of Internal Medicine, CHA Bundang Medical Center, Seongnam, South Korea
| | - Kamal Pandey
- Hematology and Oncology, Department of Internal Medicine, CHA Bundang Medical Center, Seongnam, South Korea.,Department of Biomedical Science, CHA Bundang Medical Center, Seongnam, South Korea
| | - Sei Kyung Chang
- Department of Radiation Oncology, CHA Bundang Medical Center, Seongnam, South Korea
| | - Ah-Young Kwon
- Department of Pathology, CHA Bundang Medical Center, Seongnam, South Korea
| | - Young Bin Cho
- Hematology and Oncology, Department of Internal Medicine, CHA Bundang Medical Center, Seongnam, South Korea
| | - Jin Hur
- Hematology and Oncology, Department of Internal Medicine, CHA Bundang Medical Center, Seongnam, South Korea.,Department of Biomedical Science, CHA Bundang Medical Center, Seongnam, South Korea
| | - Nar Bahadur Katwal
- Hematology and Oncology, Department of Internal Medicine, CHA Bundang Medical Center, Seongnam, South Korea.,Department of Biomedical Science, CHA Bundang Medical Center, Seongnam, South Korea
| | - Seung Ki Kim
- Department of Surgery, CHA Bundang Medical Center, Seongnam, South Korea
| | - Seung Ah Lee
- Department of Surgery, CHA Bundang Medical Center, Seongnam, South Korea
| | - Gun Woo Son
- Hematology and Oncology, Department of Internal Medicine, CHA Bundang Medical Center, Seongnam, South Korea
| | - Jong Min Jo
- Hematology and Oncology, Department of Internal Medicine, CHA Bundang Medical Center, Seongnam, South Korea
| | - Hee Jung Ahn
- Department of Pathology, CHA Bundang Medical Center, Seongnam, South Korea
| | - Yong Wha Moon
- Hematology and Oncology, Department of Internal Medicine, CHA Bundang Medical Center, Seongnam, South Korea
| |
Collapse
|