1
|
Ruzzi F, Riccardo F, Conti L, Tarone L, Semprini MS, Bolli E, Barutello G, Quaglino E, Lollini PL, Cavallo F. Cancer vaccines: Target antigens, vaccine platforms and preclinical models. Mol Aspects Med 2025; 101:101324. [PMID: 39631227 DOI: 10.1016/j.mam.2024.101324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 11/27/2024] [Indexed: 12/07/2024]
Abstract
This review provides a comprehensive overview of the evolving landscape of cancer vaccines, highlighting their potential to revolutionize tumor prevention. Building on the success of vaccines against virus-related cancers, such as HPV- and HBV-associated cervical and liver cancers, the current challenge is to extend these achievements to the prevention of non-viral tumors and the treatment of preneoplastic or early neoplastic lesions. This review analyzes the critical aspects of preventive anti-cancer vaccination, focusing on the choice of target antigens, the development of effective vaccine platforms and technologies, and the use of various model systems for preclinical testing, from laboratory rodents to companion animals.
Collapse
Affiliation(s)
- Francesca Ruzzi
- Laboratory of Immunology and Biology of Metastasis, Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40126, Bologna, Italy
| | - Federica Riccardo
- Laboratory of OncoImmunology, Department of Molecular Biotechnology and Health Sciences (DMBSS), University of Torino, 10126, Torino, Italy
| | - Laura Conti
- Laboratory of OncoImmunology, Department of Molecular Biotechnology and Health Sciences (DMBSS), University of Torino, 10126, Torino, Italy
| | - Lidia Tarone
- Laboratory of OncoImmunology, Department of Molecular Biotechnology and Health Sciences (DMBSS), University of Torino, 10126, Torino, Italy
| | - Maria Sofia Semprini
- Laboratory of Immunology and Biology of Metastasis, Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40126, Bologna, Italy
| | - Elisabetta Bolli
- Laboratory of OncoImmunology, Department of Molecular Biotechnology and Health Sciences (DMBSS), University of Torino, 10126, Torino, Italy
| | - Giuseppina Barutello
- Laboratory of OncoImmunology, Department of Molecular Biotechnology and Health Sciences (DMBSS), University of Torino, 10126, Torino, Italy
| | - Elena Quaglino
- Laboratory of OncoImmunology, Department of Molecular Biotechnology and Health Sciences (DMBSS), University of Torino, 10126, Torino, Italy
| | - Pier-Luigi Lollini
- Laboratory of Immunology and Biology of Metastasis, Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40126, Bologna, Italy; IRCCS Azienda Ospedaliera Universitaria di Bologna, 40138, Bologna, Italy.
| | - Federica Cavallo
- Laboratory of OncoImmunology, Department of Molecular Biotechnology and Health Sciences (DMBSS), University of Torino, 10126, Torino, Italy.
| |
Collapse
|
2
|
Chan JD, Scheffler CM, Munoz I, Sek K, Lee JN, Huang YK, Yap KM, Saw NYL, Li J, Chen AXY, Chan CW, Derrick EB, Todd KL, Tong J, Dunbar PA, Li J, Hoang TX, de Menezes MN, Petley EV, Kim JS, Nguyen D, Leung PSK, So J, Deguit C, Zhu J, House IG, Kats LM, Scott AM, Solomon BJ, Harrison SJ, Oliaro J, Parish IA, Quinn KM, Neeson PJ, Slaney CY, Lai J, Beavis PA, Darcy PK. FOXO1 enhances CAR T cell stemness, metabolic fitness and efficacy. Nature 2024; 629:201-210. [PMID: 38600376 PMCID: PMC11062918 DOI: 10.1038/s41586-024-07242-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 02/27/2024] [Indexed: 04/12/2024]
Abstract
Chimeric antigen receptor (CAR) T cell therapy has transformed the treatment of haematological malignancies such as acute lymphoblastic leukaemia, B cell lymphoma and multiple myeloma1-4, but the efficacy of CAR T cell therapy in solid tumours has been limited5. This is owing to a number of factors, including the immunosuppressive tumour microenvironment that gives rise to poorly persisting and metabolically dysfunctional T cells. Analysis of anti-CD19 CAR T cells used clinically has shown that positive treatment outcomes are associated with a more 'stem-like' phenotype and increased mitochondrial mass6-8. We therefore sought to identify transcription factors that could enhance CAR T cell fitness and efficacy against solid tumours. Here we show that overexpression of FOXO1 promotes a stem-like phenotype in CAR T cells derived from either healthy human donors or patients, which correlates with improved mitochondrial fitness, persistence and therapeutic efficacy in vivo. This work thus reveals an engineering approach to genetically enforce a favourable metabolic phenotype that has high translational potential to improve the efficacy of CAR T cells against solid tumours.
Collapse
Affiliation(s)
- Jack D Chan
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
| | - Christina M Scheffler
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
| | - Isabelle Munoz
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
| | - Kevin Sek
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
| | - Joel N Lee
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
| | - Yu-Kuan Huang
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
| | - Kah Min Yap
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
| | - Nicole Y L Saw
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
| | - Jasmine Li
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
| | - Amanda X Y Chen
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
| | - Cheok Weng Chan
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
| | - Emily B Derrick
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
| | - Kirsten L Todd
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
| | - Junming Tong
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
| | - Phoebe A Dunbar
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
| | - Jiawen Li
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
| | - Thang X Hoang
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
| | - Maria N de Menezes
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
| | - Emma V Petley
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
| | - Joelle S Kim
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
| | - Dat Nguyen
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
| | - Patrick S K Leung
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, Australia
| | - Joan So
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
| | - Christian Deguit
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
| | - Joe Zhu
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
| | - Imran G House
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
| | - Lev M Kats
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
| | - Andrew M Scott
- Olivia Newton-John Cancer Research Institute and School of Cancer Medicine, La Trobe University, Melbourne, Victoria, Australia
- Faculty of Medicine, The University of Melbourne, Parkville, Victoria, Australia
| | - Benjamin J Solomon
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
| | - Simon J Harrison
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
- Clinical Haematology and Centre of Excellence for Cellular Immunotherapies, Peter MacCallum Cancer Centre and Royal Melbourne Hospital, Melbourne, Victoria, Australia
| | - Jane Oliaro
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
| | - Ian A Parish
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
| | - Kylie M Quinn
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| | - Paul J Neeson
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
| | - Clare Y Slaney
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
| | - Junyun Lai
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia.
| | - Paul A Beavis
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia.
| | - Phillip K Darcy
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia.
- Clinical Haematology and Centre of Excellence for Cellular Immunotherapies, Peter MacCallum Cancer Centre and Royal Melbourne Hospital, Melbourne, Victoria, Australia.
- Department of Immunology, Monash University, Clayton, Victoria, Australia.
| |
Collapse
|
3
|
Trotter TN, Wilson A, McBane J, Dagotto CE, Yang XY, Wei JP, Lei G, Thrash H, Snyder JC, Lyerly HK, Hartman ZC. Overcoming Xenoantigen Immunity to Enable Cellular Tracking and Gene Regulation with Immune-competent "NoGlow" Mice. CANCER RESEARCH COMMUNICATIONS 2024; 4:1050-1062. [PMID: 38592453 PMCID: PMC11003454 DOI: 10.1158/2767-9764.crc-24-0062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 03/26/2024] [Accepted: 04/02/2024] [Indexed: 04/10/2024]
Abstract
The ability to temporally regulate gene expression and track labeled cells makes animal models powerful biomedical tools. However, sudden expression of xenobiotic genes [e.g., GFP, luciferase (Luc), or rtTA3] can trigger inadvertent immunity that suppresses foreign protein expression or results in complete rejection of transplanted cells. Germline exposure to foreign antigens somewhat addresses these challenges; however, native fluorescence and bioluminescence abrogates the utility of reporter proteins and highly spatiotemporally restricted expression can lead to suboptimal xenoantigen tolerance. To overcome these unwanted immune responses and enable reliable cell tracking/gene regulation, we developed a novel mouse model that selectively expresses antigen-intact but nonfunctional forms of GFP and Luc, as well as rtTA3, after CRE-mediated recombination. Using tissue-specific CREs, we observed model and sex-based differences in immune tolerance to the encoded xenoantigens, illustrating the obstacles of tolerizing animals to foreign genes and validating the utility of these "NoGlow" mice to dissect mechanisms of central and peripheral tolerance. Critically, tissue unrestricted NoGlow mice possess no detectable background fluorescence or luminescence and exhibit limited adaptive immunity against encoded transgenic xenoantigens after vaccination. Moreover, we demonstrate that NoGlow mice allow tracking and tetracycline-inducible gene regulation of triple-transgenic cells expressing GFP/Luc/rtTA3, in contrast to transgene-negative immune-competent mice that eliminate these cells or prohibit metastatic seeding. Notably, this model enables de novo metastasis from orthotopically implanted, triple-transgenic tumor cells, despite high xenoantigen expression. Altogether, the NoGlow model provides a critical resource for in vivo studies across disciplines, including oncology, developmental biology, infectious disease, autoimmunity, and transplantation. SIGNIFICANCE Multitolerant NoGlow mice enable tracking and gene manipulation of transplanted tumor cells without immune-mediated rejection, thus providing a platform to investigate novel mechanisms of adaptive immunity related to metastasis, immunotherapy, and tolerance.
Collapse
Affiliation(s)
| | - Andrea Wilson
- Department of Pathology, Duke University, Durham, North Carolina
| | - Jason McBane
- Department of Surgery, Duke University, Durham, North Carolina
| | | | - Xiao-Yi Yang
- Department of Surgery, Duke University, Durham, North Carolina
| | - Jun-Ping Wei
- Department of Surgery, Duke University, Durham, North Carolina
| | - Gangjun Lei
- Department of Surgery, Duke University, Durham, North Carolina
| | - Hannah Thrash
- Department of Pharmacology and Cancer Biology, Duke University, Durham, North Carolina
| | - Joshua C. Snyder
- Department of Surgery, Duke University, Durham, North Carolina
- Department of Cell Biology, Duke University, Durham, North Carolina
| | - Herbert Kim Lyerly
- Department of Surgery, Duke University, Durham, North Carolina
- Department of Pathology, Duke University, Durham, North Carolina
- Department of Integrative Immunobiology, Duke University, Durham, North Carolina
| | - Zachary C. Hartman
- Department of Surgery, Duke University, Durham, North Carolina
- Department of Pathology, Duke University, Durham, North Carolina
- Department of Integrative Immunobiology, Duke University, Durham, North Carolina
| |
Collapse
|
4
|
House IG, Derrick EB, Sek K, Chen AXY, Li J, Lai J, Todd KL, Munoz I, Michie J, Chan CW, Huang YK, Chan JD, Petley EV, Tong J, Nguyen D, Engel S, Savas P, Hogg SJ, Vervoort SJ, Kearney CJ, Burr ML, Lam EYN, Gilan O, Bedoui S, Johnstone RW, Dawson MA, Loi S, Darcy PK, Beavis PA. CRISPR-Cas9 screening identifies an IRF1-SOCS1-mediated negative feedback loop that limits CXCL9 expression and antitumor immunity. Cell Rep 2023; 42:113014. [PMID: 37605534 DOI: 10.1016/j.celrep.2023.113014] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 06/13/2023] [Accepted: 08/07/2023] [Indexed: 08/23/2023] Open
Abstract
CXCL9 expression is a strong predictor of response to immune checkpoint blockade therapy. Accordingly, we sought to develop therapeutic strategies to enhance the expression of CXCL9 and augment antitumor immunity. To perform whole-genome CRISPR-Cas9 screening for regulators of CXCL9 expression, a CXCL9-GFP reporter line is generated using a CRISPR knockin strategy. This approach finds that IRF1 limits CXCL9 expression in both tumor cells and primary myeloid cells through induction of SOCS1, which subsequently limits STAT1 signaling. Thus, we identify a subset of STAT1-dependent genes that do not require IRF1 for their transcription, including CXCL9. Targeting of either IRF1 or SOCS1 potently enhances CXCL9 expression by intratumoral macrophages, which is further enhanced in the context of immune checkpoint blockade therapy. We hence show a non-canonical role for IRF1 in limiting the expression of a subset of STAT1-dependent genes through induction of SOCS1.
Collapse
Affiliation(s)
- Imran G House
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia.
| | - Emily B Derrick
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Kevin Sek
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Amanda X Y Chen
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Jasmine Li
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Junyun Lai
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Kirsten L Todd
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Isabelle Munoz
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Jessica Michie
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Cheok Weng Chan
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Yu-Kuan Huang
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Jack D Chan
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Emma V Petley
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Junming Tong
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - DatMinh Nguyen
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Sven Engel
- Department of Microbiology and Immunology at the Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC, Australia; Institute of Experimental Immunology, University of Bonn, Bonn, Germany
| | - Peter Savas
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia; Division of Research, Peter MacCallum Cancer Centre, University of Melbourne, Melbourne, VIC, Australia
| | - Simon J Hogg
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia; Division of Research, Peter MacCallum Cancer Centre, University of Melbourne, Melbourne, VIC, Australia
| | - Stephin J Vervoort
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia; Division of Research, Peter MacCallum Cancer Centre, University of Melbourne, Melbourne, VIC, Australia
| | - Conor J Kearney
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, 3084, Australia; School of Cancer Medicine, La Trobe University, Melbourne, VIC, 3086, Australia
| | - Marian L Burr
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia; ACRF Department of Cancer Biology and Therapeutics, The John Curtin School of Medical Research, The Australian National University, Canberra, ACT 2601, Australia; Department of Anatomical Pathology, The Royal Melbourne Hospital, Melbourne, VIC 3050, Australia
| | - Enid Y N Lam
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia; Division of Research, Peter MacCallum Cancer Centre, University of Melbourne, Melbourne, VIC, Australia
| | - Omer Gilan
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia; Division of Research, Peter MacCallum Cancer Centre, University of Melbourne, Melbourne, VIC, Australia
| | - Sammy Bedoui
- Department of Microbiology and Immunology at the Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC, Australia; Institute of Experimental Immunology, University of Bonn, Bonn, Germany
| | - Ricky W Johnstone
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia; Division of Research, Peter MacCallum Cancer Centre, University of Melbourne, Melbourne, VIC, Australia
| | - Mark A Dawson
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia; Division of Research, Peter MacCallum Cancer Centre, University of Melbourne, Melbourne, VIC, Australia; Department of Haematology, Peter MacCallum Cancer Centre and The Royal Melbourne Hospital, Melbourne, VIC 3052, Australia; Centre for Cancer Research, The University of Melbourne, Melbourne, VIC 3000, Australia
| | - Sherene Loi
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia; Division of Research, Peter MacCallum Cancer Centre, University of Melbourne, Melbourne, VIC, Australia
| | - Phillip K Darcy
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia; Department of Immunology, Monash University, Clayton, VIC, Australia.
| | - Paul A Beavis
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia.
| |
Collapse
|
5
|
Jacob JB, Wei KC, Bepler G, Reyes JD, Cani A, Polin L, White K, Kim S, Viola N, McGrath J, Guastella A, Yin C, Mi QS, Kidder BL, Wagner KU, Ratner S, Phillips V, Xiu J, Parajuli P, Wei WZ. Identification of actionable targets for breast cancer intervention using a diversity outbred mouse model. iScience 2023; 26:106320. [PMID: 36968078 PMCID: PMC10034465 DOI: 10.1016/j.isci.2023.106320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 01/16/2023] [Accepted: 02/26/2023] [Indexed: 03/06/2023] Open
Abstract
HER2-targeted therapy has improved breast cancer survival, but treatment resistance and disease prevention remain major challenges. Genes that enable HER2/Neu oncogenesis are the next intervention targets. A bioinformatics discovery platform of HER2/Neu-expressing Diversity Outbred (DO) F1 Mice was established to identify cancer-enabling genes. Quantitative Trait Loci (QTL) associated with onset ages and growth rates of spontaneous mammary tumors were sought. Twenty-six genes in 3 QTL contain sequence variations unique to the genetic backgrounds that are linked to aggressive tumors and 21 genes are associated with human breast cancer survival. Concurrent identification of TSC22D3, a transcription factor, and its target gene LILRB4, a myeloid cell checkpoint receptor, suggests an immune axis for regulation, or intervention, of disease. We also investigated TIEG1 gene that impedes tumor immunity but suppresses tumor growth. Although not an actionable target, TIEG1 study revealed genetic regulation of tumor progression, forming the basis of the genetics-based discovery platform.
Collapse
Affiliation(s)
- Jennifer B. Jacob
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, 48201, USA
| | - Kuang-Chung Wei
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, 48201, USA
| | - Gerold Bepler
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, 48201, USA
| | - Joyce D. Reyes
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, 48201, USA
| | - Andi Cani
- Department of Internal Medicine, Rogel Cancer Center, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Lisa Polin
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, 48201, USA
| | - Kathryn White
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, 48201, USA
| | - Seongho Kim
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, 48201, USA
| | - Nerissa Viola
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, 48201, USA
| | - Julie McGrath
- Clinical and Translational Research, Caris Life Sciences, Irving, TX75039, USA
| | - Anthony Guastella
- Clinical and Translational Research, Caris Life Sciences, Irving, TX75039, USA
| | - CongCong Yin
- Department of Immunology, Henry Ford Health System, Detroit, MI48202, USA
| | - Qing-Shen Mi
- Department of Immunology, Henry Ford Health System, Detroit, MI48202, USA
| | - Benjamin L. Kidder
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, 48201, USA
| | - Kay-Uwe Wagner
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, 48201, USA
| | - Stuart Ratner
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, 48201, USA
| | - Victoria Phillips
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, 48201, USA
| | - Joanne Xiu
- Clinical and Translational Research, Caris Life Sciences, Irving, TX75039, USA
| | - Prahlad Parajuli
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, 48201, USA
| | - Wei-Zen Wei
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, 48201, USA
| |
Collapse
|
6
|
Kaneko Y, Yamatsugu K, Yamashita T, Takahashi K, Tanaka T, Aki S, Tatsumi T, Kawamura T, Miura M, Ishii M, Ohkubo K, Osawa T, Kodama T, Ishikawa S, Tsukagoshi M, Chansler M, Sugiyama A, Kanai M, Katoh H. Pathological complete remission of relapsed tumor by photo-activating antibody-mimetic drug conjugate treatment. Cancer Sci 2022; 113:4350-4362. [PMID: 36121618 DOI: 10.1111/cas.15565] [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: 03/28/2022] [Revised: 08/12/2022] [Accepted: 08/23/2022] [Indexed: 02/03/2023] Open
Abstract
Antibody-mimetic drug conjugate is a novel noncovalent conjugate consisting of an antibody-mimetic recognizing a target molecule on the cancer cell surface and low-molecular-weight payloads that kill the cancer cells. In this study, the efficacy of a photo-activating antibody-mimetic drug conjugate targeting HER2-expressing tumors was evaluated in mice, by using the affibody that recognize HER2 (ZHER2:342 ) as a target molecule and an axially substituted silicon phthalocyanine (a novel potent photo-activating compound) as a payload. The first treatment with the photo-activating antibody-mimetic drug conjugates reduced the size of all HER2-expressing KPL-4 xenograft tumors macroscopically. However, during the observation period, relapsed tumors gradually appeared in approximately 50% of the animals. To evaluate the efficacy of repeated antibody-mimetic drug conjugate treatment, animals with relapsed tumors were treated again with the same regimen. After the second observation period, the mouse tissues were examined histopathologically. Unexpectedly, all relapsed tumors were eradicated, and all animals were diagnosed with pathological complete remission. After the second treatment, skin wounds healed rapidly, and no significant side effects were observed in other organs, except for occasional microscopic granulomatous tissues beneath the serosa of the liver in a few mice. Repeated treatments seemed to be well tolerated. These results indicate the promising efficacy of the repeated photo-activating antibody-mimetic drug conjugate treatment against HER2-expressing tumors.
Collapse
Affiliation(s)
- Yudai Kaneko
- Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan.,Medical & Biological Laboratories Co., Ltd, Tokyo, Japan
| | - Kenzo Yamatsugu
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Takefumi Yamashita
- Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Kazuki Takahashi
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Toshiya Tanaka
- Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Sho Aki
- Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Toshifumi Tatsumi
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Takeshi Kawamura
- Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan.,Isotope Science Center, The University of Tokyo, Tokyo, Japan
| | - Mai Miura
- Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Masazumi Ishii
- Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Kei Ohkubo
- Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Osaka, Japan.,Institute for Advanced Co-Creation Studies, Osaka University, Osaka, Japan
| | - Tsuyoshi Osawa
- Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Tatsuhiko Kodama
- Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Shumpei Ishikawa
- Department of Preventive Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | | | | | - Akira Sugiyama
- Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan.,Isotope Science Center, The University of Tokyo, Tokyo, Japan
| | - Motomu Kanai
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Hiroto Katoh
- Department of Preventive Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
7
|
Bai B, Wang H, Li Y, de Haan K, Colonnese F, Wan Y, Zuo J, Doan NB, Zhang X, Zhang Y, Li J, Yang X, Dong W, Darrow MA, Kamangar E, Lee HS, Rivenson Y, Ozcan A. Label-Free Virtual HER2 Immunohistochemical Staining of Breast Tissue using Deep Learning. BME FRONTIERS 2022; 2022:9786242. [PMID: 37850170 PMCID: PMC10521710 DOI: 10.34133/2022/9786242] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 08/25/2022] [Indexed: 10/19/2023] Open
Abstract
The immunohistochemical (IHC) staining of the human epidermal growth factor receptor 2 (HER2) biomarker is widely practiced in breast tissue analysis, preclinical studies, and diagnostic decisions, guiding cancer treatment and investigation of pathogenesis. HER2 staining demands laborious tissue treatment and chemical processing performed by a histotechnologist, which typically takes one day to prepare in a laboratory, increasing analysis time and associated costs. Here, we describe a deep learning-based virtual HER2 IHC staining method using a conditional generative adversarial network that is trained to rapidly transform autofluorescence microscopic images of unlabeled/label-free breast tissue sections into bright-field equivalent microscopic images, matching the standard HER2 IHC staining that is chemically performed on the same tissue sections. The efficacy of this virtual HER2 staining framework was demonstrated by quantitative analysis, in which three board-certified breast pathologists blindly graded the HER2 scores of virtually stained and immunohistochemically stained HER2 whole slide images (WSIs) to reveal that the HER2 scores determined by inspecting virtual IHC images are as accurate as their immunohistochemically stained counterparts. A second quantitative blinded study performed by the same diagnosticians further revealed that the virtually stained HER2 images exhibit a comparable staining quality in the level of nuclear detail, membrane clearness, and absence of staining artifacts with respect to their immunohistochemically stained counterparts. This virtual HER2 staining framework bypasses the costly, laborious, and time-consuming IHC staining procedures in laboratory and can be extended to other types of biomarkers to accelerate the IHC tissue staining used in life sciences and biomedical workflow.
Collapse
Affiliation(s)
- Bijie Bai
- Electrical and Computer Engineering Department, University of California, Los Angeles, CA 90095, USA
- Bioengineering Department, University of California, Los Angeles 90095, USA
- California NanoSystems Institute (CNSI), University of California, Los Angeles, CA, USA
| | - Hongda Wang
- Electrical and Computer Engineering Department, University of California, Los Angeles, CA 90095, USA
- Bioengineering Department, University of California, Los Angeles 90095, USA
- California NanoSystems Institute (CNSI), University of California, Los Angeles, CA, USA
| | - Yuzhu Li
- Electrical and Computer Engineering Department, University of California, Los Angeles, CA 90095, USA
- Bioengineering Department, University of California, Los Angeles 90095, USA
- California NanoSystems Institute (CNSI), University of California, Los Angeles, CA, USA
| | - Kevin de Haan
- Electrical and Computer Engineering Department, University of California, Los Angeles, CA 90095, USA
- Bioengineering Department, University of California, Los Angeles 90095, USA
- California NanoSystems Institute (CNSI), University of California, Los Angeles, CA, USA
| | | | - Yujie Wan
- Physics and Astronomy Department, University of California, Los Angeles, CA 90095, USA
| | - Jingyi Zuo
- Computer Science Department, University of California, Los Angeles, CA, USA
| | - Ngan B. Doan
- Translational Pathology Core Laboratory, University of California, Los Angeles, CA 90095, USA
| | - Xiaoran Zhang
- Electrical and Computer Engineering Department, University of California, Los Angeles, CA 90095, USA
| | - Yijie Zhang
- Electrical and Computer Engineering Department, University of California, Los Angeles, CA 90095, USA
- Bioengineering Department, University of California, Los Angeles 90095, USA
- California NanoSystems Institute (CNSI), University of California, Los Angeles, CA, USA
| | - Jingxi Li
- Electrical and Computer Engineering Department, University of California, Los Angeles, CA 90095, USA
- Bioengineering Department, University of California, Los Angeles 90095, USA
- California NanoSystems Institute (CNSI), University of California, Los Angeles, CA, USA
| | - Xilin Yang
- Electrical and Computer Engineering Department, University of California, Los Angeles, CA 90095, USA
- Bioengineering Department, University of California, Los Angeles 90095, USA
- California NanoSystems Institute (CNSI), University of California, Los Angeles, CA, USA
| | - Wenjie Dong
- Statistics Department, University of California, Los Angeles, CA 90095, USA
| | - Morgan Angus Darrow
- Department of Pathology and Laboratory Medicine, University of California at Davis, Sacramento, CA 95817, USA
| | - Elham Kamangar
- Department of Pathology and Laboratory Medicine, University of California at Davis, Sacramento, CA 95817, USA
| | - Han Sung Lee
- Department of Pathology and Laboratory Medicine, University of California at Davis, Sacramento, CA 95817, USA
| | - Yair Rivenson
- Electrical and Computer Engineering Department, University of California, Los Angeles, CA 90095, USA
- Bioengineering Department, University of California, Los Angeles 90095, USA
- California NanoSystems Institute (CNSI), University of California, Los Angeles, CA, USA
| | - Aydogan Ozcan
- Electrical and Computer Engineering Department, University of California, Los Angeles, CA 90095, USA
- Bioengineering Department, University of California, Los Angeles 90095, USA
- California NanoSystems Institute (CNSI), University of California, Los Angeles, CA, USA
- Department of Surgery, University of California, Los Angeles, CA 90095, USA
| |
Collapse
|
8
|
Prevention and Therapy of Metastatic HER-2 + Mammary Carcinoma with a Human Candidate HER-2 Virus-like Particle Vaccine. Biomedicines 2022; 10:biomedicines10102654. [PMID: 36289916 PMCID: PMC9599132 DOI: 10.3390/biomedicines10102654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 10/17/2022] [Accepted: 10/17/2022] [Indexed: 11/16/2022] Open
Abstract
Vaccines are a promising therapeutic alternative to monoclonal antibodies against HER-2+ breast cancer. We present the preclinical activity of an ES2B-C001, a VLP-based vaccine being developed for human breast cancer therapy. FVB mice challenged with HER-2+ mammary carcinoma cells QD developed progressive tumors, whereas all mice vaccinated with ES2B-C001+Montanide ISA 51, and 70% of mice vaccinated without adjuvant, remained tumor-free. ES2B-C001 completely inhibited lung metastases in mice challenged intravenously. HER-2 transgenic Delta16 mice developed mammary carcinomas by 4−8 months of age; two administrations of ES2B-C001+Montanide prevented tumor onset for >1 year. Young Delta16 mice challenged intravenously with QD cells developed a mean of 68 lung nodules in 13 weeks, whereas all mice vaccinated with ES2B-C001+Montanide, and 73% of mice vaccinated without adjuvant, remained metastasis-free. ES2B-C001 in adjuvant elicited strong anti-HER-2 antibody responses comprising all Ig isotypes; titers ranging from 1−10 mg/mL persisted for many months. Antibodies inhibited the 3D growth of human HER-2+ trastuzumab-sensitive and -resistant breast cancer cells. Vaccination did not induce cytokine storms; however, it increased the ELISpot frequency of IFN-γ secreting HER-2-specific splenocytes. ES2B-C001 is a promising candidate vaccine for the therapy of tumors expressing HER-2. Preclinical results warrant further development towards human clinical studies.
Collapse
|
9
|
Nguyen-Hoai T, Hohn O, Pezzutto A, Westermann J. Gene Gun Her2/neu DNA Vaccination: Evaluation of Vaccine Efficacy in a Syngeneic Her2/neu Mouse Tumor Model. METHODS IN MOLECULAR BIOLOGY (CLIFTON, N.J.) 2022; 2521:129-154. [PMID: 35732996 DOI: 10.1007/978-1-0716-2441-8_7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
Genetic vaccination using naked plasmid DNA is an immunization strategy both against infectious diseases and cancer.In order to improve efficacy of DNA vaccines, particularly in large animals and humans, different strategies have been pursued. These vaccination strategies are based on different application routes, schedules and coexpression of immunomodulatory molecules as adjuvants. Our mouse tumor model offers the possibility to investigate Her2/neu DNA vaccines in different settings, that is, intramuscular or intradermal application with or without coexpression of adjuvants. The immunogenicity of predicted peptides for Her2/neu specific memory T cells were screened and confirmed after intramuscular and intradermal application. Protection from tumor growth in tumor challenge experiments and both T cell and humoral immune responses against Her2/neu peptides are used as surrogate parameters for vaccine efficacy.
Collapse
Affiliation(s)
- Tam Nguyen-Hoai
- Experimental and Clinical Research Center, Charité - Universitätsmedizin Berlin, Berlin, Germany.
| | | | - Antonio Pezzutto
- Charité - Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
| | - Jörg Westermann
- Department Hematology, Oncology and Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
10
|
Abe S, Nagata H, Crosby EJ, Inoue Y, Kaneko K, Liu CX, Yang X, Wang T, Acharya CR, Agarwal P, Snyder J, Gwin W, Morse MA, Zhong P, Lyerly HK, Osada T. Combination of ultrasound-based mechanical disruption of tumor with immune checkpoint blockade modifies tumor microenvironment and augments systemic antitumor immunity. J Immunother Cancer 2022; 10:jitc-2021-003717. [PMID: 35039461 PMCID: PMC8765068 DOI: 10.1136/jitc-2021-003717] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/13/2021] [Indexed: 02/02/2023] Open
Abstract
Background Despite multimodal adjuvant management with radiotherapy, chemotherapy and hormonal therapies, most surgically resected primary breast cancers relapse or metastasize. A potential solution to late and distant recurrence is to augment systemic antitumor immunity, in part by appropriately presenting tumor antigens, but also by modulating the immunosuppressive tumor microenvironment (TME). We previously validated this concept in models of murine carcinoma treated with a novel predominately microcavitating version of high-intensity focused ultrasound (HIFU), mechanical high-intensity focused ultrasound (M-HIFU). Here we elucidated the mechanisms of enhanced antitumor immunity by M-HIFU over conventional thermal high-intensity focused ultrasound (T-HIFU) and investigated the potential of the combinatorial strategy with an immune checkpoint inhibitor, anti-PD-L1 antibody. Methods The antitumor efficacy of treatments was investigated in syngeneic murine breast cancer models using triple-negative (E0771) or human ErbB-2 (HER2) expressing (MM3MG-HER2) tumors in C57BL/6 or BALB/c mice, respectively. Induction of systemic antitumor immunity by the treatments was tested using bilateral tumor implantation models. Flow cytometry, immunohistochemistry, and single-cell RNA sequencing were performed to elucidate detailed effects of HIFU treatments or combination treatment on TME, including the activation status of CD8 T cells and polarization of tumor-associated macrophages (TAMs). Results More potent systemic antitumor immunity and tumor growth suppression were induced by M-HIFU compared with T-HIFU. Molecular characterization of the TME after M-HIFU by single-cell RNA sequencing demonstrated repolarization of TAM to the immunostimulatory M1 subtype compared with TME post-T-HIFU. Concurrent anti-PD-L1 antibody administration or depletion of CD4+ T cells containing a population of regulatory T cells markedly increased T cell-mediated antitumor immunity and tumor growth suppression at distant, untreated tumor sites in M-HIFU treated mice compared with M-HIFU monotherapy. CD8 T and natural killer cells played major roles as effector cells in the combination treatment. Conclusions Physical disruption of the TME by M-HIFU repolarizes TAM, enhances T-cell infiltration, and, when combined with anti-PD-L1 antibody, mediates superior systemic antitumor immune responses and distant tumor growth suppression. These findings suggest M-HIFU combined with anti-PD-L1 may be useful in reducing late recurrence or metastasis when applied to primary tumors.
Collapse
Affiliation(s)
- Shinya Abe
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA.,Department of Surgical Oncology, Faculty of Medicine, The University of Tokyo Graduate School of Medicine, Bunkyo-ku, Tokyo, Japan
| | - Hiroshi Nagata
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA.,Department of Surgical Oncology, Faculty of Medicine, The University of Tokyo Graduate School of Medicine, Bunkyo-ku, Tokyo, Japan
| | - Erika J Crosby
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA
| | - Yoshiyuki Inoue
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA.,Department of Surgery, Jichi Medical University, Shimotsuke, Tochigi, Japan
| | - Kensuke Kaneko
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA.,Department of Surgical Oncology, Faculty of Medicine, The University of Tokyo Graduate School of Medicine, Bunkyo-ku, Tokyo, Japan
| | - Cong-Xiao Liu
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA
| | - Xiao Yang
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA
| | - Tao Wang
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA
| | - Chaitanya R Acharya
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA
| | - Pankaj Agarwal
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA
| | - Joshua Snyder
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA
| | - William Gwin
- Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Michael A Morse
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA.,Department of Medicine, Duke University Medical Center, Durham, North Carolina, USA
| | - Pei Zhong
- Department of Mechanical Engineering and Materials Science, Duke University, Durham, North Carolina, USA
| | - Herbert Kim Lyerly
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA
| | - Takuya Osada
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA
| |
Collapse
|
11
|
Targeting PTPN22 does not enhance the efficacy of CAR T cells in solid tumours. Mol Cell Biol 2022; 42:e0044921. [PMID: 35041491 DOI: 10.1128/mcb.00449-21] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Adoptive cell therapy with chimeric antigen receptor (CAR) T cells has revolutionised the treatment of certain B cell malignancies, but has been in ineffective against solid tumours. Recent studies have highlighted the potential of targeting negative regulators of T cell signalling to enhance the efficacy and extend the utility of CAR T cells to solid tumours. Autoimmunity-linked protein tyrosine phosphatase N22 (PTPN22) has been proposed as a target for cancer immunotherapy. Here we have used CRISPR/Cas9 gene-editing to generate PTPN22-deficient (Ptpn22Δ/Δ) mice (C57BL/6) and assessed the impact of PTPN22 deficiency on the cytotoxicity and efficacy of CAR T cells in vitro and in vivo. As reported previously, PTPN22 deficiency was accompanied by the promotion of effector T cell responses ex vivo and the repression of syngeneic tumour growth in vivo. However, PTPN22-deficiency did not enhance the cytotoxic activity of murine CAR T cells targeting the extracellular domain of the human oncoprotein HER2 in vitro. Moreover, PTPN22-deficient α-HER2 CAR T cells or ovalbumin-specific OT-I CD8+ T cells adoptively transferred into mice bearing HER2+ mammary tumours or ovalbumin-expressing mammary or colorectal tumours respectively were no more effective than their wild type counterparts in suppressing tumour growth. The deletion of PTPN22 using CRISPR/Cas9 gene-editing also did not affect the cytotoxic activity of human CAR T cells targeting the Lewis Y antigen that is expressed by many human solid tumours. Therefore, PTPN22-deficiency does not enhance the anti-tumour activity of CAR T cells in solid organ malignancies.
Collapse
|
12
|
Wiede F, Lu KH, Du X, Zeissig MN, Xu R, Goh PK, Xirouchaki CE, Hogarth SJ, Greatorex S, Sek K, Daly RJ, Beavis PA, Darcy PK, Tonks NK, Tiganis T. PTP1B is an intracellular checkpoint that limits T cell and CAR T cell anti-tumor immunity. Cancer Discov 2021; 12:752-773. [PMID: 34794959 DOI: 10.1158/2159-8290.cd-21-0694] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 10/01/2021] [Accepted: 11/15/2021] [Indexed: 11/16/2022]
Abstract
Immunotherapies aimed at alleviating the inhibitory constraints on Tcells have revolutionised cancer management. To date, these have focused on the blockade of cell surface checkpoints such as PD-1. Herein we identify protein-tyrosine-phosphatase-1B (PTP1B) as an intracellular checkpoint that is upregulated in T cells in tumors. We show that the increased PTP1B limits T cell expansion and cytotoxicity to contribute to tumor growth. T cell-specific PTP1B deletion increased STAT-5 signaling and this enhanced the antigen-induced expansion and cytotoxicity of CD8+ T cells to suppress tumor growth. The pharmacological inhibition of PTP1B recapitulated the T cell-mediated repression of tumor growth and enhanced the response to PD-1 blockade. Furthermore, the deletion or inhibition of PTP1B enhanced the efficacy of adoptively-transferred chimeric-antigen-receptor (CAR) T cells against solid tumors. Our findings identify PTP1B as an intracellular checkpoint whose inhibition can alleviate the inhibitory constraints on T cells and CAR T cells to combat cancer.
Collapse
Affiliation(s)
- Florian Wiede
- Biochemistry and Molecular Biology, Monash University
| | - Kun-Hui Lu
- Cancer Research, Peter MacCallum Cancer Centre
| | - Xin Du
- Peter MacCallum Cancer Centre
| | | | | | - Pei Kee Goh
- Biochemistry and Molecular Biology, Monash University
| | | | | | | | - Kevin Sek
- Cancer Immunology Program, Peter MacCallum Cancer Research Centre
| | - Roger J Daly
- Cancer Program, Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University
| | - Paul A Beavis
- Cancer Immunology Program, Peter MacCallum Cancer Research Centre
| | | | | | - Tony Tiganis
- Biochemistry and Molecular Biology, Monash University
| |
Collapse
|
13
|
Preclinical models and technologies to advance nanovaccine development. Adv Drug Deliv Rev 2021; 172:148-182. [PMID: 33711401 DOI: 10.1016/j.addr.2021.03.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 02/26/2021] [Accepted: 03/01/2021] [Indexed: 12/13/2022]
Abstract
The remarkable success of targeted immunotherapies is revolutionizing cancer treatment. However, tumor heterogeneity and low immunogenicity, in addition to several tumor-associated immunosuppression mechanisms are among the major factors that have precluded the success of cancer vaccines as targeted cancer immunotherapies. The exciting outcomes obtained in patients upon the injection of tumor-specific antigens and adjuvants intratumorally, reinvigorated interest in the use of nanotechnology to foster the delivery of vaccines to address cancer unmet needs. Thus, bridging nano-based vaccine platform development and predicted clinical outcomes the selection of the proper preclinical model will be fundamental. Preclinical models have revealed promising outcomes for cancer vaccines. However, only few cases were associated with clinical responses. This review addresses the major challenges related to the translation of cancer nano-based vaccines to the clinic, discussing the requirements for ex vivo and in vivo models of cancer to ensure the translation of preclinical success to patients.
Collapse
|
14
|
Tissues and Tumor Microenvironment (TME) in 3D: Models to Shed Light on Immunosuppression in Cancer. Cells 2021; 10:cells10040831. [PMID: 33917037 PMCID: PMC8067689 DOI: 10.3390/cells10040831] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 03/28/2021] [Accepted: 04/02/2021] [Indexed: 12/26/2022] Open
Abstract
Immunosuppression in cancer has emerged as a major hurdle to immunotherapy efforts. Immunosuppression can arise from oncogene-induced signaling within the tumor as well as from tumor-associated immune cells. Understanding various mechanisms by which the tumor can undermine and evade therapy is critical in improving current cancer immunotherapies. While mouse models have allowed for the characterization of key immune cell types and their role in tumor development, extrapolating these mechanisms to patients has been challenging. There is need for better models to unravel the effects of genetic alterations inherent in tumor cells and immune cells isolated from tumors on tumor growth and to investigate the feasibility of immunotherapy. Three-dimensional (3D) organoid model systems have developed rapidly over the past few years and allow for incorporation of components of the tumor microenvironment such as immune cells and the stroma. This bears great promise for derivation of patient-specific models in a dish for understanding and determining the impact on personalized immunotherapy. In this review, we will highlight the significance of current experimental models employed in the study of tumor immunosuppression and evaluate current tumor organoid-immune cell co-culture systems and their potential impact in shedding light on cancer immunosuppression.
Collapse
|
15
|
Chung WP, Huang WL, Liao WA, Huang WL, Liu YY, Su WC. Development of the CK-MB-1 trastuzumab-resistant HER2-positive breast cancer cell line and xenograft animal models. Cancer Med 2021; 10:2370-2379. [PMID: 33665980 PMCID: PMC7982635 DOI: 10.1002/cam4.3824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 10/28/2020] [Accepted: 02/19/2021] [Indexed: 11/10/2022] Open
Abstract
Background Patients with human epidermal growth factor receptor 2 (HER2)‐positive breast cancer who fail to respond to anti‐HER2 treatments have poor prognoses. Most trastuzumab‐resistant breast cancer cell lines available from biobanks feature either phosphoinositide‐3‐kinase, catalytic, alpha (PIK3CA) mutation or the loss of phosphatase and tensin homolog (PTEN). However, PIK3CA mutations and/or PTEN loss do not account for most trastuzumab‐resistant tumors in humans. Methods Breast cancer cells were collected from one patient's malignant ascites. These cells were cultured and maintained to develop a stable cell line, which we named CK‐MB‐1. We used western blotting to evaluate protein expression. The PIK3CA status of CK‐MB‐1 cells was analyzed using Sanger sequencing and validated using next‐generation sequencing. In vivo, CK‐MB‐1 xenograft tumor models were developed in zebrafish and immunodeficient mice. Results CK‐MB‐1 cells maintained the major characteristics of the parental tumor including HER2 positivity and estrogen receptor negativity. The HER2 gene amplification of CK‐MB‐1 cells was detected by fluorescence in situ hybridization. The integrity of PTEN was confirmed by its positive protein expression and the absence of gene mutations. No common PIK3CA mutation was detected. Compared with the findings in two other HER2‐positive trastuzumab‐resistant cell lines, CK‐MB‐1 cells exhibited greater resistance to trastuzumab, chemotherapeutics, and small‐molecule drugs. Trastuzumab resistance in CK‐MB‐1 cells was confirmed in vivo using the NOD SCID mouse model. Conclusions CK‐MB‐1 cells represent a stable HER2‐positive trastuzumab‐resistant breast cancer cell line. The resistance of CK‐MB‐1 cells does not originate from the PTEN or phosphoinositide 3‐kinase signaling pathway, which can provide an alternative approach for potential drugs.
Collapse
Affiliation(s)
- Wei-Pang Chung
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Department of Oncology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Center of Applied Nanomedicine, National Cheng Kung University, Tainan, Taiwan
| | - Wei-Lun Huang
- Center of Applied Nanomedicine, National Cheng Kung University, Tainan, Taiwan
| | - Wei-An Liao
- Department of Pathology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Wan-Ling Huang
- Center of Applied Nanomedicine, National Cheng Kung University, Tainan, Taiwan
| | - You-Yu Liu
- Center of Applied Nanomedicine, National Cheng Kung University, Tainan, Taiwan
| | - Wu-Chou Su
- Department of Oncology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Center of Applied Nanomedicine, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
16
|
Li Y, Sun Y, Kulke M, Hechler T, Van der Jeught K, Dong T, He B, Miller KD, Radovich M, Schneider BP, Pahl A, Zhang X, Lu X. Targeted immunotherapy for HER2-low breast cancer with 17p loss. Sci Transl Med 2021; 13:eabc6894. [PMID: 33568521 PMCID: PMC8351376 DOI: 10.1126/scitranslmed.abc6894] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 01/13/2021] [Indexed: 12/21/2022]
Abstract
The clinical challenge for treating HER2 (human epidermal growth factor receptor 2)-low breast cancer is the paucity of actionable drug targets. HER2-targeted therapy often has poor clinical efficacy for this disease due to the low level of HER2 protein on the cancer cell surface. We analyzed breast cancer genomics in the search for potential drug targets. Heterozygous loss of chromosome 17p is one of the most frequent genomic events in breast cancer, and 17p loss involves a massive deletion of genes including the tumor suppressor TP53 Our analyses revealed that 17p loss leads to global gene expression changes and reduced tumor infiltration and cytotoxicity of T cells, resulting in immune evasion during breast tumor progression. The 17p deletion region also includes POLR2A, a gene encoding the catalytic subunit of RNA polymerase II that is essential for cell survival. Therefore, breast cancer cells with heterozygous loss of 17p are extremely sensitive to the inhibition of POLR2A via a specific small-molecule inhibitor, α-amanitin. Here, we demonstrate that α-amanitin-conjugated trastuzumab (T-Ama) potentiated the HER2-targeted therapy and exhibited superior efficacy in treating HER2-low breast cancer with 17p loss. Moreover, treatment with T-Ama induced immunogenic cell death in breast cancer cells and, thereby, delivered greater efficacy in combination with immune checkpoint blockade therapy in preclinical HER2-low breast cancer models. Collectively, 17p loss not only drives breast tumorigenesis but also confers therapeutic vulnerabilities that may be used to develop targeted precision immunotherapy.
Collapse
Affiliation(s)
- Yujing Li
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Yifan Sun
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Michael Kulke
- Heidelberg Pharma Research GmbH, Ladenburg 68526, Germany
| | | | - Kevin Van der Jeught
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Tianhan Dong
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Bin He
- Departments of Surgery and Urology, Immunobiology and Transplant Science Center, Houston Methodist Cancer Center, Houston Methodist Research Institute, Houston Methodist Hospital, Houston, TX 77030, USA
- Department of Medicine, Weill Cornell Medicine of Cornell University, New York, NY 10065, USA
| | - Kathy D Miller
- Indiana University Melvin and Bren Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Division of Hematology/Oncology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Milan Radovich
- Indiana University Melvin and Bren Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Bryan P Schneider
- Indiana University Melvin and Bren Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Division of Hematology/Oncology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Andreas Pahl
- Heidelberg Pharma Research GmbH, Ladenburg 68526, Germany
| | - Xinna Zhang
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
- Indiana University Melvin and Bren Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Xiongbin Lu
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
- Indiana University Melvin and Bren Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Indiana University Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|
17
|
De Lucia M, Cotugno G, Bignone V, Garzia I, Nocchi L, Langone F, Petrovic B, Sasso E, Pepe S, Froechlich G, Gentile C, Zambrano N, Campadelli-Fiume G, Nicosia A, Scarselli E, D'Alise AM. Retargeted and Multi-cytokine-Armed Herpes Virus Is a Potent Cancer Endovaccine for Local and Systemic Anti-tumor Treatment. MOLECULAR THERAPY-ONCOLYTICS 2020; 19:253-264. [PMID: 33209980 PMCID: PMC7658578 DOI: 10.1016/j.omto.2020.10.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 10/08/2020] [Indexed: 12/19/2022]
Abstract
Oncolytic viruses (OVs) are novel anti-tumor agents with the ability to selectively infect and kill tumor cells while sparing normal tissue. Beyond tumor cytolysis, OVs are capable of priming an anti-tumor immune response via lysis and cross-presentation of locally expressed endogenous tumor antigens, acting as an “endovaccine.” The effectiveness of OVs, similar to other immunotherapies, can be hampered by an immunosuppressive tumor microenvironment. In this study, we modified a previously generated oncolytic herpes simplex virus (oHSV) retargeted to the human HER2 (hHER2) tumor molecule and encoding murine interleukin-12 (mIL-12), by insertion of a second immunomodulatory molecule, murine granulocyte-macrophage colony-stimulating factor (mGM-CSF), to maximize therapeutic efficacy. We assessed the efficacy of this double-armed virus (R-123) compared to singly expressing GM-CSF and IL-12 oHSVs in tumor-bearing mice. While monotherapies were poorly effective, combination with α-PD1 enhanced the anti-tumor response, with the highest efficacy of 100% response rate achieved by the combination of R-123 and α-PD1. Efficacy was T cell-dependent, and the induced immunity was long lasting and able to reject a second contralateral tumor. Importantly, systemic delivery of R-123 combined with α-PD1 was effective in inhibiting the development of tumor metastasis. As such, this approach could have a significant therapeutic impact paving the way for further development of this platform in cancer immunotherapy.
Collapse
Affiliation(s)
- Maria De Lucia
- Nouscom S.r.l., Via Castel Romano 100, 00128 Rome, Italy
| | | | | | - Irene Garzia
- Nouscom S.r.l., Via Castel Romano 100, 00128 Rome, Italy
| | - Linda Nocchi
- Nouscom S.r.l., Via Castel Romano 100, 00128 Rome, Italy
| | | | | | - Emanuele Sasso
- Nouscom S.r.l., Via Castel Romano 100, 00128 Rome, Italy
| | - Simona Pepe
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, 40126 Bologna, Italy
| | - Guendalina Froechlich
- CEINGE-Biotecnologie Avanzate S.C. aR.L., via Gaetano Salvatore 486, 80145 Naples, Italy.,Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, Via Sergio Pansini 5, 80131 Napoli, Italy
| | - Chiara Gentile
- CEINGE-Biotecnologie Avanzate S.C. aR.L., via Gaetano Salvatore 486, 80145 Naples, Italy.,Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, Via Sergio Pansini 5, 80131 Napoli, Italy
| | - Nicola Zambrano
- CEINGE-Biotecnologie Avanzate S.C. aR.L., via Gaetano Salvatore 486, 80145 Naples, Italy.,Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, Via Sergio Pansini 5, 80131 Napoli, Italy
| | - Gabriella Campadelli-Fiume
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, 40126 Bologna, Italy
| | - Alfredo Nicosia
- CEINGE-Biotecnologie Avanzate S.C. aR.L., via Gaetano Salvatore 486, 80145 Naples, Italy.,Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, Via Sergio Pansini 5, 80131 Napoli, Italy
| | | | | |
Collapse
|
18
|
Wei WZ, Gibson HM, Jacob JB, Frelinger JA, Berzofsky JA, Maeng H, Dyson G, Reyes JD, Pilon-Thomas S, Ratner S, Wei KC. Diversity Outbred Mice Reveal the Quantitative Trait Locus and Regulatory Cells of HER2 Immunity. THE JOURNAL OF IMMUNOLOGY 2020; 205:1554-1563. [PMID: 32796024 DOI: 10.4049/jimmunol.2000466] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 07/11/2020] [Indexed: 01/14/2023]
Abstract
The genetic basis and mechanisms of disparate antitumor immune response was investigated in Diversity Outbred (DO) F1 mice that express human HER2. DO mouse stock samples nearly the entire genetic repertoire of the species. We crossed DO mice with syngeneic HER2 transgenic mice to study the genetics of an anti-self HER2 response in a healthy outbred population. Anti-HER2 IgG was induced by Ad/E2TM or naked pE2TM, both encoding HER2 extracellular and transmembrane domains. The response of DO F1 HER2 transgenic mice was remarkably variable. Still, immune sera inhibited HER2+ SKBR3 cell survival in a dose-dependent fashion. Using DO quantitative trait locus (QTL) analysis, we mapped the QTL that influences both total IgG and IgG2(a/b/c) Ab response to either Ad/E2TM or pE2TM. QTL from these four datasets identified a region in chromosome 17 that was responsible for regulating the response. A/J and NOD segments of genes in this region drove elevated HER2 Ig levels. This region is rich in MHC-IB genes, several of which interact with inhibitory receptors of NK cells. (B6xA/J)F1 and (B6xNOD)F1 HER2 transgenic mice received Ad/E2TM after NK cell depletion, and they produced less HER2 IgG, demonstrating positive regulatory function of NK cells. Depletion of regulatory T cells enhanced response. Using DO QTL analysis, we show that MHC-IB reactive NK cells exert positive influence on the immunity, countering negative regulation by regulatory T cells. This new, to our knowledge, DO F1 platform is a powerful tool for revealing novel immune regulatory mechanisms and for testing new interventional strategies.
Collapse
Affiliation(s)
- Wei-Zen Wei
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI 48201;
| | - Heather M Gibson
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI 48201
| | - Jennifer B Jacob
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI 48201
| | - Jeffrey A Frelinger
- Valley Fever Center of Excellence, Department of Immunobiology, University of Arizona, Tucson, AZ 85724
| | - Jay A Berzofsky
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892; and
| | - Hoyoung Maeng
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892; and
| | - Gregory Dyson
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI 48201
| | - Joyce D Reyes
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI 48201
| | - Shari Pilon-Thomas
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612
| | - Stuart Ratner
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI 48201
| | - Kuang-Chung Wei
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI 48201
| |
Collapse
|
19
|
Crosby EJ, Acharya CR, Haddad AF, Rabiola CA, Lei G, Wei JP, Yang XY, Wang T, Liu CX, Wagner KU, Muller WJ, Chodosh LA, Broadwater G, Hyslop T, Shepherd JH, Hollern DP, He X, Perou CM, Chai S, Ashby BK, Vincent BG, Snyder JC, Force J, Morse MA, Lyerly HK, Hartman ZC. Stimulation of Oncogene-Specific Tumor-Infiltrating T Cells through Combined Vaccine and αPD-1 Enable Sustained Antitumor Responses against Established HER2 Breast Cancer. Clin Cancer Res 2020; 26:4670-4681. [PMID: 32732224 DOI: 10.1158/1078-0432.ccr-20-0389] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 04/17/2020] [Accepted: 06/25/2020] [Indexed: 12/15/2022]
Abstract
PURPOSE Despite promising advances in breast cancer immunotherapy, augmenting T-cell infiltration has remained a significant challenge. Although neither individual vaccines nor immune checkpoint blockade (ICB) have had broad success as monotherapies, we hypothesized that targeted vaccination against an oncogenic driver in combination with ICB could direct and enable antitumor immunity in advanced cancers. EXPERIMENTAL DESIGN Our models of HER2+ breast cancer exhibit molecular signatures that are reflective of advanced human HER2+ breast cancer, with a small numbers of neoepitopes and elevated immunosuppressive markers. Using these, we vaccinated against the oncogenic HER2Δ16 isoform, a nondriver tumor-associated gene (GFP), and specific neoepitopes. We further tested the effect of vaccination or anti-PD-1, alone and in combination. RESULTS We found that only vaccination targeting HER2Δ16, a driver of oncogenicity and HER2-therapeutic resistance, could elicit significant antitumor responses, while vaccines targeting a nondriver tumor-specific antigen or tumor neoepitopes did not. Vaccine-induced HER2-specific CD8+ T cells were essential for responses, which were more effective early in tumor development. Long-term tumor control of advanced cancers occurred only when HER2Δ16 vaccination was combined with αPD-1. Single-cell RNA sequencing of tumor-infiltrating T cells revealed that while vaccination expanded CD8 T cells, only the combination of vaccine with αPD-1 induced functional gene expression signatures in those CD8 T cells. Furthermore, we show that expanded clones are HER2-reactive, conclusively demonstrating the efficacy of this vaccination strategy in targeting HER2. CONCLUSIONS Combining oncogenic driver targeted vaccines with selective ICB offers a rational paradigm for precision immunotherapy, which we are clinically evaluating in a phase II trial (NCT03632941).
Collapse
Affiliation(s)
- Erika J Crosby
- Department of Surgery, Division of Surgical Sciences, Duke University, Durham North Carolina
| | - Chaitanya R Acharya
- Department of Surgery, Division of Surgical Sciences, Duke University, Durham North Carolina
| | - Anthony-Fayez Haddad
- Department of Surgery, Division of Surgical Sciences, Duke University, Durham North Carolina
| | - Christopher A Rabiola
- Department of Surgery, Division of Surgical Sciences, Duke University, Durham North Carolina
| | - Gangjun Lei
- Department of Surgery, Division of Surgical Sciences, Duke University, Durham North Carolina
| | - Jun-Ping Wei
- Department of Surgery, Division of Surgical Sciences, Duke University, Durham North Carolina
| | - Xiao-Yi Yang
- Department of Surgery, Division of Surgical Sciences, Duke University, Durham North Carolina
| | - Tao Wang
- Department of Surgery, Division of Surgical Sciences, Duke University, Durham North Carolina
| | - Cong-Xiao Liu
- Department of Surgery, Division of Surgical Sciences, Duke University, Durham North Carolina
| | - Kay U Wagner
- Department of Oncology, Wayne State University, Barbara Ann Karmanos Cancer Institute, Detroit, Michigan
| | - William J Muller
- Departments of Biochemistry and Medicine, Goodman Cancer Center, McGill University, Montreal, Quebec
| | - Lewis A Chodosh
- Department of Cancer Biology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Gloria Broadwater
- Department of Biostatistics and Bioinformatics, Duke University, Durham, North Carolina
| | - Terry Hyslop
- Department of Biostatistics and Bioinformatics, Duke University, Durham, North Carolina
| | - Jonathan H Shepherd
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina.,Department of Genetics, University of North Carolina, Chapel Hill, North Carolina
| | - Daniel P Hollern
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina.,Department of Genetics, University of North Carolina, Chapel Hill, North Carolina
| | - Xiaping He
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina.,Department of Genetics, University of North Carolina, Chapel Hill, North Carolina
| | - Charles M Perou
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina.,Department of Genetics, University of North Carolina, Chapel Hill, North Carolina
| | - Shengjie Chai
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina.,Department of Medicine, Division of Hematology/Oncology, University of North Carolina, Chapel Hill, North Carolina
| | - Benjamin K Ashby
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina.,Department of Medicine, Division of Hematology/Oncology, University of North Carolina, Chapel Hill, North Carolina
| | - Benjamin G Vincent
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina.,Department of Medicine, Division of Hematology/Oncology, University of North Carolina, Chapel Hill, North Carolina.,Curriculum in Bioinformatics and Computational Biology, University of North Carolina, Chapel Hill, North Carolina.,Computational Medicine Program, University of North Carolina, Chapel Hill, North Carolina
| | - Joshua C Snyder
- Department of Surgery, Division of Surgical Sciences, Duke University, Durham North Carolina.,Department of Cell Biology, Duke University, Durham, North Carolina
| | - Jeremy Force
- Department of Medicine, Duke University, Durham, North Carolina
| | - Michael A Morse
- Department of Medicine, Duke University, Durham, North Carolina
| | - Herbert K Lyerly
- Department of Surgery, Division of Surgical Sciences, Duke University, Durham North Carolina.,Department of Immunology, Duke University, Durham, North Carolina.,Department of Pathology, Duke University, Durham, North Carolina
| | - Zachary C Hartman
- Department of Surgery, Division of Surgical Sciences, Duke University, Durham North Carolina. .,Department of Pathology, Duke University, Durham, North Carolina
| |
Collapse
|
20
|
Chan JD, von Scheidt B, Zeng B, Oliver AJ, Davey AS, Ali AI, Thomas R, Trapani JA, Darcy PK, Kershaw MH, Dolcetti R, Slaney CY. Enhancing chimeric antigen receptor T-cell immunotherapy against cancer using a nanoemulsion-based vaccine targeting cross-presenting dendritic cells. Clin Transl Immunology 2020; 9:e1157. [PMID: 32704371 PMCID: PMC7374388 DOI: 10.1002/cti2.1157] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Revised: 06/25/2020] [Accepted: 06/25/2020] [Indexed: 12/30/2022] Open
Abstract
Objectives Adoptive transfer of chimeric antigen receptor (CAR)-modified T cells is a form of cancer immunotherapy that has achieved remarkable efficacy in patients with some haematological cancers. However, challenges remain in CAR T-cell treatment of solid tumours because of tumour-mediated immunosuppression. Methods We have demonstrated that CAR T-cell stimulation through T-cell receptors (TCRs) in vivo can generate durable responses against solid tumours in a variety of murine models. Since Clec9A-targeting tailored nanoemulsion (Clec9A-TNE) vaccine enhances antitumour immune responses through selective activation of Clec9A+ cross-presenting dendritic cells (DCs), we hypothesised that Clec9A-TNE could prime DCs for antigen presentation to CAR T cells through TCRs and thus improve CAR T-cell responses against solid tumours. To test this hypothesis, we used CAR T cells expressing transgenic TCRs specific for ovalbumin (OVA) peptides SIINFEKL (CAROTI) or OVA323-339 (CAROTII). Results We demonstrated that the Clec9A-TNEs encapsulating full-length recombinant OVA protein (OVA-Clec9A-TNE) improved CAROT T-cell proliferation and inflammatory cytokine secretion in vitro. Combined treatment using the OVA-Clec9A-TNE and CAROT cells resulted in durable responses and some rejections of tumours in immunocompetent mice. Tumour regression was accompanied by enhanced CAROT cell proliferation and infiltration into the tumours. Conclusion Our study presents Clec9A-TNE as a prospective avenue to enhance CAR T-cell efficacy for solid cancers.
Collapse
Affiliation(s)
- Jack D Chan
- Cancer Immunology Program Peter MacCallum Cancer Center Melbourne VIC Australia.,Sir Peter MacCallum Department of Oncology University of Melbourne Parkville VIC Australia
| | - Bianca von Scheidt
- Cancer Immunology Program Peter MacCallum Cancer Center Melbourne VIC Australia
| | - Bijun Zeng
- The University of Queensland Diamantina Institute Translational Research Institute Woolloongabba QLD Australia
| | - Amanda J Oliver
- Cancer Immunology Program Peter MacCallum Cancer Center Melbourne VIC Australia.,Sir Peter MacCallum Department of Oncology University of Melbourne Parkville VIC Australia
| | - Ashleigh S Davey
- Cancer Immunology Program Peter MacCallum Cancer Center Melbourne VIC Australia
| | - Aesha I Ali
- Cancer Immunology Program Peter MacCallum Cancer Center Melbourne VIC Australia.,Sir Peter MacCallum Department of Oncology University of Melbourne Parkville VIC Australia
| | - Ranjeny Thomas
- The University of Queensland Diamantina Institute Translational Research Institute Woolloongabba QLD Australia
| | - Joseph A Trapani
- Cancer Immunology Program Peter MacCallum Cancer Center Melbourne VIC Australia.,Sir Peter MacCallum Department of Oncology University of Melbourne Parkville VIC Australia
| | - Phillip K Darcy
- Cancer Immunology Program Peter MacCallum Cancer Center Melbourne VIC Australia.,Sir Peter MacCallum Department of Oncology University of Melbourne Parkville VIC Australia
| | - Michael H Kershaw
- Cancer Immunology Program Peter MacCallum Cancer Center Melbourne VIC Australia.,Sir Peter MacCallum Department of Oncology University of Melbourne Parkville VIC Australia
| | - Riccardo Dolcetti
- The University of Queensland Diamantina Institute Translational Research Institute Woolloongabba QLD Australia
| | - Clare Y Slaney
- Cancer Immunology Program Peter MacCallum Cancer Center Melbourne VIC Australia.,Sir Peter MacCallum Department of Oncology University of Melbourne Parkville VIC Australia
| |
Collapse
|
21
|
Anti-PSMA CAR-engineered NK-92 Cells: An Off-the-shelf Cell Therapy for Prostate Cancer. Cells 2020; 9:cells9061382. [PMID: 32498368 PMCID: PMC7349573 DOI: 10.3390/cells9061382] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 05/29/2020] [Accepted: 05/31/2020] [Indexed: 01/22/2023] Open
Abstract
Prostate cancer (PCa) has become the most common cancer among males in Europe and the USA. Adoptive immunotherapy appears a promising strategy to control the advanced stages of the disease by specifically targeting the tumor, in particular through chimeric antigen receptor T (CAR-T) cell therapy. Despite the advancements of CAR-T technology in the treatment of hematological malignancies, solid tumors still represent a challenge. To overcome current limits, other cellular effectors than T lymphocytes are under study as possible candidates for CAR-engineered cancer immunotherapy. A novel approach involves the NK-92 cell line, which mediates strong cytotoxic responses against a variety of tumor cells but has no effect on non-malignant healthy counterparts. Here, we report a novel therapeutic approach against PCa based on engineering of NK-92 cells with a CAR recognizing the human prostate-specific membrane antigen (PSMA), which is overexpressed in prostatic neoplastic cells. More importantly, the potential utility of NK-92/CAR cells to treat PCa has not yet been explored. Upon CAR transduction, NK-92/CAR cells acquired high and specific lytic activity against PSMA-expressing prostate cancer cells in vitro, and also underwent degranulation and produced high levels of IFN-γ in response to antigen recognition. Lethal irradiation of the effectors, a safety measure requested for the clinical application of retargeted NK-92 cells, fully abrogated replication but did not impact on phenotype and short-term functionality. PSMA-specific recognition and antitumor activity were retained in vivo, as adoptive transfer of irradiated NK-92/CAR cells in prostate cancer-bearing mice restrained tumor growth and improved survival. Anti-PSMA CAR-modified NK-92 cells represent a universal, off-the-shelf, renewable, and cost-effective product endowed with relevant potentialities as a therapeutic approach for PCa immunotherapy.
Collapse
|
22
|
Lai J, Mardiana S, House IG, Sek K, Henderson MA, Giuffrida L, Chen AXY, Todd KL, Petley EV, Chan JD, Carrington EM, Lew AM, Solomon BJ, Trapani JA, Kedzierska K, Evrard M, Vervoort SJ, Waithman J, Darcy PK, Beavis PA. Adoptive cellular therapy with T cells expressing the dendritic cell growth factor Flt3L drives epitope spreading and antitumor immunity. Nat Immunol 2020; 21:914-926. [PMID: 32424363 DOI: 10.1038/s41590-020-0676-7] [Citation(s) in RCA: 132] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 03/31/2020] [Indexed: 12/21/2022]
Abstract
Adoptive cell therapies using genetically engineered T cell receptor or chimeric antigen receptor T cells are emerging forms of immunotherapy that redirect T cells to specifically target cancer. However, tumor antigen heterogeneity remains a key challenge limiting their efficacy against solid cancers. Here, we engineered T cells to secrete the dendritic cell (DC) growth factor Fms-like tyrosine kinase 3 ligand (Flt3L). Flt3L-secreting T cells expanded intratumoral conventional type 1 DCs and substantially increased host DC and T cell activation when combined with immune agonists poly (I:C) and anti-4-1BB. Importantly, combination therapy led to enhanced inhibition of tumor growth and the induction of epitope spreading towards antigens beyond those recognized by adoptively transferred T cells in solid tumor models of T cell receptor and chimeric antigen receptor T cell therapy. Our data suggest that augmenting endogenous DCs is a promising strategy to overcome the clinical problem of antigen-negative tumor escape following adoptive cell therapy.
Collapse
Affiliation(s)
- Junyun Lai
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia
| | - Sherly Mardiana
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia
| | - Imran G House
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia
| | - Kevin Sek
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia
| | - Melissa A Henderson
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia
| | - Lauren Giuffrida
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia
| | - Amanda X Y Chen
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia
| | - Kirsten L Todd
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia
| | - Emma V Petley
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia
| | - Jack D Chan
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia
| | - Emma M Carrington
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia.,Department of Medical Biology, University of Melbourne, Melbourne, Victoria, Australia
| | - Andrew M Lew
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia.,Department of Medical Biology, University of Melbourne, Melbourne, Victoria, Australia
| | - Benjamin J Solomon
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia
| | - Joseph A Trapani
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia
| | - Katherine Kedzierska
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Maximilien Evrard
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Stephin J Vervoort
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia
| | - Jason Waithman
- Telethon Kids Institute, University of Western Australia, Perth, Western Australia, Australia
| | - Phillip K Darcy
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia. .,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia. .,Department of Pathology, University of Melbourne, Parkville, Victoria, Australia. .,Department of Immunology, Monash University, Clayton, Victoria, Australia.
| | - Paul A Beavis
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia. .,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia.
| |
Collapse
|
23
|
Sasso E, Froechlich G, Cotugno G, D'Alise AM, Gentile C, Bignone V, De Lucia M, Petrovic B, Campadelli-Fiume G, Scarselli E, Nicosia A, Zambrano N. Replicative conditioning of Herpes simplex type 1 virus by Survivin promoter, combined to ERBB2 retargeting, improves tumour cell-restricted oncolysis. Sci Rep 2020; 10:4307. [PMID: 32152425 PMCID: PMC7062820 DOI: 10.1038/s41598-020-61275-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Accepted: 02/20/2020] [Indexed: 12/13/2022] Open
Abstract
Oncolytic virotherapy is emerging as a promising therapeutic option for solid tumours. Several oncolytic vectors in clinical testing are based on attenuated viruses; thus, efforts are being taken to develop a new repertoire of oncolytic viruses, based on virulent viral genomes. This possibility, however, raises concerns dealing with the safety features of the virulent phenotypes. We generated a double regulated Herpes simplex type-1 virus (HSV-1), in which tumour cell restricted replicative potential was combined to selective entry via ERBB2 receptor retargeting. The transcriptional control of the viral alpha4 gene encoding for the infected cell protein-4 (ICP4) by the cellular Survivin/BIRC5 promoter conferred a tumour cell-restricted replicative potential to a virulent HSV-1 genome. The combination of the additional ERBB2 retargeting further improved the selectivity for tumour cells, conferring to the double regulated virus a very limited ability to infect and propagate in non-cancerous cells. Accordingly, a suitable replicative and cytotoxic potential was maintained in tumour cell lines, allowing the double regulated virus to synergize in vivo with immune checkpoint (anti-PD-1) blockade in immunocompetent mice. Thus, restricting the replicative spectrum and tropism of virulent HSV-1 genomes by combination of conditional replication and retargeting provides an improved safety, does not alter the oncolytic strength, and is exploitable for its therapeutic potential with immune checkpoint blockade in cancer.
Collapse
Affiliation(s)
- Emanuele Sasso
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, Via S. Pansini, 5, 80131, Naples, Italy. .,CEINGE Biotecnologie Avanzate S.C.aR.L., Via G. Salvatore 486, 80145, Naples, Italy. .,Nouscom S.R.L., Via di Castel Romano 100, 00128, Rome, Italy.
| | | | | | | | - Chiara Gentile
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, Via S. Pansini, 5, 80131, Naples, Italy.,CEINGE Biotecnologie Avanzate S.C.aR.L., Via G. Salvatore 486, 80145, Naples, Italy
| | | | - Maria De Lucia
- Nouscom S.R.L., Via di Castel Romano 100, 00128, Rome, Italy
| | | | - Gabriella Campadelli-Fiume
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Via San Giacomo 12, 40126, Bologna, Italy
| | - Elisa Scarselli
- Nouscom S.R.L., Via di Castel Romano 100, 00128, Rome, Italy
| | - Alfredo Nicosia
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, Via S. Pansini, 5, 80131, Naples, Italy.,CEINGE Biotecnologie Avanzate S.C.aR.L., Via G. Salvatore 486, 80145, Naples, Italy.,Nouscom S.R.L., Via di Castel Romano 100, 00128, Rome, Italy
| | - Nicola Zambrano
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, Via S. Pansini, 5, 80131, Naples, Italy.,CEINGE Biotecnologie Avanzate S.C.aR.L., Via G. Salvatore 486, 80145, Naples, Italy
| |
Collapse
|
24
|
Wiede F, Lu K, Du X, Liang S, Hochheiser K, Dodd GT, Goh PK, Kearney C, Meyran D, Beavis PA, Henderson MA, Park SL, Waithman J, Zhang S, Zhang Z, Oliaro J, Gebhardt T, Darcy PK, Tiganis T. PTPN2 phosphatase deletion in T cells promotes anti-tumour immunity and CAR T-cell efficacy in solid tumours. EMBO J 2020; 39:e103637. [PMID: 31803974 PMCID: PMC6960448 DOI: 10.15252/embj.2019103637] [Citation(s) in RCA: 86] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 11/04/2019] [Accepted: 11/08/2019] [Indexed: 12/18/2022] Open
Abstract
Although adoptive T-cell therapy has shown remarkable clinical efficacy in haematological malignancies, its success in combating solid tumours has been limited. Here, we report that PTPN2 deletion in T cells enhances cancer immunosurveillance and the efficacy of adoptively transferred tumour-specific T cells. T-cell-specific PTPN2 deficiency prevented tumours forming in aged mice heterozygous for the tumour suppressor p53. Adoptive transfer of PTPN2-deficient CD8+ T cells markedly repressed tumour formation in mice bearing mammary tumours. Moreover, PTPN2 deletion in T cells expressing a chimeric antigen receptor (CAR) specific for the oncoprotein HER-2 increased the activation of the Src family kinase LCK and cytokine-induced STAT-5 signalling, thereby enhancing both CAR T-cell activation and homing to CXCL9/10-expressing tumours to eradicate HER-2+ mammary tumours in vivo. Our findings define PTPN2 as a target for bolstering T-cell-mediated anti-tumour immunity and CAR T-cell therapy against solid tumours.
Collapse
Affiliation(s)
- Florian Wiede
- Monash Biomedicine Discovery InstituteMonash UniversityClaytonVic.Australia
- Department of Biochemistry and Molecular BiologyMonash UniversityClaytonVic.Australia
- Peter MacCallum Cancer CentreMelbourneVic.Australia
| | - Kun‐Hui Lu
- Peter MacCallum Cancer CentreMelbourneVic.Australia
| | - Xin Du
- Peter MacCallum Cancer CentreMelbourneVic.Australia
- Sir Peter MacCallum Department of OncologyThe University of MelbourneMelbourneVic.Australia
| | - Shuwei Liang
- Monash Biomedicine Discovery InstituteMonash UniversityClaytonVic.Australia
- Department of Biochemistry and Molecular BiologyMonash UniversityClaytonVic.Australia
- Peter MacCallum Cancer CentreMelbourneVic.Australia
| | - Katharina Hochheiser
- Peter MacCallum Cancer CentreMelbourneVic.Australia
- Department of Microbiology and ImmunologyThe University of MelbourneMelbourneVic.Australia
- Peter Doherty Institute for Infection and ImmunityMelbourneVic.Australia
| | - Garron T Dodd
- Monash Biomedicine Discovery InstituteMonash UniversityClaytonVic.Australia
- Department of Biochemistry and Molecular BiologyMonash UniversityClaytonVic.Australia
| | - Pei K Goh
- Monash Biomedicine Discovery InstituteMonash UniversityClaytonVic.Australia
- Department of Biochemistry and Molecular BiologyMonash UniversityClaytonVic.Australia
- Peter MacCallum Cancer CentreMelbourneVic.Australia
| | | | | | - Paul A Beavis
- Peter MacCallum Cancer CentreMelbourneVic.Australia
- Sir Peter MacCallum Department of OncologyThe University of MelbourneMelbourneVic.Australia
| | | | - Simone L Park
- Department of Microbiology and ImmunologyThe University of MelbourneMelbourneVic.Australia
- Peter Doherty Institute for Infection and ImmunityMelbourneVic.Australia
| | - Jason Waithman
- Telethon Kids InstituteUniversity of Western AustraliaPerthWAAustralia
| | - Sheng Zhang
- Department of Medicinal Chemistry and Molecular PharmacologyInstitute for Drug DiscoveryPurdue UniversityWest LafayetteINUSA
| | - Zhong‐Yin Zhang
- Department of Medicinal Chemistry and Molecular PharmacologyInstitute for Drug DiscoveryPurdue UniversityWest LafayetteINUSA
| | - Jane Oliaro
- Peter MacCallum Cancer CentreMelbourneVic.Australia
- Sir Peter MacCallum Department of OncologyThe University of MelbourneMelbourneVic.Australia
| | - Thomas Gebhardt
- Department of Microbiology and ImmunologyThe University of MelbourneMelbourneVic.Australia
- Peter Doherty Institute for Infection and ImmunityMelbourneVic.Australia
| | - Phillip K Darcy
- Peter MacCallum Cancer CentreMelbourneVic.Australia
- Sir Peter MacCallum Department of OncologyThe University of MelbourneMelbourneVic.Australia
| | - Tony Tiganis
- Monash Biomedicine Discovery InstituteMonash UniversityClaytonVic.Australia
- Department of Biochemistry and Molecular BiologyMonash UniversityClaytonVic.Australia
- Peter MacCallum Cancer CentreMelbourneVic.Australia
| |
Collapse
|
25
|
An HER2 DNA vaccine with evolution-selected amino acid substitutions reveals a fundamental principle for cancer vaccine formulation in HER2 transgenic mice. Cancer Immunol Immunother 2019; 68:1143-1155. [PMID: 31177328 DOI: 10.1007/s00262-019-02333-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 03/26/2019] [Indexed: 12/19/2022]
Abstract
Enhancement of endogenous immunity to tumor-associated self-antigens and neoantigens is the goal of preventive vaccination. Toward this goal, we compared the efficacy of the following HER2 DNA vaccine constructs: vaccines encoding wild-type HER2, hybrid HER2 vaccines consisting of human HER2 and rat Neu, HER2 vaccines with single residue substitutions and a novel human HER2 DNA vaccine, ph(es)E2TM. ph(es)E2TM was designed to contain five evolution-selected substitutions: M198V, Q398R, F425L, H473R and A622T that occur frequently in 12 primate HER2 sequences. These ph(es)E2TM substitutions score 0 to 1 in blocks substitutions matrix (BLOSUM), indicating minimal biochemical alterations. h(es)E2TM recombinant protein is recognized by a panel of anti-HER2 mAbs, demonstrating the preservation of HER2 protein structure. Compared to native human HER2, electrovaccination of HER2 transgenic mice with ph(es)E2TM induced a threefold increase in HER2-binding antibody (Ab) and elevated levels of IFNγ-producing T cells. ph(es)E2TM, but not pE2TM immune serum, recognized HER2 peptide p95 355LPESFDGDPASNTAP369, suggesting a broadening of epitope recognition induced by the minimally modified HER2 vaccine. ph(es)E2TM vaccination reduced tumor growth more effectively than wild-type HER2 or HER2 vaccines with more extensive modifications. The elevation of tumor immunity by ph(es)E2TM vaccination would create a favorable tumor microenvironment for neoantigen priming, further enhancing the protective immunity. The fundamental principle of exploiting evolution-selected amino acid substitutions is novel, effective and applicable to vaccine development in general.
Collapse
|
26
|
Cianciaruso C, Beltraminelli T, Duval F, Nassiri S, Hamelin R, Mozes A, Gallart-Ayala H, Ceada Torres G, Torchia B, Ries CH, Ivanisevic J, De Palma M. Molecular Profiling and Functional Analysis of Macrophage-Derived Tumor Extracellular Vesicles. Cell Rep 2019; 27:3062-3080.e11. [PMID: 31167148 PMCID: PMC6581796 DOI: 10.1016/j.celrep.2019.05.008] [Citation(s) in RCA: 128] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 04/10/2019] [Accepted: 04/30/2019] [Indexed: 12/14/2022] Open
Abstract
Extracellular vesicles (EVs), including exosomes, modulate multiple aspects of cancer biology. Tumor-associated macrophages (TAMs) secrete EVs, but their molecular features and functions are poorly characterized. Here, we report methodology for the enrichment, quantification, and proteomic and lipidomic analysis of EVs released from mouse TAMs (TAM-EVs). Compared to source TAMs, TAM-EVs present molecular profiles associated with a Th1/M1 polarization signature, enhanced inflammation and immune response, and a more favorable patient prognosis. Accordingly, enriched TAM-EV preparations promote T cell proliferation and activation ex vivo. TAM-EVs also contain bioactive lipids and biosynthetic enzymes, which may alter pro-inflammatory signaling in the cancer cells. Thus, whereas TAMs are largely immunosuppressive, their EVs may have the potential to stimulate, rather than limit, anti-tumor immunity.
Collapse
Affiliation(s)
- Chiara Cianciaruso
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland.
| | - Tim Beltraminelli
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Florent Duval
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Sina Nassiri
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Romain Hamelin
- Proteomics Core Facility, School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - André Mozes
- Flow Cytometry Core Facility, School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Hector Gallart-Ayala
- Metabolomics Platform, Faculty of Biology and Medicine, University of Lausanne, 1005 Lausanne, Switzerland
| | - Gerardo Ceada Torres
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Bruno Torchia
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Carola H Ries
- Roche Innovation Center Munich, Roche Pharma Research and Early Development, 82377 Penzberg, Germany
| | - Julijana Ivanisevic
- Metabolomics Platform, Faculty of Biology and Medicine, University of Lausanne, 1005 Lausanne, Switzerland
| | - Michele De Palma
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland.
| |
Collapse
|
27
|
Crosby EJ, Gwin W, Blackwell K, Marcom PK, Chang S, Maecker HT, Broadwater G, Hyslop T, Kim S, Rogatko A, Lubkov V, Snyder JC, Osada T, Hobeika AC, Morse MA, Lyerly HK, Hartman ZC. Vaccine-Induced Memory CD8 + T Cells Provide Clinical Benefit in HER2 Expressing Breast Cancer: A Mouse to Human Translational Study. Clin Cancer Res 2019; 25:2725-2736. [PMID: 30635338 PMCID: PMC6497539 DOI: 10.1158/1078-0432.ccr-18-3102] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 11/28/2018] [Accepted: 01/08/2019] [Indexed: 01/23/2023]
Abstract
PURPOSE Immune-based therapy for metastatic breast cancer has had limited success, particularly in molecular subtypes with low somatic mutations rates. Strategies to augment T-cell infiltration of tumors include vaccines targeting established oncogenic drivers such as the genomic amplification of HER2. We constructed a vaccine based on a novel alphaviral vector encoding a portion of HER2 (VRP-HER2). PATIENTS AND METHODS In preclinical studies, mice were immunized with VRP-HER2 before or after implantation of hHER2+ tumor cells and HER2-specific immune responses and antitumor function were evaluated. We tested VRP-HER2 in a phase I clinical trial where subjects with advanced HER2-overexpressing malignancies in cohort 1 received VRP-HER2 every 2 weeks for a total of 3 doses. In cohort 2, subjects received the same schedule concurrently with a HER2-targeted therapy. RESULTS Vaccination in preclinical models with VRP-HER2 induced HER2-specific T cells and antibodies while inhibiting tumor growth. VRP-HER2 was well tolerated in patients and vaccination induced HER2-specific T cells and antibodies. Although a phase I study, there was 1 partial response and 2 patients with continued stable disease. Median OS was 50.2 months in cohort 1 (n = 4) and 32.7 months in cohort 2 (n = 18). Perforin expression by memory CD8 T cells post-vaccination significantly correlated with improved PFS. CONCLUSIONS VRP-HER2 increased HER2-specific memory CD8 T cells and had antitumor effects in preclinical and clinical studies. The expansion of HER2-specific memory CD8 T cells in vaccinated patients was significantly correlated with increased PFS. Subsequent studies will seek to enhance T-cell activity by combining with anti-PD-1.
Collapse
Affiliation(s)
- Erika J Crosby
- Department of Surgery, Division of Surgical Sciences, Duke University Medical Center, Durham, North Carolina
| | - William Gwin
- Department of Medicine, Division of Medical Oncology, Duke University Medical Center, Durham, North Carolina
- Department of Medicine, Division of Medical Oncology, University of Washington, Seattle, Washington
| | - Kimberly Blackwell
- Department of Medicine, Division of Medical Oncology, Duke University Medical Center, Durham, North Carolina
| | - Paul K Marcom
- Department of Medicine, Division of Medical Oncology, Duke University Medical Center, Durham, North Carolina
| | - Serena Chang
- Department of Microbiology and Immunology, Institute for Immunity, Transplantation, and Infection, Stanford University, Stanford, California
| | - Holden T Maecker
- Department of Microbiology and Immunology, Institute for Immunity, Transplantation, and Infection, Stanford University, Stanford, California
| | - Gloria Broadwater
- Department of Biostatistics and Bioinformatics, Duke University Medical Center, Durham, North Carolina
| | - Terry Hyslop
- Department of Biostatistics and Bioinformatics, Duke University Medical Center, Durham, North Carolina
| | - Sungjin Kim
- Department of Biomedical Sciences, Biostatistics and Bioinformatics Research Center, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | - Andre Rogatko
- Department of Biomedical Sciences, Biostatistics and Bioinformatics Research Center, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | - Veronica Lubkov
- Department of Surgery, Division of Surgical Sciences, Duke University Medical Center, Durham, North Carolina
| | - Joshua C Snyder
- Department of Surgery, Division of Surgical Sciences, Duke University Medical Center, Durham, North Carolina
- Department of Cell Biology, Duke University Medical Center, Durham, North Carolina
| | - Takuya Osada
- Department of Surgery, Division of Surgical Sciences, Duke University Medical Center, Durham, North Carolina
| | - Amy C Hobeika
- Department of Surgery, Division of Surgical Sciences, Duke University Medical Center, Durham, North Carolina
| | - Michael A Morse
- Department of Surgery, Division of Surgical Sciences, Duke University Medical Center, Durham, North Carolina
- Department of Medicine, Division of Medical Oncology, Duke University Medical Center, Durham, North Carolina
| | - H Kim Lyerly
- Department of Surgery, Division of Surgical Sciences, Duke University Medical Center, Durham, North Carolina.
| | - Zachary C Hartman
- Department of Surgery, Division of Surgical Sciences, Duke University Medical Center, Durham, North Carolina.
| |
Collapse
|
28
|
Miller D, Ingersoll MA, Lin MF. ErbB-2 signaling in advanced prostate cancer progression and potential therapy. Endocr Relat Cancer 2019; 26:R195-R209. [PMID: 31294537 PMCID: PMC6628717 DOI: 10.1530/erc-19-0009] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Currently, prostate cancer (PCa) remains the most commonly diagnosed solid tumor and the second leading cause of cancer-related deaths in US men. Most of these deaths are attributed to the development of castration-resistant (CR) PCa. ErbB-2 and ErbB family members have been demonstrated to contribute to the progression of this lethal disease. In this review, we focus on updating the role of ErbB-2 in advanced PCa progression and its regulation, including its regulation via ligand activation, miRNAs and protein phosphorylation. We also discuss its downstream signaling pathways, including AKT, ERK1/2 and STATs, involved in advanced PCa progression. Additionally, we evaluate the potential of ErbB-2, focusing on its protein hyper-phosphorylation status, as a biomarker for aggressive PCa as well as the effectiveness of ErbB-2 as a target for the treatment of CR PCa via a multitude of approaches, including orally available inhibitors, intratumoral expression of cPAcP, vaccination and immunotherapy.
Collapse
Affiliation(s)
- Dannah Miller
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Nebraska Medical Center, Omaha, NE
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Matthew A. Ingersoll
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Nebraska Medical Center, Omaha, NE
| | - Ming-Fong Lin
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Nebraska Medical Center, Omaha, NE
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
- Section of Urology, Department of Surgery, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
- College of Pharmacy, Kaohsiung Medical University, Kaohsiung, Taiwan
- Corresponding Author: Ming-Fong Lin, Ph. D., Department of Biochemistry and Molecular Biology, College of Medicine, University of Nebraska Medical Center, 985870 Nebraska Medical Center, Omaha, NE 68198-5870, USA, TEL: (402) 559-6658, FAX: (402) 559-6650, (MFL)
| |
Collapse
|
29
|
Leoni V, Vannini A, Gatta V, Rambaldi J, Sanapo M, Barboni C, Zaghini A, Nanni P, Lollini PL, Casiraghi C, Campadelli-Fiume G. A fully-virulent retargeted oncolytic HSV armed with IL-12 elicits local immunity and vaccine therapy towards distant tumors. PLoS Pathog 2018; 14:e1007209. [PMID: 30080893 PMCID: PMC6095629 DOI: 10.1371/journal.ppat.1007209] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 08/16/2018] [Accepted: 07/11/2018] [Indexed: 12/12/2022] Open
Abstract
Oncolytic herpes simplex viruses (oHSVs) showed efficacy in clinical trials and practice. Most of them gain cancer-specificity from deletions/mutations in genes that counteract the host response, and grow selectively in cancer cells defective in anti-viral response. Because of the deletions/mutations, they are frequently attenuated or over-attenuated. We developed next-generation oHSVs, which carry no deletion/mutation, gain cancer-specificity from specific retargeting to tumor cell receptors-e.g. HER2 (human epidermal growth factor receptor 2)-hence are fully-virulent in the targeted cancer cells. The type of immunotherapy they elicit was not predictable, since non-attenuated HSVs induce and then dampen the innate response, whereas deleted/attenuated viruses fail to contrast it, and since the retargeted oHSVs replicate efficiently in tumor cells, but spare other cells in the tumor. We report on the first efficacy study of HER2-retargeted, fully-virulent oHSVs in immunocompetent mice. Their safety profile was very high. Both the unarmed R-LM113 and the IL-12-armed R-115 inhibited the growth of the primary HER2-Lewis lung carcinoma-1 (HER2-LLC1) tumor, R-115 being constantly more efficacious. All the mice that did not die because of the primary treated tumors, were protected from the growth of contralateral untreated tumors. The long-term survivors were protected from a second contralateral tumor, providing additional evidence for an abscopal immunotherapeutic effect. Analysis of the local response highlighted that particularly R-115 unleashed the immunosuppressive tumor microenvironment, i.e. induced immunomodulatory cytokines, including IFNγ, T-bet which promoted Th1 polarization. Some of the tumor infiltrating cells, e.g. CD4+, CD335+ cells were increased in the tumors of all responders mice, irrespective of which virus was employed, whereas CD8+, Foxp3+, CD141+ were increased and CD11b+ cells were decreased preferentially in R-115-treated mice. The durable response included a breakage of tolerance towards both HER2 and the wt tumor cells, and underscored a systemic immunotherapeutic vaccine response.
Collapse
Affiliation(s)
- Valerio Leoni
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| | - Andrea Vannini
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| | - Valentina Gatta
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| | - Julie Rambaldi
- Department of Veterinary Medical Sciences, University of Bologna, Bologna, Italy
| | - Mara Sanapo
- Department of Veterinary Medical Sciences, University of Bologna, Bologna, Italy
| | - Catia Barboni
- Department of Veterinary Medical Sciences, University of Bologna, Bologna, Italy
| | - Anna Zaghini
- Department of Veterinary Medical Sciences, University of Bologna, Bologna, Italy
| | - Patrizia Nanni
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| | - Pier-Luigi Lollini
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| | - Costanza Casiraghi
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| | - Gabriella Campadelli-Fiume
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
- * E-mail:
| |
Collapse
|
30
|
Heterologous human/rat HER2-specific exosome-targeted T cell vaccine stimulates potent humoral and CTL responses leading to enhanced circumvention of HER2 tolerance in double transgenic HLA-A2/HER2 mice. Vaccine 2018; 36:1414-1422. [PMID: 29415817 DOI: 10.1016/j.vaccine.2018.01.078] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Revised: 01/05/2018] [Accepted: 01/29/2018] [Indexed: 01/20/2023]
Abstract
DNA vaccines composed of heterologous human HER2 and rat neu sequences induce stronger antibody response and protective antitumor immunity than either HER2 or neu DNA vaccines in transgenic mice. We previously developed HER2-specific exosome-targeted T-cell vaccine HER2-TEXO capable of stimulating HER2-specific CD8+ T-cell responses, but only leading to partial protective immunity in double-transgenic HLA-A2/HER2 mice with self-immune tolerance to HER2. Here, we constructed an adenoviral vector AdVHuRt expressing HuRt fusion protein composed of NH2-HER21-407 (Hu) and COOH-neu408-690 (Rt) fragments, and developed a heterologous human/rat HER2-specific exosome-targeted T-cell vaccine HuRt-TEXO using polyclonal CD4+ T-cells uptaking exosomes released by AdVHuRt-transfected dendritic cells. We found that the HuRt-TEXO vaccine stimulates enhanced CD4+ T-cell responses leading to increased induction of HER2-specific antibody (∼70 µg/ml) compared to that (∼40 µg/ml) triggered by the homologous HER2-TEXO vaccine. By using PE-H-2Kd/HER223-71 tetramer, we determined that HuRt-TEXO stimulates stronger HER2-specific CD8+ T-cell responses eradicating 90% of HER2-specific target cells, while HER2-TEXO-induced CD8+ T-cell responses only eliminating 53% targets. Furthermore, HuRt-TEXO, but not HER2-TEXO vaccination, is capable of suppressing early stage-established HER2-expressing 4T1HER2 breast cancer in its lung metastasis or subcutaneous form in BALB/c mice, and of completely protecting transgenic HLA-A2/HER2 mice from growth of HLA-A2/HER2-expressing BL6-10A2/HER2 melanoma. HuRt-TEXO-stimulated HER2-specific CD8+ T-cells not only are cytolytic to trastuzumab-resistant HLA-A2/HER2-expressing BT474/A2 breast tumor cells in vitro but also eradicates pre-established BT474/A2 tumors in athymic nude mice. Therefore, our novel heterologous human/rat HER2-specific T-cell vaccine HuRt-TEXO, circumventing HER2 tolerance, may provide a new therapeutic alternative for patients with trastuzumab-resistant HER2+ breast tumor.
Collapse
|
31
|
Yong CSM, John LB, Devaud C, Prince MH, Johnstone RW, Trapani JA, Darcy PK, Kershaw MH. A role for multiple chimeric antigen receptor-expressing leukocytes in antigen-specific responses to cancer. Oncotarget 2018; 7:34582-98. [PMID: 27153556 PMCID: PMC5085178 DOI: 10.18632/oncotarget.9149] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Accepted: 04/16/2016] [Indexed: 12/18/2022] Open
Abstract
While adoptive immunotherapy using chimeric antigen receptor (CAR)-modified T cells can induce remission of some tumors, the role of other CAR-modified leukocytes is not well characterized. In this study, we characterize the function of leukocytes including natural killer (NK) cells, macrophages and CAR T cells from transgenic mice expressing a CAR under the control of the pan-hematopoietic promoter, vav, and determine the ability of these mice to respond to ERB expressing tumors. We demonstrate the anti-tumor functions of leukocytes, including antigen specific cytotoxicity and cytokine secretion. The adoptive transfer of CAR T cells provided a greater survival advantage in the E0771ERB tumor model than their wildtype (WT) counterparts. In addition, CAR NK cells and CAR T cells also mediated increased survival in the RMAERB tumor model. When challenged with Her2 expressing tumors, F38 mice were shown to mount an effective immune response, resulting in tumor rejection and long-term survival. This was shown to be predominantly dependent on both CD8+ T cells and NK cells. However, macrophages and CD4+ T cells were also shown to contribute to this response. Overall, this study highlights the use of the vav-CAR mouse model as a unique tool to determine the anti-tumor function of various immune subsets, either alone or when acting alongside CAR T cells in adoptive immunotherapy.
Collapse
Affiliation(s)
- Carmen S M Yong
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia
| | - Liza B John
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia
| | - Christel Devaud
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia.,Institut de Recherche en Santé Digestive, Université de Toulouse, INPT, INRA, INSERM UMR1220, UPS, France
| | - Miles H Prince
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia
| | - Ricky W Johnstone
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia
| | - Joseph A Trapani
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia
| | - Phillip K Darcy
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia.,Department of Immunology, Monash University, Prahran Victoria, Australia
| | - Michael H Kershaw
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia.,Department of Immunology, Monash University, Prahran Victoria, Australia
| |
Collapse
|
32
|
Westwood JA, Ellis S, Danne J, Johnson C, Oorschot V, Ramm G, Tscharke DC, Davenport AJ, Whisstock JC, Darcy PK, Kershaw MH, Slaney CY. An ultrastructural investigation of tumors undergoing regression mediated by immunotherapy. Oncotarget 2017; 8:115215-115229. [PMID: 29383154 PMCID: PMC5777766 DOI: 10.18632/oncotarget.23215] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Accepted: 07/14/2017] [Indexed: 12/31/2022] Open
Abstract
While immunotherapy employing chimeric antigen receptor (CAR) T cells can be effective against a variety of tumor types, little is known about what happens within the tumor at an ultrastructural level during tumor regression. Here, we used transmission electron microscopy to investigate morphologic and cellular features of tumors responding to immunotherapy composed of adoptive transfer of dual-specific CAR T cells and a vaccine, supported by preconditioning irradiation and interleukin-2. Tumors responded rapidly, and large areas of cell death were apparent by 4 days after treatment. The pleomorphic and metabolically active nature of tumor cells and phagocytic activity of macrophages were apparent in electron microscopic images of tumors prior to treatment. Following treatment, morphologic features of various types of tumor cell death were observed, including apoptosis, paraptosis and necrosis. Large numbers of lipid droplets were evident in tumor cells undergoing apoptosis. Macrophages were the predominant leukocyte type infiltrating tumors before treatment. Macrophages decreased in frequency and number after treatment, whereas an increasing accumulation of neutrophils and T lymphocytes was observed following treatment. Phagocytic activity of macrophages and neutrophils was apparent, while T cells could be observed in close association with tumor cells with potential immunological synapses present. These observations highlight the cellular composition and ultrastructural appearance of tumors undergoing regression mediated by immunotherapy.
Collapse
Affiliation(s)
- Jennifer A Westwood
- Cancer Immunology Program, Peter MacCallum Cancer Center, Melbourne, Australia
| | - Sarah Ellis
- Cancer Immunology Program, Peter MacCallum Cancer Center, Melbourne, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Australia
| | - Jill Danne
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Australia
| | - Chad Johnson
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Australia
| | - Viola Oorschot
- Monash Ramaciotti Centre for Cryo Electron Microscopy, Monash University, Clayton, Australia
| | - Georg Ramm
- Monash Ramaciotti Centre for Cryo Electron Microscopy, Monash University, Clayton, Australia
| | - David C Tscharke
- John Curtin School of Medical Research, Australian National University, Canberra, Australia
| | | | - James C Whisstock
- The ARC Centre of Excellence in Advanced Molecular Imaging, Monash University, Melbourne, Australia
| | - Phillip K Darcy
- Cancer Immunology Program, Peter MacCallum Cancer Center, Melbourne, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Australia
| | - Michael H Kershaw
- Cancer Immunology Program, Peter MacCallum Cancer Center, Melbourne, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Australia
| | - Clare Y Slaney
- Cancer Immunology Program, Peter MacCallum Cancer Center, Melbourne, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Australia
| |
Collapse
|
33
|
Dai X, He Y, Yao W, Gao X. Establishment of a spontaneous metastasis tumor model for human ErbB-2 vaccine. Biomed Pharmacother 2017; 88:991-998. [DOI: 10.1016/j.biopha.2017.01.099] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Revised: 01/16/2017] [Accepted: 01/16/2017] [Indexed: 12/13/2022] Open
|
34
|
Mardiana S, John LB, Henderson MA, Slaney CY, von Scheidt B, Giuffrida L, Davenport AJ, Trapani JA, Neeson PJ, Loi S, Haynes NM, Kershaw MH, Beavis PA, Darcy PK. A Multifunctional Role for Adjuvant Anti-4-1BB Therapy in Augmenting Antitumor Response by Chimeric Antigen Receptor T Cells. Cancer Res 2017; 77:1296-1309. [PMID: 28082401 DOI: 10.1158/0008-5472.can-16-1831] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Revised: 11/23/2016] [Accepted: 12/12/2016] [Indexed: 11/16/2022]
Abstract
Adoptive immunotherapy utilizing chimeric antigen receptor (CAR) T cells has demonstrated high success rates in hematologic cancers, but results against solid malignancies have been limited to date, due in part to the immunosuppressive tumor microenvironment. Activation of the 4-1BB (CD137) pathway using an agonistic α-4-1BB antibody is known to provide strong costimulatory signals for augmenting and diversifying T-cell responses. We therefore hypothesized that a combination of α-4-1BB and CAR T-cell therapy would result in improved antitumor responses. Using a human-Her2 self-antigen mouse model, we report here that α-4-1BB significantly enhanced CAR T-cell efficacy directed against the Her2 antigen in two different established solid tumor settings. Treatment also increased the expression of IFNγ and the proliferation marker Ki67 in tumor-infiltrating CAR T cells when combined with α-4-1BB. Strikingly, α-4-1BB significantly reduced host immunosuppressive cells at the tumor site, including regulatory T cells and myeloid-derived suppressor cells, correlating with an increased therapeutic response. We conclude that α-4-1BB has a multifunctional role for enhancing CAR T-cell responses and that this combination therapy has high translational potential, given current phase I/II clinical trials with α-4-1BB against various types of cancer. Cancer Res; 77(6); 1296-309. ©2017 AACR.
Collapse
Affiliation(s)
- Sherly Mardiana
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Australia
| | - Liza B John
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Australia
| | - Melissa A Henderson
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Australia
| | - Clare Y Slaney
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Australia
| | - Bianca von Scheidt
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Australia
| | - Lauren Giuffrida
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Australia
| | - Alexander J Davenport
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Australia
| | - Joseph A Trapani
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Australia
| | - Paul J Neeson
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Australia
| | - Sherene Loi
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Australia
| | - Nicole M Haynes
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Australia.,Cancer Therapeutics Program, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Michael H Kershaw
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Australia.,Department of Pathology, The University of Melbourne, Melbourne, Australia.,Department of Immunology, Monash University, Melbourne, Australia
| | - Paul A Beavis
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Australia. .,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Australia
| | - Phillip K Darcy
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Australia. .,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Australia.,Department of Pathology, The University of Melbourne, Melbourne, Australia.,Department of Immunology, Monash University, Melbourne, Australia
| |
Collapse
|
35
|
Slaney CY, von Scheidt B, Davenport AJ, Beavis PA, Westwood JA, Mardiana S, Tscharke DC, Ellis S, Prince HM, Trapani JA, Johnstone RW, Smyth MJ, Teng MW, Ali A, Yu Z, Rosenberg SA, Restifo NP, Neeson P, Darcy PK, Kershaw MH. Dual-specific Chimeric Antigen Receptor T Cells and an Indirect Vaccine Eradicate a Variety of Large Solid Tumors in an Immunocompetent, Self-antigen Setting. Clin Cancer Res 2016; 23:2478-2490. [PMID: 27965307 DOI: 10.1158/1078-0432.ccr-16-1860] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Revised: 11/22/2016] [Accepted: 11/30/2016] [Indexed: 11/16/2022]
Abstract
Purpose: While adoptive transfer of T cells bearing a chimeric antigen receptor (CAR) can eliminate substantial burdens of some leukemias, the ultimate challenge remains the eradication of large solid tumors for most cancers. We aimed to develop an immunotherapy approach effective against large tumors in an immunocompetent, self-antigen preclinical mouse model.Experimental Design: In this study, we generated dual-specific T cells expressing both a CAR specific for Her2 and a TCR specific for the melanocyte protein (gp100). We used a regimen of adoptive cell transfer incorporating vaccination (ACTIV), with recombinant vaccinia virus expressing gp100, to treat a range of tumors including orthotopic breast tumors and large liver tumors.Results: ACTIV therapy induced durable complete remission of a variety of Her2+ tumors, some in excess of 150 mm2, in immunocompetent mice expressing Her2 in normal tissues, including the breast and brain. Vaccinia virus induced extensive proliferation of T cells, leading to massive infiltration of T cells into tumors. Durable tumor responses required the chemokine receptor CXCR3 and exogenous IL2, but were independent of IFNγ. Mice were resistant to tumor rechallenge, indicating immune memory involving epitope spreading. Evidence of limited neurologic toxicity was observed, associated with infiltration of cerebellum by T cells, but was only transient.Conclusions: This study supports a view that it is possible to design a highly effective combination immunotherapy for solid cancers, with acceptable transient toxicity, even when the target antigen is also expressed in vital tissues. Clin Cancer Res; 23(10); 2478-90. ©2016 AACR.
Collapse
Affiliation(s)
- Clare Y Slaney
- Cancer Immunology Program, Peter MacCallum Cancer Center, Melbourne, Victoria, Australia. .,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia
| | - Bianca von Scheidt
- Cancer Immunology Program, Peter MacCallum Cancer Center, Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia
| | - Alexander J Davenport
- Cancer Immunology Program, Peter MacCallum Cancer Center, Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia
| | - Paul A Beavis
- Cancer Immunology Program, Peter MacCallum Cancer Center, Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia
| | - Jennifer A Westwood
- Cancer Immunology Program, Peter MacCallum Cancer Center, Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia
| | - Sherly Mardiana
- Cancer Immunology Program, Peter MacCallum Cancer Center, Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia
| | - David C Tscharke
- John Curtin School of Medical Research, Australian National University, Canberra, Australia
| | - Sarah Ellis
- Cancer Immunology Program, Peter MacCallum Cancer Center, Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia
| | - H Miles Prince
- Cancer Immunology Program, Peter MacCallum Cancer Center, Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia
| | - Joseph A Trapani
- Cancer Immunology Program, Peter MacCallum Cancer Center, Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia
| | - Ricky W Johnstone
- Cancer Immunology Program, Peter MacCallum Cancer Center, Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia
| | - Mark J Smyth
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Michele W Teng
- Cancer Immunoregulation and Immunotherapy Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Aesha Ali
- Cancer Immunology Program, Peter MacCallum Cancer Center, Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia
| | - Zhiya Yu
- Center for Cancer Research, National Cancer Institute, National Institute of Health, Bethesda, Maryland
| | - Steven A Rosenberg
- Center for Cancer Research, National Cancer Institute, National Institute of Health, Bethesda, Maryland
| | - Nicholas P Restifo
- Center for Cancer Research, National Cancer Institute, National Institute of Health, Bethesda, Maryland
| | - Paul Neeson
- Cancer Immunology Program, Peter MacCallum Cancer Center, Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia
| | - Phillip K Darcy
- Cancer Immunology Program, Peter MacCallum Cancer Center, Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia.,Department of Immunology, Monash University, Clayton, Australia
| | - Michael H Kershaw
- Cancer Immunology Program, Peter MacCallum Cancer Center, Melbourne, Victoria, Australia. .,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia.,Department of Immunology, Monash University, Clayton, Australia
| |
Collapse
|
36
|
Fry EA, Taneja P, Inoue K. Clinical applications of mouse models for breast cancer engaging HER2/neu. INTEGRATIVE CANCER SCIENCE AND THERAPEUTICS 2016; 3:593-603. [PMID: 28133539 PMCID: PMC5267336 DOI: 10.15761/icst.1000210] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Human c-ErbB2 (HER2) has long been used as a marker of breast cancer (BC) for sub-categorization for the prediction of prognosis, and determination of therapeutic strategies. HER2 overexpressing BCs are more invasive/metastatic; but patients respond to monoclonal antibody therapy with trastuzumab or tyrosine kinase inhibitors, at least at early stages. To date, numerous mouse models that faithfully reproduce HER2(+) BCs have been created in mice. We recently reviewed different mouse models of BC overexpressing wild type or mutant neu driven by MMTV, neu, or doxycycline-inducible promoters. These mice have been used to demonstrate the histopathology, oncogenic signaling pathways initiated by aberrant overexpression of HER2 in the mammary epithelium, and interaction between oncogenes and tumor suppressor genes at molecular levels. In this review, we focus on their clinical applications. They can be used to test the efficacy of HER(2) inhibitors before starting clinical trials, characterize the tumor-initiating cells that could be the cause of relapse after therapy as well as to analyze the molecular mechanisms of therapeutic resistance targeting HER2. MMTV-human ErbB2 (HER2) mouse models have recently been established since the monoclonal antibody to HER2 (trastuzumab; Herceptin®) does not recognize the rat neu protein. It has been reported that early intervention with HER2 monoclonal antibody would be beneficial for preventing mammary carcinogenesis. MDA-7/IL-24 as well as naturally-occurring chemicals have also been tested using MMTV-neu models. Recent studies have shown that MMTV-neu models are useful to develop vaccines to HER2 for immunotherapy. The mouse models employing HER2/neu will be essential for future antibody or drug screenings to overcome resistance to trastuzumab or HER(2)-specific tyrosine kinase inhibitors.
Collapse
Affiliation(s)
- Elizabeth A. Fry
- The Department of Pathology, Wake Forest University Health Sciences, Medical Center Boulevard, Winston-Salem, NC 27157 USA
| | - Pankaj Taneja
- Department of Biotechnology, Sharda University, Knowledge Park III, Greater Noida 201306, India
| | - Kazushi Inoue
- The Department of Pathology, Wake Forest University Health Sciences, Medical Center Boulevard, Winston-Salem, NC 27157 USA
| |
Collapse
|
37
|
Gross G, Eshhar Z. Therapeutic Potential of T Cell Chimeric Antigen Receptors (CARs) in Cancer Treatment: Counteracting Off-Tumor Toxicities for Safe CAR T Cell Therapy. Annu Rev Pharmacol Toxicol 2016; 56:59-83. [PMID: 26738472 DOI: 10.1146/annurev-pharmtox-010814-124844] [Citation(s) in RCA: 108] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
A chimeric antigen receptor (CAR) is a recombinant fusion protein combining an antibody-derived targeting fragment with signaling domains capable of activating T cells. Recent early-phase clinical trials have demonstrated the remarkable ability of CAR-modified T cells to eliminate B cell malignancies. This review describes the choice of target antigens and CAR manipulations to maximize antitumor specificity. Benefits and current limitations of CAR-modified T cells are discussed, with a special focus on the distribution of tumor antigens on normal tissues and the risk of on-target, off-tumor toxicities in the clinical setting. We present current methodologies for pre-evaluating these risks and review the strategies for counteracting potential off-tumor effects. Successful implementation of these approaches will improve the safety and efficacy of CAR T cell therapy and extend the range of cancer patients who may be treated.
Collapse
Affiliation(s)
- Gideon Gross
- Laboratory of Immunology, MIGAL, Galilee Research Institute, Kiryat Shmona 11016, Israel; .,Department of Biotechnology, Tel-Hai College, Upper Galilee 12210, Israel.,Center of Cancer Research, Tel Aviv Sourasky Medical Center, Tel Aviv 64239, Israel
| | - Zelig Eshhar
- Center of Cancer Research, Tel Aviv Sourasky Medical Center, Tel Aviv 64239, Israel.,Department of Immunology, The Weizmann Institute of Science, Rehovot 76100, Israel;
| |
Collapse
|
38
|
Yong CSM, Westwood JA, Schröder J, Papenfuss AT, von Scheidt B, Moeller M, Devaud C, Darcy PK, Kershaw MH. Expression of a Chimeric Antigen Receptor in Multiple Leukocyte Lineages in Transgenic Mice. PLoS One 2015; 10:e0140543. [PMID: 26505904 PMCID: PMC4624721 DOI: 10.1371/journal.pone.0140543] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 08/24/2015] [Indexed: 12/25/2022] Open
Abstract
Genetically modified CD8+ T lymphocytes have shown significant anti-tumor effects in the adoptive immunotherapy of cancer, with recent studies highlighting a potential role for a combination of other immune subsets to enhance these results. However, limitations in present genetic modification techniques impose difficulties in our ability to fully explore the potential of various T cell subsets and assess the potential of other leukocytes armed with chimeric antigen receptors (CARs). To address this issue, we generated a transgenic mouse model using a pan-hematopoietic promoter (vav) to drive the expression of a CAR specific for a tumor antigen. Here we present a characterization of the immune cell compartment in two unique vav-CAR transgenic mice models, Founder 9 (F9) and Founder 38 (F38). We demonstrate the vav promoter is indeed capable of driving the expression of a CAR in cells from both myeloid and lymphoid lineage, however the highest level of expression was observed in T lymphocytes from F38 mice. Lymphoid organs in vav-CAR mice were smaller and had reduced cell numbers compared to the wild type (WT) controls. Furthermore, the immune composition of F9 mice differed greatly with a significant reduction in lymphocytes found in the thymus, lymph node and spleen of these mice. To gain insight into the altered immune phenotype of F9 mice, we determined the chromosomal integration site of the transgene in both mouse strains using whole genome sequencing (WGS). We demonstrated that compared to the 7 copies found in F38 mice, F9 mice harbored almost 270 copies. These novel vav-CAR models provide a ready source of CAR expressing myeloid and lymphoid cells and will aid in facilitating future experiments to delineate the role for other leukocytes for adoptive immunotherapy against cancer.
Collapse
Affiliation(s)
- Carmen S. M. Yong
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Jennifer A. Westwood
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Jan Schröder
- Bioinformatics Division, The Walter & Eliza Hall Institute of Medical Research, Parkville, Victoria, 3052, Australia
- Bioinformatics and Cancer Genomics, Peter MacCallum Cancer Centre, East Melbourne, Victoria, 3002, Australia
- Department of Computing and Information Systems, The University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Anthony T. Papenfuss
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria 3010, Australia
- Bioinformatics Division, The Walter & Eliza Hall Institute of Medical Research, Parkville, Victoria, 3052, Australia
- Bioinformatics and Cancer Genomics, Peter MacCallum Cancer Centre, East Melbourne, Victoria, 3002, Australia
- Department of Computing and Information Systems, The University of Melbourne, Parkville, Victoria, 3010, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Bianca von Scheidt
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Maria Moeller
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Christel Devaud
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria 3010, Australia
- INSERM U1043 Centre de Physiopathologie Toulouse Purpan (CPTP), Toulouse, France
- * E-mail: (MK); (PD); (CD)
| | - Phillip K. Darcy
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria 3010, Australia
- Department of Immunology, Monash University, Prahran Victoria 3181 Australia
- * E-mail: (MK); (PD); (CD)
| | - Michael H. Kershaw
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria 3010, Australia
- Department of Immunology, Monash University, Prahran Victoria 3181 Australia
- * E-mail: (MK); (PD); (CD)
| |
Collapse
|
39
|
Embryonic Lethality in Homozygous Human Her-2 Transgenic Mice Due to Disruption of the Pds5b Gene. PLoS One 2015; 10:e0136817. [PMID: 26334628 PMCID: PMC4559457 DOI: 10.1371/journal.pone.0136817] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Accepted: 08/07/2015] [Indexed: 12/29/2022] Open
Abstract
The development of antigen-targeted therapeutics is dependent on the preferential expression of tumor-associated antigens (TAA) at targetable levels on the tumor. Tumor-associated antigens can be generated de novo or can arise from altered expression of normal basal proteins, such as the up-regulation of human epidermal growth factor receptor 2 (Her2/ErbB2). To properly assess the development of Her2 therapeutics in an immune tolerant model, we previously generated a transgenic mouse model in which expression of the human Her2 protein was present in both the brain and mammary tissue. This mouse model has facilitated the development of Her2 targeted therapies in a clinically relevant and suitable model. While heterozygous Her2+/- mice appear to develop in a similar manner to wild type mice (Her2-/-), it has proven difficult to generate homozygous Her2+/+ mice, potentially due to embryonic lethality. In this study, we performed whole genome sequencing to determine if the integration site of the Her2 transgene was responsible for this lethality. Indeed, we report that the Her2 transgene had integrated into the Pds5b (precocious dissociation of sisters) gene on chromosome 5, as a 162 copy concatemer. Furthermore, our findings demonstrate that Her2+/+ mice, similar to Pds5b-/- mice, are embryonic lethal and confirm the necessity for Pds5b in embryonic development. This study confirms the value of whole genome sequencing in determining the integration site of transgenes to gain insight into associated phenotypes.
Collapse
|
40
|
Wei WZ, Jones RF, Juhasz C, Gibson H, Veenstra J. Evolution of animal models in cancer vaccine development. Vaccine 2015; 33:7401-7407. [PMID: 26241945 DOI: 10.1016/j.vaccine.2015.07.075] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Accepted: 07/02/2015] [Indexed: 12/29/2022]
Abstract
Advances in cancer vaccine development are facilitated by animal models reflecting key features of human cancer and its interface with host immunity. Several series of transplantable preneoplastic and neoplastic mouse mammary lesions have been used to delineate mechanisms of anti-tumor immunity. Mimicking immune tolerance to tumor-associated antigens (TAA) such as HER2/neu, transgenic mice developing spontaneous mammary tumors are strong model systems for pre-clinical vaccine testing. In these models, HER2 DNA vaccines are easily administered, well-tolerated, and induce both humoral and cellular immunity. Although engineered mouse strains have advanced cancer immunotherapy, basic shortcomings remain. For example, multiple mouse strains have to be tested to recapitulate genetic regulation of immune tolerance in humans. Outbred domestic felines more closely parallel humans in the natural development of HER2 positive breast cancer and their varying genetic background. Electrovaccination with heterologous HER2 DNA induces robust adaptive immune responses in cats. Importantly, homologous feline HER2 DNA with a single amino acid substitution elicits unique antibodies to feline mammary tumor cells, unlocking a new vaccine principle. As an alternative approach to targeted vaccination, non-surgical tumor ablation such as cryoablation induces anti-tumor immunity via in situ immunization, particularly when combined with toll-like receptor (TLR) agonist. As strategies for vaccination advance, non-invasive monitoring of host response becomes imperative. As an example, magnetic resonance imaging (MRI) and positron emission tomography (PET) scanning following administration of tryptophan metabolism tracer [11C]-alpha-methyl-tryptophan (AMT) provides non-invasive imaging of both tumor growth and metabolic activities. Because AMT is a substrate of indoleamine-pyrrole 2,3-dioxygenase (IDO), an enzyme that produces the immune regulatory molecule kynurenine, AMT imaging can provide novel insight of host response. In conclusion, new feline models improve the predictive power of cancer immunotherapy and real-time PET imaging enables mechanistic monitoring of host immunity. Strategic utilization of these new tools will expedite cancer vaccine development.
Collapse
Affiliation(s)
- Wei-Zen Wei
- Karmanos Cancer Institute, Wayne State University, Detroit, MI 48201, United States.
| | - Richard F Jones
- Karmanos Cancer Institute, Wayne State University, Detroit, MI 48201, United States
| | - Csaba Juhasz
- Karmanos Cancer Institute, Wayne State University, Detroit, MI 48201, United States
| | - Heather Gibson
- Karmanos Cancer Institute, Wayne State University, Detroit, MI 48201, United States
| | - Jesse Veenstra
- Karmanos Cancer Institute, Wayne State University, Detroit, MI 48201, United States
| |
Collapse
|
41
|
Gibson HM, Veenstra JJ, Jones R, Vaishampayan U, Sauerbrey M, Bepler G, Lum L, Reyes J, Weise A, Wei WZ. Induction of HER2 Immunity in Outbred Domestic Cats by DNA Electrovaccination. Cancer Immunol Res 2015; 3:777-86. [PMID: 25711535 PMCID: PMC4491035 DOI: 10.1158/2326-6066.cir-14-0175] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Accepted: 02/14/2015] [Indexed: 12/20/2022]
Abstract
Domestic cats share human living environments and genetic traits. They develop spontaneous feline mammary carcinoma (FMC) with similar histopathology to human breast cancer. HER2 and AKT phosphorylation was demonstrated in primary FMC by immunoblot analysis, indicating HER2 as a therapeutic target. FMC lines K12 and K248 expressing HER1, HER2, and HER3 were sensitive to receptor tyrosine kinase (RTK) inhibitors gefitinib and lapatinib. To test HER2 vaccine response in cats, purpose-bred, healthy cats were electrovaccinated with heterologous (xenogeneic) or point-mutated feline HER2 DNA. T-cell reactivity to feline self-HER2 was detected in 4 of 10 cats that received bear HER2, human-rat fusion HER2 (E2Neu) or mutant feline HER2 (feHER2-K), which contains a single amino acid substitution. The variable T-cell responses may resemble that in the genetically heterogeneous human population. All immune sera to heterologous HER2 recognized feline HER2 expressed in 3T3 cells (3T3/HER2), but not that in FMC K12 or K248. Immune sera to mutant pfeHER2-K bound 3T3/HER2 cells weakly, but they showed better recognition of K12 and K248 cells that also express HER1 and HER3, suggesting distinct HER2 epitopes displayed by FMC that may be simulated by feHER2-K. In summary, HER2 DNA electroporation overcomes T-cell immune tolerance in approximately 40% of healthy cats and induces antibodies with distinct specificity. Vaccination studies in domestic cats can expedite vaccine iteration to guide human vaccine design and better predict outcome, with the added benefit of helping feline mammary tumor patients.
Collapse
Affiliation(s)
- Heather M Gibson
- Karmanos Cancer Institute, Wayne State University, Detroit, Michigan
| | - Jesse J Veenstra
- Karmanos Cancer Institute, Wayne State University, Detroit, Michigan
| | - Richard Jones
- Karmanos Cancer Institute, Wayne State University, Detroit, Michigan
| | - Ulka Vaishampayan
- Karmanos Cancer Institute, Wayne State University, Detroit, Michigan
| | | | - Gerold Bepler
- Karmanos Cancer Institute, Wayne State University, Detroit, Michigan
| | - Lawrence Lum
- Karmanos Cancer Institute, Wayne State University, Detroit, Michigan
| | - Joyce Reyes
- Karmanos Cancer Institute, Wayne State University, Detroit, Michigan
| | - Amy Weise
- Karmanos Cancer Institute, Wayne State University, Detroit, Michigan
| | - Wei-Zen Wei
- Karmanos Cancer Institute, Wayne State University, Detroit, Michigan.
| |
Collapse
|
42
|
Gene Gun Her2/neu DNA Vaccination: Evaluation of Vaccine Efficacy in a Syngeneic Her2/neu Mouse Tumor Model. Methods Mol Biol 2015; 1317:17-37. [PMID: 26072399 DOI: 10.1007/978-1-4939-2727-2_2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Genetic vaccination using naked plasmid DNA is an immunization strategy both against infectious diseases and cancer. In order to improve the efficacy of DNA vaccines, particularly in large animals and humans, different strategies have been pursued. These vaccination strategies are based on different application routes, schedules, and coexpression of immunomodulatory molecules as adjuvants. Our mouse tumor model offers the possibility to investigate Her2/neu DNA vaccines in different settings, i.e., intramuscular or intradermal application with or without coexpression of adjuvants. Protection from tumor growth in tumor challenge experiments and both T cell and humoral immune responses against Her2/neu peptides are used as surrogate parameters for vaccine efficacy.
Collapse
|
43
|
Liu H, Yang B, Sun T, Lin L, Hu Y, Deng M, Yang J, Liu T, Li J, Sun S, Jiao S. Specific growth inhibition of ErbB2‑expressing human breast cancer cells by genetically modified NK‑92 cells. Oncol Rep 2014; 33:95-102. [PMID: 25333815 DOI: 10.3892/or.2014.3548] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2014] [Accepted: 03/10/2014] [Indexed: 11/06/2022] Open
Abstract
The natural killer cell line NK‑92 shows great cytotoxicity against various types of cancer. Several types of solid tumor cells, however, can effectively resist NK-mediated lysis by interaction of major histocompatibility complex (MHC) molecules with NK cell inhibitory receptors. To generate a eukaryotic expression vector encoding chimeric antigen receptor scFv anti-erbB2-CD28-ζ and to investigate the expression and action of this chimeric antigen receptor in cancer cells both in vitro and in vivo, NK‑92 cells were genetically modified with an scFv anti-erbB2-CD28-ζ chimeric recep-tor by optimized electro-poration using the Amaxa Nucleofector system. The expression of the chimeric receptor was evaluated by RT-PCR and immunofluorescence. The ability of the genetically modified NK‑92 cells to induce cell death in tumor targets was assessed in vitro and in vivo. The transduced NK‑92-anti-erbB2 scFv-CD28-ζ cells expressing high levels of the fusion protein on the cell surface were analyzed by fluorescence-activated cell-sorting (FACS) analysis. These cells specifically enhanced the cell death of the erbB2‑expressing human breast cancer cell lines MDA-MB-453 and SKBr3. Furthermore, adoptive transfer of genetically modified NK‑92 cells specifically reduced tumor size and lung metastasis of nude mice bearing established MDA-MB-453 cells, and significantly enhanced the survival period of these mice. The genetically modified NK‑92 cells significantly enhanced the killing of erbB2‑expressing cancer and may be a novel therapeutic strategy for erbB2‑expressing cancer cells.
Collapse
Affiliation(s)
- Hui Liu
- Department of Internal Oncology, 301 General Hospital, Beijing, P.R. China
| | - Bo Yang
- Department of Internal Oncology, 301 General Hospital, Beijing, P.R. China
| | - Tingting Sun
- Department of Internal Neurology, Hainan Branch of 301 General Hospital, Hainan, P.R. China
| | - Lin Lin
- Department of Internal Oncology, 301 General Hospital, Beijing, P.R. China
| | - Yi Hu
- Department of Internal Oncology, 301 General Hospital, Beijing, P.R. China
| | - Muhong Deng
- Department of Internal Oncology, 301 General Hospital, Beijing, P.R. China
| | - Junlan Yang
- Department of Internal Oncology, 301 General Hospital, Beijing, P.R. China
| | - Tianyi Liu
- Tumor Central Laboratory, 301 General Hospital, Beijing, P.R. China
| | - Jinyu Li
- Department of Internal Oncology, 301 General Hospital, Beijing, P.R. China
| | - Shengjie Sun
- Department of Internal Oncology, 301 General Hospital, Beijing, P.R. China
| | - Shunchang Jiao
- Department of Internal Oncology, 301 General Hospital, Beijing, P.R. China
| |
Collapse
|
44
|
Castagnoli L, Iezzi M, Ghedini GC, Ciravolo V, Marzano G, Lamolinara A, Zappasodi R, Gasparini P, Campiglio M, Amici A, Chiodoni C, Palladini A, Lollini PL, Triulzi T, Menard S, Nanni P, Tagliabue E, Pupa SM. Activated d16HER2 Homodimers and SRC Kinase Mediate Optimal Efficacy for Trastuzumab. Cancer Res 2014; 74:6248-59. [DOI: 10.1158/0008-5472.can-14-0983] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
45
|
Cavallo F, Aurisicchio L, Mancini R, Ciliberto G. Xenogene vaccination in the therapy of cancer. Expert Opin Biol Ther 2014; 14:1427-42. [DOI: 10.1517/14712598.2014.927433] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
46
|
Aurisicchio L, Peruzzi D, Koo G, Wei WZ, La Monica N, Ciliberto G. Immunogenicity and therapeutic efficacy of a dual-component genetic cancer vaccine cotargeting carcinoembryonic antigen and HER2/neu in preclinical models. Hum Gene Ther 2014; 25:121-31. [PMID: 24195644 PMCID: PMC3922413 DOI: 10.1089/hum.2013.103] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2013] [Accepted: 11/05/2013] [Indexed: 12/18/2022] Open
Abstract
Several cancer vaccine efforts have been directed to simultaneously cotarget multiple tumor antigens, with the intent to achieve broader immune responses and more effective control of cancer growth. Genetic cancer vaccines based on in vivo muscle electro-gene-transfer of plasmid DNA (DNA-EGT) and adenoviral vectors represent promising modalities to elicit powerful immune responses against tumor-associated antigens (TAAs) such as carcinoembryonic antigen (CEA) and human epidermal growth factor receptor-2 (HER2)/neu. Combinations of these modalities of immunization (heterologous prime-boost) can induce superior immune reactions as compared with single-modality vaccines. We have generated a dual component-dual target genetic cancer vaccine consisting of a DNA moiety containing equal amounts of two plasmids, one encoding the extracellular and transmembrane domains of HER2 (ECD.TM) and the other encoding CEA fused to the B subunit of Escherichia coli heat-labile toxin (LTB), and of an adenoviral subtype 6 dicistronic vector carrying the same two tumor antigens gene constructs. The CEA/HER2 vaccine was tested in two different CEA/HER2 double-transgenic mouse models and in NOD/scid-DR1 mice engrafted with the human immune system. The immune response was measured by enzyme-linked immunospot assay, flow cytometry, and ELISA. The CEA/HER2 vaccine was able to break immune tolerance against both antigens. Induction of a T cell and antibody immune response was detected in immune-tolerant mice. Most importantly, the vaccine was able to slow the growth of HER2/neu⁺ and CEA⁺ tumors. A significant T cell response was measured in NOD/scid-DR1 mice engrafted with human cord blood cells. In conclusion, the CEA/HER2 genetic vaccine was immunogenic and able to confer significant therapeutic effects. These data warrant the evaluation of this vaccination strategy in human clinical trials.
Collapse
Affiliation(s)
- Luigi Aurisicchio
- Istituto di Ricerche di Biologia Molecolare (IRBM), 00040 Pomezia, Rome, Italy
| | - Daniela Peruzzi
- Istituto di Ricerche di Biologia Molecolare (IRBM), 00040 Pomezia, Rome, Italy
| | - Gloria Koo
- Memorial Sloan-Kettering Cancer Center, New York, NY 10021
| | - Wei-Zen Wei
- Karmanos Cancer Institute, Wayne State University, Detroit, MI 48201
| | - Nicola La Monica
- Istituto di Ricerche di Biologia Molecolare (IRBM), 00040 Pomezia, Rome, Italy
| | - Gennaro Ciliberto
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), National Cancer Institute G. Pascale, 80131 Naples, Italy
| |
Collapse
|
47
|
De Giovanni C, Nicoletti G, Quaglino E, Landuzzi L, Palladini A, Ianzano ML, Dall'Ora M, Grosso V, Ranieri D, Laranga R, Croci S, Amici A, Penichet ML, Iezzi M, Cavallo F, Nanni P, Lollini PL. Vaccines against human HER2 prevent mammary carcinoma in mice transgenic for human HER2. Breast Cancer Res 2014; 16:R10. [PMID: 24451168 PMCID: PMC3979148 DOI: 10.1186/bcr3602] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2013] [Accepted: 01/13/2014] [Indexed: 01/26/2023] Open
Abstract
Introduction The availability of mice transgenic for the human HER2 gene (huHER2) and prone to the development of HER2-driven mammary carcinogenesis (referred to as FVB-huHER2 mice) prompted us to study active immunopreventive strategies targeting the human HER2 molecule in a tolerant host. Methods FVB-huHER2 mice were vaccinated with either IL-12-adjuvanted human HER2-positive cancer cells or DNA vaccine carrying chimeric human-rat HER2 sequences. Onset and number of mammary tumors were recorded to evaluate vaccine potency. Mice sera were collected and passively transferred to xenograft-bearing mice to assess their antitumor efficacy. Results Both cell and DNA vaccines significantly delayed tumor onset, leading to about 65% tumor-free mice at 70 weeks, whereas mock-vaccinated FVB-huHER2 controls developed mammary tumors at a median age of 45 weeks. In the DNA vaccinated group, 65% of mice were still tumor-free at about 90 weeks of age. The number of mammary tumors per mouse was also significantly reduced in vaccinated mice. Vaccines broke the immunological tolerance to the huHER2 transgene, inducing both humoral and cytokine responses. The DNA vaccine mainly induced a high and sustained level of anti-huHER2 antibodies, the cell vaccine also elicited interferon (IFN)-γ production. Sera of DNA-vaccinated mice transferred to xenograft-carrying mice significantly inhibited the growth of human HER2-positive cancer cells. Conclusions Anti-huHER2 antibodies elicited in the tolerant host exert antitumor activity.
Collapse
|
48
|
Pavlenko M, Leder C, Pisa P. Plasmid DNA vaccines against cancer: cytotoxic T-lymphocyte induction against tumor antigens. Expert Rev Vaccines 2014; 4:315-27. [PMID: 16026247 DOI: 10.1586/14760584.4.3.315] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
In recent years, a number of tumor vaccination strategies have been developed. Most of these rely on the identification of tumor antigens that can be recognized by the immune system. DNA vaccination represents one such approach for the induction of both humoral and cellular immune responses against tumor antigens. Studies in animal models have demonstrated the feasibility of utilizing DNA vaccination to elicit protective antitumor immune responses. However, most tumor antigens expressed by cancer cells in humans are weakly immunogenic, and therefore require the development of strategies to potentiate DNA vaccine efficacy in the clinical setting. This review focuses on recent advances in understanding of the immunology of DNA vaccines, as well as strategies used to increase DNA vaccine potency with respect to cytotoxic T-lymphocyte activity.
Collapse
Affiliation(s)
- Maxim Pavlenko
- Department of Oncology and Pathology, Cancer Center Karolinska, Karolinska Institute, Stockholm S-171 76, Sweden.
| | | | | |
Collapse
|
49
|
Riccardo F, Bolli E, Macagno M, Arigoni M, Cavallo F, Quaglino E. Chimeric DNA Vaccines: An Effective Way to Overcome Immune Tolerance. Curr Top Microbiol Immunol 2014; 405:99-122. [PMID: 25294003 DOI: 10.1007/82_2014_426] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The fact that cancer immunotherapy is considered to be a safe and successful weapon for use in combination with surgery, radiation, and chemotherapy treatments means that it has recently been chosen as Breakthrough of the Year 2013 by Science editors. Anticancer vaccines have been extensively tested, in this field, both in preclinical cancer models and in the clinic. However, tumor-associated antigens (TAAs) are often self-tolerated molecules and cancer patients suffer from strong immunosuppressive effects, meaning that the triggering of an effective anti-tumor immune response is difficult. One possible means to overcome immunological tolerance to self-TAAs is of course the use of vaccines that code for xenogeneic proteins. However, a low-affinity antibody response against the self-homologous protein expressed by cancer cells is generally induced by xenovaccination. This issue becomes extremely limiting when working with tumors in which the contribution of the humoral rather than the cellular immune response is required if tumor growth is to be hampered. A possible way to avoid this problem is to use hybrid vaccines which code for chimeric proteins that include both homologous and xenogeneic moieties. In fact, a superior protective anti-tumor immune response against ErbB2+ transplantable and autochthonous mammary tumors was observed over plasmids that coded for the fully rat or fully human proteins when hybrid plasmids that coded for chimeric rat/human ErbB2 protein were tested in ErbB2 transgenic mice. In principle, these findings may become the basis for a new rational means of designing effective vaccines against TAAs.
Collapse
Affiliation(s)
- Federica Riccardo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, via Nizza 52, 10126, Torino, Italy
| | - Elisabetta Bolli
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, via Nizza 52, 10126, Torino, Italy
| | - Marco Macagno
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, via Nizza 52, 10126, Torino, Italy
| | - Maddalena Arigoni
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, via Nizza 52, 10126, Torino, Italy
| | - Federica Cavallo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, via Nizza 52, 10126, Torino, Italy
| | - Elena Quaglino
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, via Nizza 52, 10126, Torino, Italy.
| |
Collapse
|
50
|
Wang SH, Lu L, Fan Y, Wicha MS, Cao Z, Chang AE, Xia JC, Baker JR, Li Q. Characterization of a novel transgenic mouse tumor model for targeting HER2+ cancer stem cells. Int J Biol Sci 2013; 10:25-32. [PMID: 24391448 PMCID: PMC3879588 DOI: 10.7150/ijbs.6309] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2013] [Accepted: 10/06/2013] [Indexed: 01/03/2023] Open
Abstract
HER2 is an oncogenic tumor-associated antigen overexpressed in 20-25% of breast cancers, which is associated with increased invasion, metastasis of the disease and resistance to therapy. Recent studies have further shown that HER2 can increase the population of breast cancer stem cells (BCSCs). However, there is currently no in vivo model for the study of HER2+ BCSCs. In this study, we characterized a mouse breast cancer model for HER2+ BCSCs. This was accomplished by inoculating mouse mammary tumor EO771 cells engineered with human wild-type HER2 (EO771E2) into C57BL/6 HER2 transgenic mice to test and confirm the stable human HER2 expression in the model. More importantly, we detected a subpopulation of EO771E2 cells with a high activity of aldehyde dehydrogenases (ALDHhigh). We demonstrated that the isolated ALDHhigh EO771E2 cells possessed key properties of BCSCs including enhanced tumorigenicity, generation of heterogeneous tumors and the capacity to self-renewal in vitro. In conclusion, the tumors formed in C57BL/6 HER2 transgenic mice with EO771E2 cell injection revealed stable and functional human HER2 expression. These tumors contain a subset of ALDHhigh cells which are small in number, but are enriched in cancer stem cells. This model is deemed to be useful for experiments aimed to develop novel treatments to target HER2+ BCSCs.
Collapse
Affiliation(s)
- Su He Wang
- 1. Michigan Nanotechnology Institute for Medicine and Biological Sciences
| | - Lin Lu
- 2. University of Michigan Comprehensive Cancer Center, Ann Arbor, Michigan 48109, USA ; 3. State Key Laboratory of Oncology in Southern China and Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China
| | - Yongyi Fan
- 1. Michigan Nanotechnology Institute for Medicine and Biological Sciences
| | - Max S Wicha
- 2. University of Michigan Comprehensive Cancer Center, Ann Arbor, Michigan 48109, USA
| | - Zhengyi Cao
- 1. Michigan Nanotechnology Institute for Medicine and Biological Sciences
| | - Alfred E Chang
- 2. University of Michigan Comprehensive Cancer Center, Ann Arbor, Michigan 48109, USA
| | - Jian-chuan Xia
- 3. State Key Laboratory of Oncology in Southern China and Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China
| | - James R Baker
- 1. Michigan Nanotechnology Institute for Medicine and Biological Sciences
| | - Qiao Li
- 2. University of Michigan Comprehensive Cancer Center, Ann Arbor, Michigan 48109, USA
| |
Collapse
|