1
|
Rakhmilevich AL, Tsarovsky NW, Felder M, Zaborek J, Moram S, Erbe AK, Pieper AA, Spiegelman DV, Cheng EM, Witt CM, Overwijk WW, Morris ZS, Sondel PM. A combined radio-immunotherapy regimen eradicates late-stage tumors in mice. Front Immunol 2024; 15:1419773. [PMID: 39076988 PMCID: PMC11284032 DOI: 10.3389/fimmu.2024.1419773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 07/02/2024] [Indexed: 07/31/2024] Open
Abstract
Background The majority of experimental approaches for cancer immunotherapy are tested against relatively small tumors in tumor-bearing mice, because in most cases advanced cancers are resistant to the treatments. In this study, we asked if even late-stage mouse tumors can be eradicated by a rationally designed combined radio-immunotherapy (CRI) regimen. Methods CRI consisted of local radiotherapy, intratumoral IL-12, slow-release systemic IL-2 and anti- CTLA-4 antibody. Therapeutic effects of CRI against several weakly immunogenic and immunogenic mouse tumors including B78 melanoma, MC38 and CT26 colon carcinomas and 9464D neuroblastoma were evaluated. Immune cell depletion and flow cytometric analysis were performed to determine the mechanisms of the antitumor effects. Results Tumors with volumes of 2,000 mm3 or larger were eradicated by CRI. Flow analyses of the tumors revealed reduction of T regulatory (Treg) cells and increase of CD8/Treg ratios following CRI. Rapid shrinkage of the treated tumors did not require T cells, whereas T cells were involved in the systemic effect against the distant tumors. Cured mice developed immunological memory. Conclusions These findings underscore that rationally designed combination immunotherapy regimens can be effective even against large, late-stage tumors.
Collapse
Affiliation(s)
| | - Noah W. Tsarovsky
- Department of Human Oncology, University of Wisconsin-Madison, Madison, WI, United States
| | - Mildred Felder
- Department of Human Oncology, University of Wisconsin-Madison, Madison, WI, United States
| | - Jen Zaborek
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI, United States
| | - Sritha Moram
- Department of Human Oncology, University of Wisconsin-Madison, Madison, WI, United States
| | - Amy K. Erbe
- Department of Human Oncology, University of Wisconsin-Madison, Madison, WI, United States
| | - Alexander A. Pieper
- Department of Human Oncology, University of Wisconsin-Madison, Madison, WI, United States
| | - Dan V. Spiegelman
- Department of Human Oncology, University of Wisconsin-Madison, Madison, WI, United States
| | - Emily M. Cheng
- Department of Human Oncology, University of Wisconsin-Madison, Madison, WI, United States
| | - Cole M. Witt
- Department of Human Oncology, University of Wisconsin-Madison, Madison, WI, United States
| | | | - Zachary S. Morris
- Department of Human Oncology, University of Wisconsin-Madison, Madison, WI, United States
| | - Paul M. Sondel
- Department of Human Oncology, University of Wisconsin-Madison, Madison, WI, United States
- Department of Pediatrics, University of Wisconsin-Madison, Madison, WI, United States
| |
Collapse
|
2
|
Nguyen A, Brown D, Krishnan R, Bastin D, Deng L, Chen L, Salem O, Walsh SR, Bramson JL, Wan Y. HDACi-dependent Microenvironmental Normalization Overcomes Tumor Burden-induced T-cell Exhaustion. Clin Cancer Res 2023; 29:4289-4305. [PMID: 37561398 DOI: 10.1158/1078-0432.ccr-22-2181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 08/17/2022] [Accepted: 08/07/2023] [Indexed: 08/11/2023]
Abstract
PURPOSE T-cell exhaustion limits immunotherapy for the treatment of solid tumors. Although immune checkpoint blockade and adoptive T-cell therapy (ACT) can mediate tumor regression, their potency is often determined by tumor burden. Here, we identified tumor burden-related pathway changes that are conducive to T-cell exhaustion. We then determined whether microenvironmental reprogramming via epigenetic modulation could reverse T-cell exhaustion and improve immunotherapeutic responsiveness. EXPERIMENTAL DESIGN We developed a murine syngeneic tumor model wherein an increased burden ablated therapeutic responsiveness to ACT, which corresponded with systemic induction of T-cell exhaustion. Transcriptome analysis of these large tumors allowed us to characterize changes to immunosuppressive pathway expression during class I histone deacetylase inhibitor MS-275 treatment. We then measured the therapeutic impact of MS-275 during ACT and assessed T-cell exhaustion by transcriptome/phenotypic analysis. RESULTS ACT durably regressed small tumors but failed to control large tumors, which were associated with systemic T-cell exhaustion and ablation of T-cell responses. Large tumors were defined by an immunosuppressive pathway signature. MS-275 reversed this pathway signature and promoted durable regression of large tumors during ACT. Prototypical exhaustion marker Tim-3 was selectively upregulated in transferred T cells despite displaying a reduced exhaustion signature. Instead, we observed enhanced activation-dependent signaling correlating with enrichment of the IL2-STAT5 signaling axis. Activated CD8+ T-cell responses were predominantly skewed toward terminal effector cell-like CD44+ Tim-3hi TCF1- CD127- KLRG1+ differentiation. CONCLUSIONS Tumor burden-induced pathway changes can be reversed through epigenetic reprogramming, enabling the conversion from T-cell exhaustion to effector lineage differentiation.
Collapse
Affiliation(s)
- Andrew Nguyen
- Department of Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, Canada
| | - Dominique Brown
- Department of Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, Canada
| | - Ramya Krishnan
- Department of Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, Canada
| | - Donald Bastin
- Department of Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, Canada
| | - Li Deng
- Department of Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, Canada
| | - Lan Chen
- Department of Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, Canada
| | - Omar Salem
- Department of Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, Canada
| | - Scott R Walsh
- Department of Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, Canada
| | - Jonathan L Bramson
- Department of Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, Canada
| | - Yonghong Wan
- Department of Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, Canada
| |
Collapse
|
3
|
Dendritic cell-mimicking scaffolds for ex vivo T cell expansion. Bioact Mater 2023; 21:241-252. [PMID: 36157246 PMCID: PMC9474324 DOI: 10.1016/j.bioactmat.2022.08.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Revised: 08/10/2022] [Accepted: 08/23/2022] [Indexed: 11/20/2022] Open
Abstract
We propose an ex vivo T cell expansion system that mimics natural antigen-presenting cells (APCs) for adoptive cell therapy (ACT). Microfiber scaffolds coated with dendritic cell (DC) membrane replicate physicochemical properties of dendritic cells specific for T cell activation such as rapid recognition by T cells, long duration of T cell tethering, and DC-specific co-stimulatory cues. The DC membrane-coated scaffold is first surface-immobilized with T cell stimulatory ligands, anti-CD3 (αCD3) and anti-CD28 (αCD28) antibodies, followed by adsorption of releasable interleukin-2 (IL-2). The scaffolds present both surface and soluble cues to T cells ex vivo in the same way that these cues are presented by natural APCs in vivo. We demonstrate that the DC-mimicking scaffold promotes greater polyclonal expansion of primary human T cells as compared to αCD3/αCD28-functionalized Dynabead. More importantly, major histocompatibility complex molecules derived from the DC membrane of the scaffold allow antigen-specific T cell expansion with target cell-specific killing ability. In addition, most of the expanded T cells (∼97%) can be harvested from the scaffold by density gradient centrifugation. Overall, the DC-mimicking scaffold offers a scalable, modular, and customizable platform for rapid expansion of highly functional T cells for ACT. The scaffold mimics physicochemical properties of natural antigen-presenting cells. The scaffold presents T cell stimulatory cues as antigen-presenting cell does. This platform supports both polyclonal and antigen-specific T cell expansion. This platform offers a large-scale manufacturing system for adoptive cell therapy.
Collapse
|
4
|
Medrano RFV, Salles TA, Dariolli R, Antunes F, Feitosa VA, Hunger A, Catani JPP, Mendonça SA, Tamura RE, Lana MG, Rodrigues EG, Strauss BE. Potentiation of combined p19Arf and interferon-beta cancer gene therapy through its association with doxorubicin chemotherapy. Sci Rep 2022; 12:13636. [PMID: 35948616 PMCID: PMC9365852 DOI: 10.1038/s41598-022-17775-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 07/30/2022] [Indexed: 11/11/2022] Open
Abstract
Balancing safety and efficacy is a major consideration for cancer treatments, especially when combining cancer immunotherapy with other treatment modalities such as chemotherapy. Approaches that induce immunogenic cell death (ICD) are expected to eliminate cancer cells by direct cell killing as well as activation of an antitumor immune response. We have developed a gene therapy approach based on p19Arf and interferon-β gene transfer that, similar to conventional inducers of ICD, results in the release of DAMPS and immune activation. Here, aiming to potentiate this response, we explore whether association between our approach and treatment with doxorubicin (Dox), a known inducer of ICD, could further potentiate treatment efficacy without inducing cardiotoxicity, a critical side effect of Dox. Using central composite rotational design analysis, we show that cooperation between gene transfer and chemotherapy killed MCA205 and B16F10 cells and permitted the application of reduced viral and drug doses. The treatments also cooperated to induce elevated levels of ICD markers in MCA205, which correlated with improved efficacy of immunotherapy in vivo. Treatment of subcutaneous MCA205 tumors associating gene transfer and low dose (10 mg/kg) chemotherapy resulted in inhibition of tumor progression. Moreover, the reduced dose did not cause cardiotoxicity as compared to the therapeutic dose of Dox (20 mg/kg). The association of p19Arf/interferon-β gene transfer and Dox chemotherapy potentiated antitumor response and minimized cardiotoxicity.
Collapse
Affiliation(s)
- Ruan F V Medrano
- Laboratório de Vetores Virais, Centro de Investigação Translacional Em Oncologia/LIM 24, Instituto do Câncer do Estado de São Paulo (ICESP), Faculdade de Medicina, Universidade de São Paulo (FM-USP), Av. Dr. Arnaldo, 251, 8° Andar, São Paulo, SP, CEP: 01246-000, Brazil.,Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Thiago A Salles
- Laboratório de Genética e Cardiologia Molecular/LIM 13, Instituto do Coração, FM-USP, São Paulo, SP, Brazil
| | - Rafael Dariolli
- Laboratório de Genética e Cardiologia Molecular/LIM 13, Instituto do Coração, FM-USP, São Paulo, SP, Brazil.,Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Fernanda Antunes
- Laboratório de Vetores Virais, Centro de Investigação Translacional Em Oncologia/LIM 24, Instituto do Câncer do Estado de São Paulo (ICESP), Faculdade de Medicina, Universidade de São Paulo (FM-USP), Av. Dr. Arnaldo, 251, 8° Andar, São Paulo, SP, CEP: 01246-000, Brazil
| | - Valker A Feitosa
- Núcleo de Bionanomanufatura, Instituto de Pesquisas Tecnológicas (Bionano-IPT), São Paulo, SP, Brazil.,Faculdade de Ciências Farmaceuticas, Universidade Estadual Paulista Júlio de Mesquita Filho, Araraquara, SP, Brazil
| | - Aline Hunger
- Laboratório de Vetores Virais, Centro de Investigação Translacional Em Oncologia/LIM 24, Instituto do Câncer do Estado de São Paulo (ICESP), Faculdade de Medicina, Universidade de São Paulo (FM-USP), Av. Dr. Arnaldo, 251, 8° Andar, São Paulo, SP, CEP: 01246-000, Brazil.,Cristalia, Biotecnologia Unidade 1, Rodoviária SP 147, Itapira, SP, Brazil
| | - João P P Catani
- Laboratório de Vetores Virais, Centro de Investigação Translacional Em Oncologia/LIM 24, Instituto do Câncer do Estado de São Paulo (ICESP), Faculdade de Medicina, Universidade de São Paulo (FM-USP), Av. Dr. Arnaldo, 251, 8° Andar, São Paulo, SP, CEP: 01246-000, Brazil.,Vlaams Instituut Voor Biotenchnologie-UGent Center for Medical Biotechnology, Gent, Belgium
| | - Samir A Mendonça
- Laboratório de Vetores Virais, Centro de Investigação Translacional Em Oncologia/LIM 24, Instituto do Câncer do Estado de São Paulo (ICESP), Faculdade de Medicina, Universidade de São Paulo (FM-USP), Av. Dr. Arnaldo, 251, 8° Andar, São Paulo, SP, CEP: 01246-000, Brazil.,Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO, USA
| | - Rodrigo E Tamura
- Laboratório de Vetores Virais, Centro de Investigação Translacional Em Oncologia/LIM 24, Instituto do Câncer do Estado de São Paulo (ICESP), Faculdade de Medicina, Universidade de São Paulo (FM-USP), Av. Dr. Arnaldo, 251, 8° Andar, São Paulo, SP, CEP: 01246-000, Brazil.,Department of Biological Sciences, Federal University of São Paulo, Diadema, SP, Brazil
| | - Marlous G Lana
- Laboratório de Vetores Virais, Centro de Investigação Translacional Em Oncologia/LIM 24, Instituto do Câncer do Estado de São Paulo (ICESP), Faculdade de Medicina, Universidade de São Paulo (FM-USP), Av. Dr. Arnaldo, 251, 8° Andar, São Paulo, SP, CEP: 01246-000, Brazil
| | - Elaine G Rodrigues
- Department of Microbiology, Immunology and Parasitology, Paulista School of Medicine, Federal University of São Paulo (EPM-UNIFESP), São Paulo, Brazil
| | - Bryan E Strauss
- Laboratório de Vetores Virais, Centro de Investigação Translacional Em Oncologia/LIM 24, Instituto do Câncer do Estado de São Paulo (ICESP), Faculdade de Medicina, Universidade de São Paulo (FM-USP), Av. Dr. Arnaldo, 251, 8° Andar, São Paulo, SP, CEP: 01246-000, Brazil.
| |
Collapse
|
5
|
Tilsed CM, Casey TH, de Jong E, Bosco A, Zemek RM, Salmons J, Wan G, Millward MJ, Nowak AK, Lake RA, Lesterhuis WJ. Retinoic Acid Induces an IFN-Driven Inflammatory Tumour Microenvironment, Sensitizing to Immune Checkpoint Therapy. Front Oncol 2022; 12:849793. [PMID: 35402250 PMCID: PMC8988133 DOI: 10.3389/fonc.2022.849793] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 02/28/2022] [Indexed: 12/21/2022] Open
Abstract
With immune checkpoint therapy (ICT) having reshaped the treatment of many cancers, the next frontier is to identify and develop novel combination therapies to improve efficacy. Previously, we and others identified beneficial immunological effects of the vitamin A derivative tretinoin on anti-tumour immunity. Although it is known that tretinoin preferentially depletes myeloid derived suppressor cells in blood, little is known about the effects of tretinoin on the tumour microenvironment, hampering the rational design of clinical trials using tretinoin in combination with ICT. Here, we aimed to identify how tretinoin changed the tumour microenvironment in mouse tumour models, using flow cytometry and RNAseq, and we sought to use that information to establish optimal dosing and scheduling of tretinoin in combination with several ICT antibodies in multiple cancer models. We found that tretinoin rapidly induced an interferon dominated inflammatory tumour microenvironment, characterised by increased CD8+ T cell infiltration. This phenotype completely overlapped with the phenotype that was induced by ICT itself, and we confirmed that the combination further amplified this inflammatory milieu. The addition of tretinoin significantly improved the efficacy of anti-CTLA4/anti-PD-L1 combination therapy, and staggered scheduling was more efficacious than concomitant scheduling, in a dose-dependent manner. The positive effects of tretinoin could be extended to ICT antibodies targeting OX40, GITR and CTLA4 monotherapy in multiple cancer models. These data show that tretinoin induces an interferon driven, CD8+ T cell tumour microenvironment that is responsive to ICT.
Collapse
Affiliation(s)
- Caitlin M. Tilsed
- School of Biomedical Sciences, University of Western Australia, Crawley, WA, Australia
- National Centre for Asbestos Related Diseases, Institute for Respiratory Health, Nedlands, WA, Australia
| | - Thomas H. Casey
- School of Biomedical Sciences, University of Western Australia, Crawley, WA, Australia
- National Centre for Asbestos Related Diseases, Institute for Respiratory Health, Nedlands, WA, Australia
| | - Emma de Jong
- Telethon Kids Institute, University of Western Australia, Nedlands, WA, Australia
| | - Anthony Bosco
- Telethon Kids Institute, University of Western Australia, Nedlands, WA, Australia
| | - Rachael M. Zemek
- Telethon Kids Institute, University of Western Australia, Nedlands, WA, Australia
| | - Joanne Salmons
- School of Biomedical Sciences, University of Western Australia, Crawley, WA, Australia
- National Centre for Asbestos Related Diseases, Institute for Respiratory Health, Nedlands, WA, Australia
| | - Graeme Wan
- School of Biomedical Sciences, University of Western Australia, Crawley, WA, Australia
- National Centre for Asbestos Related Diseases, Institute for Respiratory Health, Nedlands, WA, Australia
| | - Michael J. Millward
- Medical School, University of Western Australia, Crawley, WA, Australia
- Department of Medical Oncology, Sir Charles Gairdner Hospital, Nedlands, WA, Australia
| | - Anna K. Nowak
- National Centre for Asbestos Related Diseases, Institute for Respiratory Health, Nedlands, WA, Australia
- Medical School, University of Western Australia, Crawley, WA, Australia
- Department of Medical Oncology, Sir Charles Gairdner Hospital, Nedlands, WA, Australia
| | - Richard A. Lake
- School of Biomedical Sciences, University of Western Australia, Crawley, WA, Australia
- National Centre for Asbestos Related Diseases, Institute for Respiratory Health, Nedlands, WA, Australia
| | - Willem Joost Lesterhuis
- School of Biomedical Sciences, University of Western Australia, Crawley, WA, Australia
- National Centre for Asbestos Related Diseases, Institute for Respiratory Health, Nedlands, WA, Australia
- Telethon Kids Institute, University of Western Australia, Nedlands, WA, Australia
- *Correspondence: Willem Joost Lesterhuis,
| |
Collapse
|
6
|
Wolf SP, Wen FT, Schreiber H. Criteria to make animal studies more relevant to treating human cancer. Curr Opin Immunol 2022; 74:25-31. [PMID: 34619458 PMCID: PMC8901458 DOI: 10.1016/j.coi.2021.09.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 08/28/2021] [Accepted: 09/11/2021] [Indexed: 02/03/2023]
Abstract
Certain aspects of experimental tumor models in mice most accurately reflect the biology and immunology of cancer in patients. A survey of experimental cancer immunotherapy papers published in 2020 shows most do not achieve cancer shrinkage although treatment is initiated at an early time point after cancer cell injection, which does not reflect cancer immunotherapy in patients. Even then, few current experimental approaches eradicate the injected malignant cells, most only delay outgrowth. The value of targeting mutation-encoded tumor-specific antigens becomes increasingly evident while problems of finding normal gene-encoded tumor-associated antigens as safe, effective targets persist. It might be time to refocus on realistic experimental settings and truly cancer-specific targets. These antigens are associated with the least risk of side effects.
Collapse
Affiliation(s)
- Steven P Wolf
- Department of Pathology, The University of Chicago, Chicago, IL, 60637, USA; David and Etta Jonas Center for Cellular Therapy, The University of Chicago, Chicago, IL, 60637, USA
| | - Frank T Wen
- Department of Medicine, The University of Chicago, Chicago, IL, 60637, USA
| | - Hans Schreiber
- Department of Pathology, The University of Chicago, Chicago, IL, 60637, USA; David and Etta Jonas Center for Cellular Therapy, The University of Chicago, Chicago, IL, 60637, USA; Committee on Cancer Biology and Committee on Immunology, The University of Chicago, Chicago, IL, 60637, USA.
| |
Collapse
|
7
|
Böttinger P, Schreiber K, Hyjek E, Krausz T, Spiotto MT, Steiner M, Idel C, Booras H, Beck-Engeser G, Riederer J, Willimsky G, Wolf SP, Karrison T, Jensen E, Weichselbaum RR, Nakamura Y, Yew PY, Lambert PF, Kurita T, Kiyotani K, Leisegang M, Schreiber H. Cooperation of genes in HPV16 E6/E7-dependent cervicovaginal carcinogenesis trackable by endoscopy and independent of exogenous estrogens or carcinogens. Carcinogenesis 2021; 41:1605-1615. [PMID: 32221533 DOI: 10.1093/carcin/bgaa027] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 03/16/2020] [Accepted: 03/26/2020] [Indexed: 02/07/2023] Open
Abstract
Human papillomavirus (HPV) infection is necessary but insufficient for progression of epithelial cells from dysplasia to carcinoma-in situ (CIS) to invasive cancer. The combination of mutant cellular and viral oncogenes that regulate progression of cervical cancer (CC) remains unclear. Using combinations of HPV16 E6/E7 (E+), mutant Kras (mKras) (K+) and/or loss of Pten (P-/-), we generated autochthonous models of CC without exogenous estrogen, carcinogen or promoters. Furthermore, intravaginal instillation of adenoCre virus enabled focal activation of the oncogenes/inactivation of the tumor suppressor gene. In P+/+ mice, E6/E7 alone (P+/+E+K-) failed to cause premalignant changes, while mKras alone (P+/+E-K+) caused persistent mucosal abnormalities in about one-third of mice, but no cancers. To develop cancer, P+/+ mice needed both E6/E7 and mKras expression. Longitudinal endoscopies of P+/+E+K+ mice predicted carcinoma development by detection of mucosal lesions, found on an average of 23 weeks prior to death, unlike longitudinal quantitative PCRs of vaginal lavage samples from the same mice. Endoscopy revealed that individual mice differed widely in the time required for mucosal lesions to appear after adenoCre and in the time required for these lesions to progress to cancer. These cancers developed in the transition zone that extends, unlike in women, from the murine cervix to the distal vagina. The P-/-E+K+ genotype led to precipitous cancer development within a few weeks and E6/E7-independent cancer development occurred in the P-/-E-K+ genotype. In the P-/-E+K- genotype, mice only developed CIS. Thus, distinct combinations of viral and cellular oncogenes are involved in distinct steps in cervical carcinogenesis.
Collapse
Affiliation(s)
- Paula Böttinger
- Department of Pathology, The University of Chicago, Chicago, IL, USA
| | - Karin Schreiber
- Department of Pathology, The University of Chicago, Chicago, IL, USA
| | - Elizabeth Hyjek
- Department of Radiation and Cellular Oncology, The University of Chicago, Chicago, IL, USA
| | - Thomas Krausz
- Department of Radiation and Cellular Oncology, The University of Chicago, Chicago, IL, USA
| | - Michael T Spiotto
- Department of Radiation and Cellular Oncology, The University of Chicago, Chicago, IL, USA
| | - Madeline Steiner
- Department of Pathology, The University of Chicago, Chicago, IL, USA
| | - Christian Idel
- Department of Pathology, The University of Chicago, Chicago, IL, USA
| | - Heather Booras
- Department of Pathology, The University of Chicago, Chicago, IL, USA
| | | | - Jessie Riederer
- Department of Pathology, The University of Chicago, Chicago, IL, USA
| | - Gerald Willimsky
- Institute of Immunology, Charité-Universitätsmedizin Berlin, Campus Buch, Berlin, Germany.,German Cancer Research Center, Heidelberg, Germany.,German Cancer Consortium, Partner site Berlin, Berlin, Germany
| | - Steven P Wolf
- Department of Pathology, The University of Chicago, Chicago, IL, USA.,Institute of Immunology, Charité-Universitätsmedizin Berlin, Campus Buch, Berlin, Germany
| | - Theodore Karrison
- Department of Public Health Sciences, The University of Chicago, Chicago, IL, USA
| | - Elizabeth Jensen
- Department of Pathology, The University of Chicago, Chicago, IL, USA
| | - Ralph R Weichselbaum
- Department of Radiation and Cellular Oncology, The University of Chicago, Chicago, IL, USA
| | - Yusuke Nakamura
- Project for Immunogenomics, Cancer Precision Medicine Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Poh Yin Yew
- Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Paul F Lambert
- McArdle Laboratory for Cancer Research/Department of Oncology, University of Wisconsin, Madison, WI, USA
| | - Takeshi Kurita
- Department of Cancer Biology and Genetics, Ohio State University, Columbus, OH, USA
| | - Kazuma Kiyotani
- Project for Immunogenomics, Cancer Precision Medicine Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Matthias Leisegang
- Institute of Immunology, Charité-Universitätsmedizin Berlin, Campus Buch, Berlin, Germany
| | - Hans Schreiber
- Department of Pathology, The University of Chicago, Chicago, IL, USA
| |
Collapse
|
8
|
Optimal combination treatment regimens of vaccine and radiotherapy augment tumor-bearing host immunity. Commun Biol 2021; 4:78. [PMID: 33469123 PMCID: PMC7815836 DOI: 10.1038/s42003-020-01598-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 12/14/2020] [Indexed: 01/29/2023] Open
Abstract
A major obstacle to immunotherapy is insufficient infiltration of effector immune cells into the tumor microenvironment. Radiotherapy greatly reduces tumor burden but relapses often occur. Here we show that the immunosuppressive tumor microenvironment was gradually established by recruiting Tregs after radiation. Despite tumors being controlled after depletion of Tregs in the irradiated area, improvement of mice survival remained poor. A much better antitumor effect was achieved with vaccination followed by radiation than other treatments. Vaccination followed by radiation recruited more effector T cells in tumor regions, which responded to high levels of chemokines. Sequential combination of vaccination and radiotherapy could elicit distinct host immune responses. Our study demonstrated that optimal combination of irradiation and vaccination is required to achieve effective antitumor immune responses. We propose a combination regimen that could be easily translated into the clinic and offer an opportunity for rational combination therapies design in cancer treatment.
Collapse
|
9
|
Morse CB, Voillet V, Bates BM, Chiu EY, Garcia NM, Gottardo R, Greenberg PD, Anderson KG. Development of a clinically relevant ovarian cancer model incorporating surgical cytoreduction to evaluate treatment of micro-metastatic disease. Gynecol Oncol 2020; 160:427-437. [PMID: 33229044 DOI: 10.1016/j.ygyno.2020.11.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Accepted: 11/08/2020] [Indexed: 12/16/2022]
Abstract
OBJECTIVES Mouse models of ovarian cancer commonly transfer large numbers of tumor cells into the peritoneal cavity to establish experimental metastatic disease, which may not adequately model early metastatic spread from a primary tumor site. We hypothesized we could develop an ovarian cancer model that predictably represents micro-metastatic disease. METHODS Murine ID8VEGF ovarian cancer cells were transduced to express enhanced luciferase (eLuc) to enable intravital detection of microscopic disease burden and injected beneath the ovarian bursa of C57Bl/6 mice. At 6 or 10 weeks after orthotopic injection, when mice had detectable metastases, hysterectomy and bilateral salpingo-oophorectomy was performed to remove all macroscopic disease, and survival monitored. Immunohistochemistry and gene expression profiling were performed on primary and metastatic tumors. RESULTS eLuc-transduced ID8VEGF cells were brighter than cells transduced with standard luciferase, enabling in vivo visualization of microscopic intra-abdominal metastases developing after orthotopic injection. Primary surgical cytoreduction removed the primary tumor mass but left minimal residual disease in all mice. Metastatic sites that developed following orthotopic injection were similar to metastatic human ovarian cancer sites. Gene expression and immune infiltration were similar between primary and metastatic mouse tumors. Surgical cytoreduction prolonged survival compared to no surgery, with earlier cytoreduction more beneficial than delayed, despite micro-metastatic disease in both settings. CONCLUSIONS Mice with primary ovarian tumors established through orthotopic injection develop progressively fatal metastatic ovarian cancer, and benefit from surgical cytoreduction to remove bulky disease. This model enables the analysis of therapeutic regimens designed to target and potentially eradicate established minimal residual disease.
Collapse
Affiliation(s)
- Christopher B Morse
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of Washington, Seattle, WA 98195, United States of America; Program in Immunology, Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, United States of America; Division of Gynecologic Oncology, Allegheny Health Network, West Penn Hospital, Mellon Pavilion, Suite 310, 4815 Liberty Avenue, Pittsburgh, PA 15224, United States of America.
| | - Valentin Voillet
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, United States of America
| | - Breanna M Bates
- Program in Immunology, Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, United States of America
| | - Edison Y Chiu
- Program in Immunology, Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, United States of America
| | - Nicolas M Garcia
- Program in Immunology, Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, United States of America
| | - Raphael Gottardo
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, United States of America
| | - Philip D Greenberg
- Program in Immunology, Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, United States of America; Divison of Medical Oncology, Department of Medicine, Department of Immunology, University of Washington, Seattle, WA 98195, United States of America.
| | - Kristin G Anderson
- Program in Immunology, Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, United States of America.
| |
Collapse
|
10
|
Guzik P, Siwowska K, Fang HY, Cohrs S, Bernhardt P, Schibli R, Müller C. Promising potential of [ 177Lu]Lu-DOTA-folate to enhance tumor response to immunotherapy-a preclinical study using a syngeneic breast cancer model. Eur J Nucl Med Mol Imaging 2020; 48:984-994. [PMID: 33078260 PMCID: PMC8041666 DOI: 10.1007/s00259-020-05054-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Accepted: 09/22/2020] [Indexed: 12/01/2022]
Abstract
PURPOSE It was previously demonstrated that radiation effects can enhance the therapy outcome of immune checkpoint inhibitors. In this study, a syngeneic breast tumor mouse model was used to investigate the effect of [177Lu]Lu-DOTA-folate as an immune stimulus to enhance anti-CTLA-4 immunotherapy. METHODS In vitro and in vivo studies were performed to characterize NF9006 breast tumor cells with regard to folate receptor (FR) expression and the possibility of tumor targeting using [177Lu]Lu-DOTA-folate. A preclinical therapy study was performed over 70 days with NF9006 tumor-bearing mice that received vehicle only (group A); [177Lu]Lu-DOTA-folate (5 MBq; 3.5 Gy absorbed tumor dose; group B); anti-CTLA-4 antibody (3 × 200 μg; group C), or both agents (group D). The mice were monitored regarding tumor growth over time and signs indicating adverse events of the treatment. RESULTS [177Lu]Lu-DOTA-folate bound specifically to NF9006 tumor cells and tissue in vitro and accumulated in NF9006 tumors in vivo. The treatment with [177Lu]Lu-DOTA-folate or an anti-CTLA-4 antibody had only a minor effect on NF9006 tumor growth and did not substantially increase the median survival time of mice (23 day and 19 days, respectively) as compared with untreated controls (12 days). [177Lu]Lu-DOTA-folate sensitized, however, the tumors to anti-CTLA-4 immunotherapy, which became obvious by reduced tumor growth and, hence, a significantly improved median survival time of mice (> 70 days). No obvious signs of adverse effects were observed in treated mice as compared with untreated controls. CONCLUSION Application of [177Lu]Lu-DOTA-folate had a positive effect on the therapy outcome of anti-CTLA-4 immunotherapy. The results of this study may open new perspectives for future clinical translation of folate radioconjugates.
Collapse
Affiliation(s)
- Patrycja Guzik
- Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institute, 5232, Villigen-PSI, Switzerland
| | - Klaudia Siwowska
- Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institute, 5232, Villigen-PSI, Switzerland
| | - Hsin-Yu Fang
- Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institute, 5232, Villigen-PSI, Switzerland
| | - Susan Cohrs
- Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institute, 5232, Villigen-PSI, Switzerland
| | - Peter Bernhardt
- Department of Radiation Physics, The Sahlgrenska Academy, University of Gothenburg, SE-413 45, Gothenburg, Sweden.,Department of Medical Physics and Medical Bioengineering, Sahlgrenska University Hospital, SE-413 45, Gothenburg, Sweden
| | - Roger Schibli
- Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institute, 5232, Villigen-PSI, Switzerland.,Department of Chemistry and Applied Biosciences, ETH Zurich, 8093, Zurich, Switzerland
| | - Cristina Müller
- Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institute, 5232, Villigen-PSI, Switzerland. .,Department of Chemistry and Applied Biosciences, ETH Zurich, 8093, Zurich, Switzerland.
| |
Collapse
|
11
|
Temples MN, Adjei IM, Nimocks PM, Djeu J, Sharma B. Engineered Three-Dimensional Tumor Models to Study Natural Killer Cell Suppression. ACS Biomater Sci Eng 2020; 6:4179-4199. [PMID: 33463353 DOI: 10.1021/acsbiomaterials.0c00259] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
A critical hurdle associated with natural killer (NK) cell immunotherapies is inadequate infiltration and function in the solid tumor microenvironment. Well-controlled 3D culture systems could advance our understanding of the role of various biophysical and biochemical cues that impact NK cell migration in solid tumors. The objectives of this study were to establish a biomaterial which (i) supports NK cell migration and (ii) recapitulates features of the in vivo solid tumor microenvironment, to study NK infiltration and function in a 3D system. Using peptide-functionalized poly(ethylene glycol)-based hydrogels, the extent of NK-92 cell migration was observed to be largely dependent on the density of integrin binding sites and the presence of matrix metalloproteinase degradable sites. When lung cancer cells were encapsulated into the hydrogels to create tumor microenvironments, the extent of NK-92 cell migration and functional activity was dependent on the cancer cell type and duration of 3D culture. NK-92 cells showed greater migration into the models consisting of nonmetastatic A549 cells relative to metastatic H1299 cells, and reduced migration in both models when cancer cells were cultured for 7 days versus 1 day. In addition, the production of NK cell-related pro-inflammatory cytokines and chemokines was reduced in H1299 models relative to A549 models. These differences in NK-92 cell migration and cytokine/chemokine production corresponded to differences in the production of various immunomodulatory molecules by the different cancer cells, namely, the H1299 models showed increased stress ligand shedding and immunosuppressive cytokine production, particularly TGF-β. Indeed, inhibition of TGF-β receptor I in NK-92 cells restored their infiltration in H1299 models to levels similar to that in A549 models and increased overall infiltration in both models. Relative to conventional 2D cocultures, NK-92 cell mediated cytotoxicity was reduced in the 3D tumor models, suggesting the hydrogel serves to mimic some features of the biophysical barriers in in vivo tumor microenvironments. This study demonstrates the feasibility of a synthetic hydrogel system for investigating the biophysical and biochemical cues impacting NK cell infiltration and NK cell-cancer cell interactions in the solid tumor microenvironment.
Collapse
Affiliation(s)
- Madison N Temples
- J. Crayton Pruitt Department of Biomedical Engineering, University of Florida, Biomedical Sciences Building JG-56, 1275 Center Drive, Gainesville, Florida 32611-6131, United States
| | - Isaac M Adjei
- J. Crayton Pruitt Department of Biomedical Engineering, University of Florida, Biomedical Sciences Building JG-56, 1275 Center Drive, Gainesville, Florida 32611-6131, United States
| | - Phoebe M Nimocks
- J. Crayton Pruitt Department of Biomedical Engineering, University of Florida, Biomedical Sciences Building JG-56, 1275 Center Drive, Gainesville, Florida 32611-6131, United States
| | - Julie Djeu
- Department of Immunology, Moffitt Cancer Center MRC 4E, 12902 Magnolia Drive, Tampa, Florida 33612-9497, United States
| | - Blanka Sharma
- J. Crayton Pruitt Department of Biomedical Engineering, University of Florida, Biomedical Sciences Building JG-56, 1275 Center Drive, Gainesville, Florida 32611-6131, United States
| |
Collapse
|
12
|
Ichikawa J, Yoshida T, Isser A, Laino AS, Vassallo M, Woods D, Kim S, Oelke M, Jones K, Schneck JP, Weber JS. Rapid Expansion of Highly Functional Antigen-Specific T Cells from Patients with Melanoma by Nanoscale Artificial Antigen-Presenting Cells. Clin Cancer Res 2020; 26:3384-3396. [PMID: 32241816 DOI: 10.1158/1078-0432.ccr-19-3487] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 02/13/2020] [Accepted: 03/30/2020] [Indexed: 01/06/2023]
Abstract
PURPOSE Generation of antigen-specific T cells from patients with cancer employs large numbers of peripheral blood cells and/or tumor-infiltrating cells to generate antigen-presenting and effector cells commonly requiring multiple rounds of restimulation ex vivo. We used a novel paramagnetic, nanoparticle-based artificial antigen-presenting cell (nano-aAPC) that combines anti-CD28 costimulatory and human MHC class I molecules that are loaded with antigenic peptides to rapidly expand tumor antigen-specific T cells from patients with melanoma. EXPERIMENTAL DESIGN Nano-aAPC-expressing HLA-A*0201 molecules and costimulatory anti-CD28 antibody and HLA-A*0201 molecules loaded with MART-1 or gp100 class I-restricted peptides were used to stimulate CD8 T cells purified from the peripheral blood of treatment-naïve or PD-1 antibody-treated patients with stage IV melanoma. Expanded cells were restimulated with fresh peptide-pulsed nano-aAPC at day 7. Phenotype analysis and functional assays including cytokine release, cytolysis, and measurement of avidity were conducted. RESULTS MART-1-specific CD8 T cells rapidly expanded up to 1,000-fold by day 14 after exposure to peptide-pulsed nano-aAPC. Expanded T cells had a predominantly stem cell memory CD45RA+/CD62L+/CD95+ phenotype; expressed ICOS, PD-1, Tim3, and LAG3; and lacked CD28. Cells from patients with melanoma were polyfunctional; highly avid; expressed IL2, IFNγ, and TNFα; and exhibited cytolytic activity against tumor cell lines. They expanded 2- to 3-fold after exposure to PD-1 antibody in vivo, and expressed a highly diverse T-cell receptor V beta repertoire. CONCLUSIONS Peptide-pulsed nano-aAPC rapidly expanded polyfunctional antigen-specific CD8 T cells with high avidity, potent lytic function, and a stem cell memory phenotype from patients with melanoma.
Collapse
Affiliation(s)
- Junya Ichikawa
- NYU Langone Medical Center, Laura and Isaac Perlmutter Cancer Center, New York, New York.
| | - Tatsuya Yoshida
- NYU Langone Medical Center, Laura and Isaac Perlmutter Cancer Center, New York, New York
| | - Ariel Isser
- Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Andressa S Laino
- NYU Langone Medical Center, Laura and Isaac Perlmutter Cancer Center, New York, New York
| | - Melinda Vassallo
- NYU Langone Medical Center, Laura and Isaac Perlmutter Cancer Center, New York, New York
| | - David Woods
- NYU Langone Medical Center, Laura and Isaac Perlmutter Cancer Center, New York, New York
| | | | | | | | | | - Jeffrey S Weber
- NYU Langone Medical Center, Laura and Isaac Perlmutter Cancer Center, New York, New York.
| |
Collapse
|
13
|
Faithful preclinical mouse models for better translation to bedside in the field of immuno-oncology. Int J Clin Oncol 2019; 25:831-841. [PMID: 31407168 DOI: 10.1007/s10147-019-01520-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 08/01/2019] [Indexed: 12/19/2022]
Abstract
The success of immunotherapy using immune checkpoint inhibitors has changed the practice of cancer treatment tremendously. However, there are still many clinical challenges, such as drug resistance, predictive biomarker development, exploration of combination therapies, and prediction of immune-related adverse events in preclinical settings. To overcome these problems, it is essential to establish faithful preclinical mouse models that recapitulate the clinical features, molecular genetics, biological heterogeneity, and immune microenvironment of human cancers. Here we review the advantages and disadvantages of current preclinical mouse models, including syngeneic murine tumor cell lines, autochthonous tumor models, cancer cell line-derived xenografts, patient-derived-xenografts, and various kinds of immunologically humanized mice. We discuss how these models should be characterized and applied in preclinical settings, and how we should prepare preclinical studies for successful translation from bench to bedside.
Collapse
|
14
|
Zhai L, Ladomersky E, Lauing KL, Wu M, Scholtens DM, Savoor R, Zhang B, Wu JD, Horbinski C, Lukas RV, Binder DC, Wainwright DA. Commentary: preclinical efficacy of immune-checkpoint monotherapy does not recapitulate corresponding biomarkers-based clinical predictions in glioblastoma by Garg et al. (2017). Oncoimmunology 2018; 8:1548242. [PMID: 30723577 DOI: 10.1080/2162402x.2018.1548242] [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: 07/30/2018] [Revised: 10/29/2018] [Accepted: 11/09/2018] [Indexed: 10/27/2022] Open
Abstract
Preclinical modeling and gene expression analyses have yielded distinct observations for the role of immune checkpoint, IDO1, in glioblastoma (GBM). Accordingly, our recent work differs with Garg et al. (2017) with respect to IDO1 among preclinical and bioinformatic GBM datasets. Here, we discuss the methodological differences that affected study interpretation, and potentially, future clinical decision-making for IDO1-targeting approaches against GBM.
Collapse
Affiliation(s)
- Lijie Zhai
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Erik Ladomersky
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Kristen L Lauing
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Meijing Wu
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Denise M Scholtens
- Department of Preventive Medicine-Biostatistics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Rohan Savoor
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Bin Zhang
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.,Department of Microbiology-Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.,Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Jennifer D Wu
- Department of Microbiology-Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.,Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Craig Horbinski
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.,Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Rimas V Lukas
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - David C Binder
- Department of Radiation Oncology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Derek A Wainwright
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.,Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.,Department of Microbiology-Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.,Department of Medicine-Division of Hematology and Oncology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
15
|
Zafar S, Sorsa S, Siurala M, Hemminki O, Havunen R, Cervera-Carrascon V, Santos JM, Wang H, Lieber A, De Gruijl T, Kanerva A, Hemminki A. CD40L coding oncolytic adenovirus allows long-term survival of humanized mice receiving dendritic cell therapy. Oncoimmunology 2018; 7:e1490856. [PMID: 30386680 PMCID: PMC6207416 DOI: 10.1080/2162402x.2018.1490856] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 06/13/2018] [Accepted: 06/13/2018] [Indexed: 12/12/2022] Open
Abstract
Dendritic cells (DCs) are crucial players in promoting immune responses. Logically, adoptive DC therapy is a promising approach in cancer immunotherapy. One of the major obstacles in cancer immunotherapy in general is the immunosuppressive tumor microenvironment, which hampers the maturation and activation of DCs. Therefore, human clinical outcomes with DC therapy alone have been disappointing. In this study, we use fully serotype 3 oncolytic adenovirus Ad3-hTERT-CMV-hCD40L, expressing human CD40L, to modulate the tumor microenvironment with subsequently improved function of DCs. We evaluated the synergistic effects of Ad3-hTERT-CMV-hCD40L and DCs in the presence of human peripheral blood mononuclear cells ex vivo and in vivo. Tumors treated with Ad3-hTERT-CMV-hCD40L and DCs featured greater antitumor effect compared with unarmed virus or either treatment alone. 100% of humanized mice survived to the end of the experiment, while mice in all other groups died by day 88. Moreover, adenovirally-delivered CD40L induced activation of DCs, leading to induction of Th1 immune responses. These results support clinical trials with Ad3-hTERT-CMV-hCD40L in patients receiving DC therapy.
Collapse
Affiliation(s)
- Sadia Zafar
- Cancer Gene Therapy Group, University of Helsinki, Helsinki, Finland
| | - Suvi Sorsa
- Cancer Gene Therapy Group, University of Helsinki, Helsinki, Finland
- TILT Biotherapeutics Ltd, Helsinki, Finland
| | - Mikko Siurala
- Cancer Gene Therapy Group, University of Helsinki, Helsinki, Finland
- TILT Biotherapeutics Ltd, Helsinki, Finland
| | - Otto Hemminki
- Cancer Gene Therapy Group, University of Helsinki, Helsinki, Finland
- Division of Urology, Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
| | - Riikka Havunen
- Cancer Gene Therapy Group, University of Helsinki, Helsinki, Finland
- TILT Biotherapeutics Ltd, Helsinki, Finland
| | - Victor Cervera-Carrascon
- Cancer Gene Therapy Group, University of Helsinki, Helsinki, Finland
- TILT Biotherapeutics Ltd, Helsinki, Finland
| | - João Manuel Santos
- Cancer Gene Therapy Group, University of Helsinki, Helsinki, Finland
- TILT Biotherapeutics Ltd, Helsinki, Finland
| | - Hongjie Wang
- Department of Pathology, University of Washington, Seattle, WA, USA
| | - Andre Lieber
- Department of Pathology, University of Washington, Seattle, WA, USA
| | - Tanja De Gruijl
- Department of Medical Oncology, VU University Medical Center, Amsterdam, the Netherlands
| | - Anna Kanerva
- Cancer Gene Therapy Group, University of Helsinki, Helsinki, Finland
- Department of Obstetrics and Gynecology, Helsinki University Central Hospital, Helsinki, Finland
| | - Akseli Hemminki
- Cancer Gene Therapy Group, University of Helsinki, Helsinki, Finland
- TILT Biotherapeutics Ltd, Helsinki, Finland
- Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
| |
Collapse
|
16
|
Contreras-Sandoval AM, Merino M, Vasquez M, Trocóniz IF, Berraondo P, Garrido MJ. Correlation between anti-PD-L1 tumor concentrations and tumor-specific and nonspecific biomarkers in a melanoma mouse model. Oncotarget 2018; 7:76891-76901. [PMID: 27764774 PMCID: PMC5363557 DOI: 10.18632/oncotarget.12727] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Accepted: 10/11/2016] [Indexed: 01/15/2023] Open
Abstract
Blockade of PD-L1 with specific monoclonal antibodies (anti-PD-L1) represents a therapeutic strategy to increase the capability of the immune system to modulate the tumor immune-resistance. The relationship between anti-PD-L1 tumor exposition and anti-tumor effect represents a challenge that has been addressed in this work through the identification of certain biomarkers implicated in the antibody's mechanism of action, using a syngeneic melanoma mouse model. The development of an in-vitro/in-vivo platform has allowed us to investigate the PD-L1 behavior after its blockage with anti-PD-L1 at cellular level and in animals. In-vitro studies showed that the complex PD-L1/anti-PD-L1 was retained mainly at the cell surface. The antibody concentration and time exposure affected directly the recycling or ligand turnover. In-vivo studies showed that anti-PD-L1 was therapeutically active at all stage of the disease, with a rapid onset, a low but durable efficacy and non-relevant toxic effect. This efficacy measured as tumor shrinkage correlated with tumor-specific infiltrating lymphocytes (TILs), which increased as antibody tumor concentrations increased. Both, TILS and antibody concentrations followed similar kinetic patterns, justifying the observed anti-PD-L1 rapid onset. Interestingly, peripheral lymphocytes (PBLs) behave as infiltrating lymphocytes, suggesting that these PBLs might be considered as a possible biomarker for antibody activity.
Collapse
Affiliation(s)
- Ana M Contreras-Sandoval
- School of Pharmacy, Department of Pharmacy and Pharmaceutical Technology, University of Navarra, 31008 Pamplona, Spain
| | - María Merino
- School of Pharmacy, Department of Pharmacy and Pharmaceutical Technology, University of Navarra, 31008 Pamplona, Spain
| | - Marcos Vasquez
- Program of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Navarra Institute for Health Research (IdiSNA), Pamplona, Navarra, 31008, Spain
| | - Iñaki F Trocóniz
- School of Pharmacy, Department of Pharmacy and Pharmaceutical Technology, University of Navarra, 31008 Pamplona, Spain
| | - Pedro Berraondo
- Program of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Navarra Institute for Health Research (IdiSNA), Pamplona, Navarra, 31008, Spain
| | - María J Garrido
- School of Pharmacy, Department of Pharmacy and Pharmaceutical Technology, University of Navarra, 31008 Pamplona, Spain
| |
Collapse
|
17
|
Abstract
DNA vaccines offer many advantages over other anti-tumor vaccine approaches due to their simplicity, ease of manufacturing, and safety. Results from several clinical trials in patients with cancer have demonstrated that DNA vaccines are safe and can elicit immune responses. However, to date few DNA vaccines have progressed beyond phase I clinical trial evaluation. Studies into the mechanism of action of DNA vaccines in terms of antigen-presenting cell types able to directly present or cross-present DNA-encoded antigens, and the activation of innate immune responses due to DNA itself, have suggested opportunities to increase the immunogenicity of these vaccines. In addition, studies into the mechanisms of tumor resistance to anti-tumor vaccination have suggested combination approaches that can increase the anti-tumor effect of DNA vaccines. This review focuses on these mechanisms of action and mechanisms of resistance using DNA vaccines, and how this information is being used to improve the anti-tumor effect of DNA vaccines. These approaches are then specifically discussed in the context of human prostate cancer, a disease for which DNA vaccines have been and continue to be explored as treatments.
Collapse
Affiliation(s)
- Christopher D Zahm
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI 53705, United States
| | - Viswa Teja Colluru
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI 53705, United States
| | - Douglas G McNeel
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI 53705, United States.
| |
Collapse
|
18
|
Abstract
Immuno-oncology (I/O) research has intensified significantly in recent years due to the breakthrough development and the regulatory approval of several immune checkpoint inhibitors, leading to the rapid expansion of the new discovery of novel I/O therapies, new checkpoint inhibitors and beyond. However, many I/O questions remain unanswered, including why only certain subsets of patients respond to these treatments, who the responders would be, and how to expand patient response (the conversion of non-responders or maximizing response in partial responders). All of these require relevant I/O experimental systems, particularly relevant preclinical animal models. Compared to other oncology drug discovery, e.g. cytotoxic and targeted drugs, a lack of relevant animal models is a major obstacle in I/O drug discovery, and an urgent and unmet need. Despite the obvious importance, and the fact that much I/O research has been performed using many different animal models, there are few comprehensive and introductory reviews on this topic. This article attempts to review the efforts in development of a variety of such models, as well as their applications and limitations for readers new to the field, particularly those in the pharmaceutical industry.
Collapse
Affiliation(s)
- Qi-Xiang Li
- Crown Bioscience Inc., 3375 Scott Blvd, Suite 108, Santa Clara, CA 95054, USA; State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100191, China.
| | - Gerold Feuer
- HuMurine Technologies, Inc., 2700 Stockton Blvd, Rm. 1403, Sacramento, CA 95817, USA
| | - Xuesong Ouyang
- Crown Bioscience Inc., 3375 Scott Blvd, Suite 108, Santa Clara, CA 95054, USA
| | - Xiaoyu An
- Crown Bioscience Inc., 3375 Scott Blvd, Suite 108, Santa Clara, CA 95054, USA; State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100191, China
| |
Collapse
|
19
|
Klinke DJ, Wang Q. Inferring the Impact of Regulatory Mechanisms that Underpin CD8+ T Cell Control of B16 Tumor Growth In vivo Using Mechanistic Models and Simulation. Front Pharmacol 2017; 7:515. [PMID: 28101055 PMCID: PMC5209634 DOI: 10.3389/fphar.2016.00515] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Accepted: 12/12/2016] [Indexed: 11/13/2022] Open
Abstract
A major barrier for broadening the efficacy of immunotherapies for cancer is identifying key mechanisms that limit the efficacy of tumor infiltrating lymphocytes. Yet, identifying these mechanisms using human samples and mouse models for cancer remains a challenge. While interactions between cancer and the immune system are dynamic and non-linear, identifying the relative roles that biological components play in regulating anti-tumor immunity commonly relies on human intuition alone, which can be limited by cognitive biases. To assist natural intuition, modeling and simulation play an emerging role in identifying therapeutic mechanisms. To illustrate the approach, we developed a multi-scale mechanistic model to describe the control of tumor growth by a primary response of CD8+ T cells against defined tumor antigens using the B16 C57Bl/6 mouse model for malignant melanoma. The mechanistic model was calibrated to data obtained following adenovirus-based immunization and validated to data obtained following adoptive transfer of transgenic CD8+ T cells. More importantly, we use simulation to test whether the postulated network topology, that is the modeled biological components and their associated interactions, is sufficient to capture the observed anti-tumor immune response. Given the available data, the simulation results also provided a statistical basis for quantifying the relative importance of different mechanisms that underpin CD8+ T cell control of B16F10 growth. By identifying conditions where the postulated network topology is incomplete, we illustrate how this approach can be used as part of an iterative design-build-test cycle to expand the predictive power of the model.
Collapse
Affiliation(s)
- David J Klinke
- Department of Chemical and Biomedical Engineering and WVU Cancer Institute, West Virginia UniversityMorgantown, WV, USA; Department of Microbiology, Immunology, and Cell Biology, West Virginia UniversityMorgantown, WV, USA
| | - Qing Wang
- Department of Computer Science, Mathematics and Engineering, Shepherd University Shepherdstown, WV, USA
| |
Collapse
|
20
|
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: 91] [Impact Index Per Article: 10.1] [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
|
21
|
Bialkowski L, van Weijnen A, Van der Jeught K, Renmans D, Daszkiewicz L, Heirman C, Stangé G, Breckpot K, Aerts JL, Thielemans K. Intralymphatic mRNA vaccine induces CD8 T-cell responses that inhibit the growth of mucosally located tumours. Sci Rep 2016; 6:22509. [PMID: 26931556 PMCID: PMC4773884 DOI: 10.1038/srep22509] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Accepted: 02/16/2016] [Indexed: 01/01/2023] Open
Abstract
The lack of appropriate mouse models is likely one of the reasons of a limited translational success rate of therapeutic vaccines against cervical cancer, as rapidly growing ectopic tumours are commonly used for preclinical studies. In this work, we demonstrate that the tumour microenvironment of TC-1 tumours differs significantly depending on the anatomical location of tumour lesions (i.e. subcutaneously, in the lungs and in the genital tract). Our data demonstrate that E7-TriMix mRNA vaccine-induced CD8+ T lymphocytes migrate into the tumour nest and control tumour growth, although they do not express mucosa-associated markers such as CD103 or CD49a. We additionally show that despite the presence of the antigen-specific T cells in the tumour lesions, the therapeutic outcomes in the genital tract model remain limited. Here, we report that such a hostile tumour microenvironment can be reversed by cisplatin treatment, leading to a complete regression of clinically relevant tumours when combined with mRNA immunization. We thereby demonstrate the necessity of utilizing clinically relevant models for preclinical evaluation of anticancer therapies and the importance of a simultaneous combination of anticancer immune response induction with targeting of tumour environment.
Collapse
Affiliation(s)
- Lukasz Bialkowski
- Laboratory of Molecular and Cellular Therapy, Vrije Universiteit Brussel, Laarbeeklaan 103E, 1090 Brussels, Belgium
| | - Alexia van Weijnen
- Laboratory of Molecular and Cellular Therapy, Vrije Universiteit Brussel, Laarbeeklaan 103E, 1090 Brussels, Belgium
| | - Kevin Van der Jeught
- Laboratory of Molecular and Cellular Therapy, Vrije Universiteit Brussel, Laarbeeklaan 103E, 1090 Brussels, Belgium
| | - Dries Renmans
- Laboratory of Molecular and Cellular Therapy, Vrije Universiteit Brussel, Laarbeeklaan 103E, 1090 Brussels, Belgium
| | - Lidia Daszkiewicz
- Laboratory of Molecular and Cellular Therapy, Vrije Universiteit Brussel, Laarbeeklaan 103E, 1090 Brussels, Belgium
| | - Carlo Heirman
- Laboratory of Molecular and Cellular Therapy, Vrije Universiteit Brussel, Laarbeeklaan 103E, 1090 Brussels, Belgium
| | - Geert Stangé
- Diabetes Research Center, Vrije Universiteit Brussel, Laarbeeklaan 103E, 1090 Brussels, Belgium
| | - Karine Breckpot
- Laboratory of Molecular and Cellular Therapy, Vrije Universiteit Brussel, Laarbeeklaan 103E, 1090 Brussels, Belgium
| | - Joeri L Aerts
- Laboratory of Molecular and Cellular Therapy, Vrije Universiteit Brussel, Laarbeeklaan 103E, 1090 Brussels, Belgium
| | - Kris Thielemans
- Laboratory of Molecular and Cellular Therapy, Vrije Universiteit Brussel, Laarbeeklaan 103E, 1090 Brussels, Belgium
| |
Collapse
|
22
|
Binder DC, Arina A, Wen F, Tu T, Zhao M, Hoffman RM, Wainwright DA, Schreiber H. Tumor relapse prevented by combining adoptive T cell therapy with Salmonella typhimurium. Oncoimmunology 2016; 5:e1130207. [PMID: 27471609 DOI: 10.1080/2162402x.2015.1130207] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Revised: 12/04/2015] [Accepted: 12/05/2015] [Indexed: 10/22/2022] Open
Abstract
We recently reported that therapeutic vaccination with live tumor antigen-producing Salmonella typhimurium rescues dysfunctional endogenous T cell responses and eradicates long-established tumors refractory to αCTLA-4 and αPD-L1 checkpoint inhibitor blockade. Here, we show that live intravenously injected or heat-killed (HK) intratumorally injected Salmonella typhimurium, even when not producing tumor antigen, synergize with adoptive T cell therapy to eradicate tumors. These data demonstrate that the combination of adoptive T cell transfer with the injection of live or dead Salmonella typhimurium is a promising approach for cancer treatment.
Collapse
Affiliation(s)
- David C Binder
- Committee on Cancer Biology, University of Chicago, Chicago, IL, USA; Department of Pathology, University of Chicago, Chicago, IL, USA
| | - Ainhoa Arina
- Department of Pathology, University of Chicago, Chicago, IL, USA; Committee on Immunology, The University of Chicago, Chicago, IL, USA
| | - Frank Wen
- Department of Pathology, University of Chicago , Chicago, IL, USA
| | - Tony Tu
- Department of Pathology, University of Chicago, Chicago, IL, USA; Committee on Immunology, The University of Chicago, Chicago, IL, USA
| | - Ming Zhao
- AntiCancer Inc. , San Diego, CA, USA
| | - Robert M Hoffman
- AntiCancer Inc., San Diego, CA, USA; Department of Surgery, University of California, San Diego, CA, USA
| | - Derek A Wainwright
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA; Northwestern Brain Tumor Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Hans Schreiber
- Committee on Cancer Biology, University of Chicago, Chicago, IL, USA; Department of Pathology, University of Chicago, Chicago, IL, USA; Committee on Immunology, The University of Chicago, Chicago, IL, USA
| |
Collapse
|
23
|
Leisegang M, Engels B, Schreiber K, Yew PY, Kiyotani K, Idel C, Arina A, Duraiswamy J, Weichselbaum RR, Uckert W, Nakamura Y, Schreiber H. Eradication of Large Solid Tumors by Gene Therapy with a T-Cell Receptor Targeting a Single Cancer-Specific Point Mutation. Clin Cancer Res 2015; 22:2734-43. [PMID: 26667491 DOI: 10.1158/1078-0432.ccr-15-2361] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Accepted: 12/07/2015] [Indexed: 11/16/2022]
Abstract
PURPOSE Cancers usually contain multiple unique tumor-specific antigens produced by single amino acid substitutions (AAS) and encoded by somatic nonsynonymous single nucleotide substitutions. We determined whether adoptively transferred T cells can reject large, well-established solid tumors when engineered to express a single type of T-cell receptor (TCR) that is specific for a single AAS. EXPERIMENTAL DESIGN By exome and RNA sequencing of an UV-induced tumor, we identified an AAS in p68 (mp68), a co-activator of p53. This AAS seemed to be an ideal tumor-specific neoepitope because it is encoded by a trunk mutation in the primary autochthonous cancer and binds with highest affinity to the MHC. A high-avidity mp68-specific TCR was used to genetically engineer T cells as well as to generate TCR-transgenic mice for adoptive therapy. RESULTS When the neoepitope was expressed at high levels and by all cancer cells, their direct recognition sufficed to destroy intratumor vessels and eradicate large, long-established solid tumors. When the neoepitope was targeted as autochthonous antigen, T cells caused cancer regression followed by escape of antigen-negative variants. Escape could be thwarted by expressing the antigen at increased levels in all cancer cells or by combining T-cell therapy with local irradiation. Therapeutic efficacies of TCR-transduced and TCR-transgenic T cells were similar. CONCLUSIONS Gene therapy with a single TCR targeting a single AAS can eradicate large established cancer, but a uniform expression and/or sufficient levels of the targeted neoepitope or additional therapy are required to overcome tumor escape. Clin Cancer Res; 22(11); 2734-43. ©2015 AACRSee related commentary by Liu, p. 2602.
Collapse
Affiliation(s)
| | - Boris Engels
- Department of Pathology, The University of Chicago, Illinois
| | - Karin Schreiber
- Department of Pathology, The University of Chicago, Illinois
| | - Poh Yin Yew
- Department of Medicine, The University of Chicago, Illinois
| | | | - Christian Idel
- Department of Pathology, The University of Chicago, Illinois
| | - Ainhoa Arina
- Department of Pathology, The University of Chicago, Illinois
| | | | - Ralph R Weichselbaum
- Department of Radiation and Cellular Oncology, The Ludwig Center for Metastasis Research, The University of Chicago, Illinois
| | - Wolfgang Uckert
- Molecular Cell Biology and Gene Therapy, Max-Delbrück-Center for Molecular Medicine, Berlin, Germany. Institute of Biology, Humboldt University Berlin, Berlin, Germany
| | | | - Hans Schreiber
- Institute of Immunology, Charité, Campus Buch, Berlin, Germany. Department of Pathology, The University of Chicago, Illinois
| |
Collapse
|
24
|
Perica K, Bieler JG, Schütz C, Varela JC, Douglass J, Skora A, Chiu YL, Oelke M, Kinzler K, Zhou S, Vogelstein B, Schneck JP. Enrichment and Expansion with Nanoscale Artificial Antigen Presenting Cells for Adoptive Immunotherapy. ACS NANO 2015; 9:6861-71. [PMID: 26171764 PMCID: PMC5082131 DOI: 10.1021/acsnano.5b02829] [Citation(s) in RCA: 114] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Adoptive immunotherapy (AIT) can mediate durable regression of cancer, but widespread adoption of AIT is limited by the cost and complexity of generating tumor-specific T cells. Here we develop an Enrichment + Expansion strategy using paramagnetic, nanoscale artificial antigen presenting cells (aAPC) to rapidly expand tumor-specific T cells from rare naïve precursors and predicted neo-epitope responses. Nano-aAPC are capable of enriching rare tumor-specific T cells in a magnetic column and subsequently activating them to induce proliferation. Enrichment + Expansion resulted in greater than 1000-fold expansion of both mouse and human tumor-specific T cells in 1 week, with nano-aAPC based enrichment conferring a proliferation advantage during both in vitro culture and after adoptive transfer in vivo. Robust T cell responses were seen not only for shared tumor antigens, but also for computationally predicted neo-epitopes. Streamlining the rapid generation of large numbers of tumor-specific T cells in a cost-effective fashion through Enrichment + Expansion can be a powerful tool for immunotherapy.
Collapse
Affiliation(s)
| | | | | | | | - Jacqueline Douglass
- ∥Ludwig Cancer Research Center and Howard Hughes Medical Institute, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
| | - Andrew Skora
- ∥Ludwig Cancer Research Center and Howard Hughes Medical Institute, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
| | | | | | - Kenneth Kinzler
- ∥Ludwig Cancer Research Center and Howard Hughes Medical Institute, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
| | - Shibin Zhou
- ∥Ludwig Cancer Research Center and Howard Hughes Medical Institute, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
| | - Bert Vogelstein
- ∥Ludwig Cancer Research Center and Howard Hughes Medical Institute, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
| | | |
Collapse
|
25
|
Pottier C, Wheatherspoon A, Roncarati P, Longuespée R, Herfs M, Duray A, Delvenne P, Quatresooz P. The importance of the tumor microenvironment in the therapeutic management of cancer. Expert Rev Anticancer Ther 2015; 15:943-54. [PMID: 26098949 DOI: 10.1586/14737140.2015.1059279] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Tumor prognosis is generally defined by various tumor parameters. However, it is well known that paracrine, endocrine and cell-cell interactions between the tumor and its microenvironment contribute to its growth. The tumor microenvironment (TME) can also influence disease prognosis and is likely to be considered as an important prognostic factor. In addition, conventional therapies can influence the microenvironment and antitumor immunity. Similarly, the TME will influence the effectiveness of therapy. The purpose of this review is to demonstrate how TME is important in therapeutic management. Key interactions between TME and different cancer therapies as well as their current clinical consequences have been described. More research is needed to establish the important network between tumor cells and their environment to highlight their relationships with conventional therapies and develop global therapeutic strategies.
Collapse
Affiliation(s)
- Charles Pottier
- Department of Pathology, University Hospital of Liège, Liège, Belgium
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Klinke DJ. Enhancing the discovery and development of immunotherapies for cancer using quantitative and systems pharmacology: Interleukin-12 as a case study. J Immunother Cancer 2015; 3:27. [PMID: 26082838 PMCID: PMC4468964 DOI: 10.1186/s40425-015-0069-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Accepted: 04/28/2015] [Indexed: 12/22/2022] Open
Abstract
Recent clinical successes of immune checkpoint modulators have unleashed a wave of enthusiasm associated with cancer immunotherapy. However, this enthusiasm is dampened by persistent translational hurdles associated with cancer immunotherapy that mirror the broader pharmaceutical industry. Specifically, the challenges associated with drug discovery and development stem from an incomplete understanding of the biological mechanisms in humans that are targeted by a potential drug and the financial implications of clinical failures. Sustaining progress in expanding the clinical benefit provided by cancer immunotherapy requires reliably identifying new mechanisms of action. Along these lines, quantitative and systems pharmacology (QSP) has been proposed as a means to invigorate the drug discovery and development process. In this review, I discuss two central themes of QSP as applied in the context of cancer immunotherapy. The first theme focuses on a network-centric view of biology as a contrast to a "one-gene, one-receptor, one-mechanism" paradigm prevalent in contemporary drug discovery and development. This theme has been enabled by the advances in wet-lab capabilities to assay biological systems at increasing breadth and resolution. The second theme focuses on integrating mechanistic modeling and simulation with quantitative wet-lab studies. Drawing from recent QSP examples, large-scale mechanistic models that integrate phenotypic signaling-, cellular-, and tissue-level behaviors have the potential to lower many of the translational hurdles associated with cancer immunotherapy. These include prioritizing immunotherapies, developing mechanistic biomarkers that stratify patient populations and that reflect the underlying strength and dynamics of a protective host immune response, and facilitate explicit sharing of our understanding of the underlying biology using mechanistic models as vehicles for dialogue. However, creating such models require a modular approach that assumes that the biological networks remain similar in health and disease. As oncogenesis is associated with re-wiring of these biological networks, I also describe an approach that combines mechanistic modeling with quantitative wet-lab experiments to identify ways in which malignant cells alter these networks, using Interleukin-12 as an example. Collectively, QSP represents a new holistic approach that may have profound implications for how translational science is performed.
Collapse
Affiliation(s)
- David J Klinke
- Department of Chemical Engineering and Mary Babb Randolph Cancer Center, West Virginia University, Morgantown, WV 25606 USA
| |
Collapse
|
27
|
Abstract
Research over the past decade has revealed the increasingly complex biologic features of the CD4(+) T-cell lineage. This T-cell subset, which was originally defined on the basis of helper activity in antibody responses, expresses receptors that recognize peptides that have been processed and presented by specialized antigen-presenting cells. At the core of the adaptive immune response, CD4 T cells display a large degree of plasticity and the ability to differentiate into multiple sublineages in response to developmental and environmental cues. These differentiated sublineages can orchestrate a broad range of effector activities during the initiation, expansion, and memory phase of an immune response. The contribution of CD4 cells to host defense against pathogenic invasion and regulation of autoimmunity is now well established. Emerging evidence suggests that CD4 cells also actively participate in shaping antitumor immunity. Here, we outline the biologic properties of CD4 T-cell subsets with an emphasis on their contribution to the antitumor response.
Collapse
Affiliation(s)
- Hye-Jung Kim
- Authors' Affiliations: Department of Microbiology & Immunobiology, Division of Immunology, Harvard Medical School, Boston, Massachusetts
| | | |
Collapse
|
28
|
Wang Q, Klinke DJ, Wang Z. CD8(+) T cell response to adenovirus vaccination and subsequent suppression of tumor growth: modeling, simulation and analysis. BMC SYSTEMS BIOLOGY 2015; 9:27. [PMID: 26048402 PMCID: PMC4458046 DOI: 10.1186/s12918-015-0168-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Accepted: 05/15/2015] [Indexed: 01/08/2023]
Abstract
BACKGROUND Using immune checkpoint modulators in the clinic to increase the number and activity of cytotoxic T lymphocytes that recognize tumor antigens can prolong survival for metastatic melanoma. Yet, only a fraction of the patient population receives clinical benefit. In short, these clinical trials demonstrate proof-of-principle but optimizing the specific therapeutic strategies remains a challenge. In many fields, CAD (computer-aided design) is a tool used to optimize integrated system behavior using a mechanistic model that is based upon knowledge of constitutive elements. The objective of this study was to develop a predictive simulation platform for optimizing anti-tumor immunity using different treatment strategies. METHODS To better understand the therapeutic role that cytotoxic CD8(+) T cells can play in controlling tumor growth, we developed a multi-scale mechanistic model of the biology using impulsive differential equations and calibrated it to a self-consistent data set. RESULTS The multi-scale model captures the activation and differentiation of naïve CD8(+) T cells into effector cytotoxic T cells in the lymph node following adenovirus-mediated vaccination against a tumor antigen, the trafficking of the resulting cytotoxic T cells into blood and tumor microenvironment, the production of cytokines within the tumor microenvironment, and the interactions between tumor cells, T cells and cytokines that control tumor growth. The calibrated model captures the modest suppression of tumor cell growth observed in the B16F10 model, a transplantable mouse model for metastatic melanoma, and was used to explore the impact of multiple vaccinations on controlling tumor growth. CONCLUSIONS Using the calibrated mechanistic model, we found that the cytotoxic CD8(+) T cell response was prolonged by multiple adenovirus vaccinations. However, the strength of the immune response cannot be improved enough by multiple adenovirus vaccinations to reduce tumor burden if the cytotoxic activity or local proliferation of cytotoxic T cells in response to tumor antigens is not greatly enhanced. Overall, this study illustrates how mechanistic models can be used for in silico screening of the optimal therapeutic dosage and timing in cancer treatment.
Collapse
Affiliation(s)
- Qing Wang
- Department of Computer Sciences, Mathematics, and Engineering, Shepherd University, Shepherdstown, 25443, WV, USA.
| | - David J Klinke
- Department of Chemical Engineering and Mary Babb Randolph Cancer Center, West Virginia University, Morgantown, 25606, WV, USA. .,Department of Microbiology, Immunology, & Cell Biology, West Virginia University, Morgantown, 25606, WV, USA.
| | - Zhijun Wang
- Department of Computer Sciences, Mathematics, and Engineering, Shepherd University, Shepherdstown, 25443, WV, USA.
| |
Collapse
|
29
|
|
30
|
Targeting cancer-specific mutations by T cell receptor gene therapy. Curr Opin Immunol 2015; 33:112-9. [PMID: 25728991 DOI: 10.1016/j.coi.2015.02.005] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2015] [Revised: 01/22/2015] [Accepted: 02/10/2015] [Indexed: 12/30/2022]
Abstract
The ease of sequencing the cancer genome, identifying all somatic mutations and grafting mutation-specific T cell receptor (TCR) genes into T cells for adoptive transfer allow, for the first time, a truly tumor-specific and effective therapy. Mutation-specific TCR gene therapy might achieve optimal efficacy with least possible toxicity. Recent clinical data confirm the long-standing evidence from experimental cancer models that antigens encoded by the tumor-specific somatic mutations are potentially the best targets for adoptive T cell therapy. Open questions are, how many somatic mutations create suitable epitopes, whether only individual-specific or also recurrent somatic mutations qualify as suitable epitopes and how neoantigen-specific TCRs are most efficiently obtained. Tumor heterogeneity needs to be considered; therefore, it will be important to identify immunogenic driver mutations that occurred early, are essential for cancer cell survival and present in all cancer cells.
Collapse
|
31
|
Binder DC, Engels B, Arina A, Yu P, Slauch JM, Fu YX, Karrison T, Burnette B, Idel C, Zhao M, Hoffman RM, Munn DH, Rowley DA, Schreiber H. Antigen-specific bacterial vaccine combined with anti-PD-L1 rescues dysfunctional endogenous T cells to reject long-established cancer. Cancer Immunol Res 2015; 1:123-33. [PMID: 24455752 DOI: 10.1158/2326-6066.cir-13-0058] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Immunogenic tumors grow progressively even when heavily infiltrated by CD8(+) T cells. We investigated how to rescue CD8(+) T cell function in long-established immunogenic melanomas that contained a high percentage of endogenous PD-1(+) tumor-specific CD8(+) T cells that were dysfunctional. Treatment with αPD-L1 and αCTLA-4 blocking antibodies did not prevent tumors from progressing rapidly. We then tested exogenous tumor-specific antigen delivery into tumors using Salmonella Typhimurium A1-R to increase antigen levels and generate a proinflammatory tumor microenvironment. Antigen-producing A1-R rescued the endogenous tumor-specific CD8(+) T cell response: proliferation was induced in the lymphoid organs and effector function was recovered in the tumor. Treatment with antigen-producing A1-R led to improved mouse survival and resulted in 32% rejection of long-established immunogenic melanomas. Following treatment with antigen-producing A1-R, the majority of tumor-specific CD8(+) T cells still expressed a high level of PD-1 in the tumor. Combining antigen-producing A1-R with αPD-L1 blocking antibody enhanced the expansion of tumor-specific CD8(+) T cells and resulted in 80% tumor rejection. Collectively, these data demonstrate a powerful new therapeutic approach to rescue dysfunctional endogenous tumor-specific CD8(+) T cells and eradicate advanced immunogenic tumors.
Collapse
Affiliation(s)
- David C Binder
- Committee on Cancer Biology, The University of Chicago, Chicago, IL 60637 ; Department of Pathology, The University of Chicago, Chicago, IL 60637
| | - Boris Engels
- Department of Pathology, The University of Chicago, Chicago, IL 60637 ; Committee on Immunology, The University of Chicago, Chicago, IL 60637
| | - Ainhoa Arina
- Department of Pathology, The University of Chicago, Chicago, IL 60637 ; Committee on Immunology, The University of Chicago, Chicago, IL 60637
| | - Ping Yu
- Committee on Immunology, The University of Chicago, Chicago, IL 60637 ; Department of Medicine, The University of Chicago, Chicago, IL 60637
| | - James M Slauch
- Department of Microbiology and College of Medicine, University of Illinois, Urbana, IL 61801
| | - Yang-Xin Fu
- Department of Pathology, The University of Chicago, Chicago, IL 60637 ; Committee on Immunology, The University of Chicago, Chicago, IL 60637
| | - Theodore Karrison
- Department of Health Sciences, The University of Chicago, Chicago, IL 60637
| | - Byron Burnette
- Department of Radiation Oncology, The University of Chicago, Chicago, IL 60637
| | - Christian Idel
- Department of Pathology, The University of Chicago, Chicago, IL 60637 ; Committee on Immunology, The University of Chicago, Chicago, IL 60637 ; Department of Otorhinolaryngology, University of Lübeck, Lübeck, Germany
| | - Ming Zhao
- AntiCancer, Inc., San Diego, CA 92111
| | - Robert M Hoffman
- AntiCancer, Inc., San Diego, CA 92111 ; Dept. of Surgery, University of California San Diego, San Diego, CA 92103-8220
| | - David H Munn
- Georgia Health Sciences University Cancer Center, Augusta, GA 30912
| | - Donald A Rowley
- Department of Pathology, The University of Chicago, Chicago, IL 60637
| | - Hans Schreiber
- Committee on Cancer Biology, The University of Chicago, Chicago, IL 60637 ; Department of Pathology, The University of Chicago, Chicago, IL 60637 ; Committee on Immunology, The University of Chicago, Chicago, IL 60637
| |
Collapse
|
32
|
Affiliation(s)
- Lorenzo Galluzzi
- Université Paris Descartes/Paris V, Sorbonne Paris Cité; Paris, France; ; Equipe 11 labellisée par la Ligue Nationale Contre le Cancer; Centre de Recherche des Cordeliers; Paris, France; ; Institut Gustave Roussy; Villejuif, France
| | | |
Collapse
|
33
|
Dai M, Yip YY, Hellstrom I, Hellstrom KE. Curing mice with large tumors by locally delivering combinations of immunomodulatory antibodies. Clin Cancer Res 2014; 21:1127-38. [PMID: 25142145 DOI: 10.1158/1078-0432.ccr-14-1339] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
PURPOSE Immunomodulatory mAbs can treat cancer, but cures are rare except for small tumors. Our objective was to explore whether the therapeutic window increases by combining mAbs with different modes of action and injecting them into tumors. EXPERIMENTAL DESIGN Combinations of mAbs to CD137/PD-1/CTLA-4 or CD137/PD-1/CTLA-4/CD19 were administrated intratumorally to mice with syngeneic tumors (B16 and SW1 melanoma, TC1 lung carcinoma), including tumors with a mean surface of approximately 80 mm(2). Survival and tumor growth were assessed. Immunologic responses were evaluated using flow cytometry and qRT-PCR. RESULTS More than 50% of tumor-bearing mice had complete regression and long-term survival after tumor injection with mAbs recognizing CD137/PD-1/CTLA-4/CD19 with similar responses in three models. Intratumoral injection was more efficacious than intraperitoneal injection in causing rejection also of untreated tumors in the same mice. The three-mAb combination could also induce regression, but was less efficacious. There were few side effects, and therapy-resistant tumors were not observed. Transplanted tumor cells rapidly caused a Th2 response with increased CD19 cells. Successful therapy shifted this response to the Th1 phenotype with decreased CD19 cells and increased numbers of long-term memory CD8 effector cells and T cells making IFNγ and TNFα. CONCLUSIONS Intratumoral injection of mAbs recognizing CD137/PD-1/CTLA-4/CD19 can eradicate established tumors and reverse a Th2 response with tumor-associated CD19 cells to Th1 immunity, whereas a combination lacking anti-CD19 is less effective. There are several human cancers for which a similar approach may provide clinical benefit.
Collapse
Affiliation(s)
- Min Dai
- University of Washington Harborview Medical Center, Department of Pathology, Seattle, Washington
| | - Yuen Yee Yip
- University of Washington Harborview Medical Center, Department of Pathology, Seattle, Washington
| | - Ingegerd Hellstrom
- University of Washington Harborview Medical Center, Department of Pathology, Seattle, Washington
| | - Karl Erik Hellstrom
- University of Washington Harborview Medical Center, Department of Pathology, Seattle, Washington.
| |
Collapse
|
34
|
Schirrmacher V. Complete remission of cancer in late-stage disease by radiation and transfer of allogeneic MHC-matched immune T cells: lessons from GvL studies in animals. Cancer Immunol Immunother 2014; 63:535-43. [PMID: 24610041 PMCID: PMC11029222 DOI: 10.1007/s00262-014-1530-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Accepted: 02/25/2014] [Indexed: 12/01/2022]
Abstract
Most immunotherapy studies in animal tumor models are performed in early stages of the disease. Reports on the studies of treatment in late stages of tumor growth and metastasis are much rarer. To guide future efforts for treatment in late-stage disease, a model of effective immune rejection of advanced metastasized cancer is reviewed and lessons therefrom are summarized. Already cachectic DBA/2 mice with a subcutaneously transplanted syngeneic tumor (ESb-MP lymphoma) of 1.5 cm diameter and with macroscopic liver and kidney metastases at 4 weeks could be successfully treated by a combination of sublethal (5 Gy) irradiation followed by a single transfer of 20 million anti-tumor immune spleen cells from tumor-resistant allogeneic MHC-B10.D2 mice. Following intravenous cell transfer, the primary tumors became encapsulated and were eventually rejected from the skin while visceral metastases gradually disappeared leaving behind only scar tissue. There was wound-healing at the site of the rejected primary tumor, and the animals survived long term without any tumor recurrence. The complete eradication of late-stage disease by adoptive cellular immunotherapy could be corroborated noninvasively by (31)P-NMR spectroscopy of primary tumors and by (1)H-NMR microimaging of liver metastases. Conclusions from functional mechanistic studies in this model are summarized and clinical implications discussed.
Collapse
Affiliation(s)
- Volker Schirrmacher
- German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany,
| |
Collapse
|
35
|
Brennen WN, Drake CG, Isaacs JT. Enhancement of the T-cell armamentarium as a cell-based therapy for prostate cancer. Cancer Res 2014; 74:3390-5. [PMID: 24747912 DOI: 10.1158/0008-5472.can-14-0249] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Prostate cancer is frequently characterized by a large inflammatory infiltrate that includes T cells. Although T cells traffic to cancer lesions in large numbers, they are unable to generate a therapeutic response because of the immunosuppressive microenvironment. Therefore, arming T cells with a cytotoxic agent that is capable of killing cancer cells independent of these immunosuppressive signals is a rational approach to enhance their potency. Essentially, the T cells would serve as a cell-based vector, or "Trojan Horse," to selectively deliver a protoxin to disseminated prostate cancer lesions. The selective delivery of a protoxin using T cells represents an ideal method to maximize their therapeutic potency through a "field effect." Because systemically infused T cells are expected to traffic to sites of inflammation other than cancer, an additional level of specificity may be needed to prevent toxicity to nontarget tissues. Toward this goal, genetic engineering can be used to make protoxin expression dependent upon T-cell recognition of the prostate-specific membrane antigen by a chimeric antigen receptor. Furthermore, selective activation of the protoxin using a tissue- or tumor-specific protease, such as PSA, can promote further specificity. Thus, T-cell potency can be enhanced by targeted protoxin secretion and greater specificity achieved using combinatorial antigen recognition and protoxin activation.
Collapse
Affiliation(s)
- W Nathaniel Brennen
- Authors' Affiliations: Chemical Therapeutics Program; Department of Oncology;
| | - Charles G Drake
- Department of Oncology; Department of Immunology; and Department of Urology, The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland
| | - John T Isaacs
- Authors' Affiliations: Chemical Therapeutics Program; Department of Oncology; Department of Urology, The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland
| |
Collapse
|
36
|
Elimination of progressive mammary cancer by repeated administrations of chimeric antigen receptor-modified T cells. Mol Ther 2014; 22:1029-38. [PMID: 24572294 DOI: 10.1038/mt.2014.28] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2013] [Accepted: 02/18/2014] [Indexed: 12/11/2022] Open
Abstract
Continuous oncogenic processes that generate cancer require an on-going treatment approach to eliminate the transformed cells, and prevent their further development. Here, we studied the ability of T cells expressing a chimeric antibody-based receptor (CAR) to offer a therapeutic benefit for breast cancer induced by erbB-2. We tested CAR-modified T cells (T-bodies) specific to erbB-2 for their antitumor potential in a mouse model overexpressing a human erbB-2 transgene that develops mammary tumors. Comparing the antitumor reactivity of CAR-modified T cells under various therapeutic settings, either prophylactic, prior to tumor development, or therapeutically. We found that repeated administration of CAR-modified T cells is required to eliminate spontaneously developing mammary cancer. Systemic, as well as intratumoral administered CAR-modified T cells accumulated at tumor sites and eventually eliminated the malignant cells. Interestingly, within a few weeks after a single CAR T cells' administration, and rejection of primary lesion, tumors usually relapsed both in treated mammary gland and at remote sites; however, repeated injections of CAR-modified T cells were able to control the secondary tumors. Since spontaneous tumors can arise repeatedly, especially in the case of syndromes characterized by specific susceptibility to cancer, multiple administrations of CAR-modified T cells can serve to control relapsing disease.
Collapse
|
37
|
Binder DC, Schreiber H. Dual blockade of PD-1 and CTLA-4 combined with tumor vaccine effectively restores T-cell rejection function in tumors--letter. Cancer Res 2014; 74:632; discussion 635. [PMID: 24408925 DOI: 10.1158/0008-5472.can-13-2216] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- David C Binder
- Authors' Affiliations: Committees on Cancer Biology and Immunology, and Department of Pathology, The University of Chicago, Chicago, Illinois
| | | |
Collapse
|
38
|
Arina A, Schreiber K, Binder DC, Karrison TG, Liu RB, Schreiber H. Adoptively transferred immune T cells eradicate established tumors despite cancer-induced immune suppression. THE JOURNAL OF IMMUNOLOGY 2013; 192:1286-93. [PMID: 24367029 DOI: 10.4049/jimmunol.1202498] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Myeloid-derived CD11b(+)Gr1(+) suppressor cells (MDSCs) and tumor-associated macrophages (TAMs) are considered a major obstacle for effective adoptive T cell therapy. Myeloid cells suppress naive T cell proliferation ex vivo and can prevent the generation of T cell responses in vivo. We find, however, that adoptively transferred immune T cells eradicate well-established tumors in the presence of MDSCs and TAMs, which are strongly immunosuppressive ex vivo. These MDSCs and TAMs were comparable in numbers and immunosuppressive capacity among different tumor models. Longitudinal microscopy of tumors in vivo revealed that after T cell transfer, tumor vasculature and cancer cells disappeared simultaneously. During T cell-mediated tumor destruction, the tumor stroma contained abundant myeloid cells (mainly TAMs) that retained their suppressive properties. Preimmunized but not naive mice resisted immune suppression caused by an unrelated tumor burden, supporting the idea that in vivo, myeloid immunosuppressive cells can suppress naive but not memory T cell responses.
Collapse
Affiliation(s)
- Ainhoa Arina
- Department of Pathology, University of Chicago, Chicago, IL 60637
| | | | | | | | | | | |
Collapse
|
39
|
|
40
|
DuPage M, Jacks T. Genetically engineered mouse models of cancer reveal new insights about the antitumor immune response. Curr Opin Immunol 2013; 25:192-9. [PMID: 23465466 DOI: 10.1016/j.coi.2013.02.005] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2013] [Accepted: 02/08/2013] [Indexed: 01/21/2023]
Abstract
Cancer is a complex disease that can originate in virtually all the tissues of the body, and tumors progress through many different stages during their development. While genetic mutations in the emerging cancer cells drive this disease, it has become increasingly clear that cancer development is strongly influenced by the surrounding microenvironment. Cells of the immune system are critical components of this extrinsic network of cancer regulators, contributing significantly to the microenvironment of most cancers and either promoting or inhibiting the initiation and progression of this disease. Genetically engineered mouse (GEM) mouse models of spontaneous cancer are starting to shape our understanding of how antitumor T cells may act to prevent or inhibit cancer progression in some settings and not others. Lessons learned from investigating spontaneous mouse cancer models have important implications for directing clinical efforts that attempt to direct a cancer patient's immune system to eradicate their disease.
Collapse
Affiliation(s)
- Michel DuPage
- Koch Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | | |
Collapse
|
41
|
Listopad JJ, Kammertoens T, Anders K, Silkenstedt B, Willimsky G, Schmidt K, Kuehl AA, Loddenkemper C, Blankenstein T. Fas expression by tumor stroma is required for cancer eradication. Proc Natl Acad Sci U S A 2013; 110:2276-81. [PMID: 23341634 PMCID: PMC3568383 DOI: 10.1073/pnas.1218295110] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The contribution of molecules such as perforin, IFN-γ (IFNγ), and particularly Fas ligand (FasL) by transferred CD8(+) effector T (T(E)) cells to rejection of large, established tumors is incompletely understood. Efficient attack against large tumors carrying a surrogate tumor antigen (mimicking a "passenger" mutation) by T(E) cells requires action of IFNγ on tumor stroma cells to avoid selection of antigen-loss variants. Because "cancer-driving" antigens (CDAs) are rarely counterselected, IFNγ may be expected to be dispensable in elimination of cancers by targeting a CDA. Here, initial regression of large, established tumors required neither IFNγ, FasL, nor perforin by transferred CD8(+) T(E) cells targeting Simian Virus (SV) 40 large T as CDA. However, cytotoxic T(E) cells lacking IFNγ or FasL could not prevent relapse despite retention of the rejection antigen by the cancer cells. Complete tumor rejection required IFNγ-regulated Fas by the tumor stroma. Therefore, T(E) cells lacking IFNγ or FasL cannot prevent progression of antigenic cancer because the tumor stroma escapes destruction if its Fas expression is down-regulated.
Collapse
Affiliation(s)
- Joanna J. Listopad
- Max Delbrück Center for Molecular Medicine, 13125 Berlin, Germany; and
- Institute of Immunology
| | - Thomas Kammertoens
- Max Delbrück Center for Molecular Medicine, 13125 Berlin, Germany; and
- Institute of Immunology
| | - Kathleen Anders
- Max Delbrück Center for Molecular Medicine, 13125 Berlin, Germany; and
| | | | - Gerald Willimsky
- Max Delbrück Center for Molecular Medicine, 13125 Berlin, Germany; and
- Institute of Immunology
| | | | - Anja A. Kuehl
- Department of Internal Medicine, Rheumatology and Clinical Immunology, and
| | | | - Thomas Blankenstein
- Max Delbrück Center for Molecular Medicine, 13125 Berlin, Germany; and
- Institute of Immunology
| |
Collapse
|
42
|
Soto CM, Stone JD, Chervin AS, Engels B, Schreiber H, Roy EJ, Kranz DM. MHC-class I-restricted CD4 T cells: a nanomolar affinity TCR has improved anti-tumor efficacy in vivo compared to the micromolar wild-type TCR. Cancer Immunol Immunother 2012; 62:359-69. [PMID: 22926060 DOI: 10.1007/s00262-012-1336-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2012] [Accepted: 08/03/2012] [Indexed: 12/29/2022]
Abstract
Clinical studies with immunotherapies for cancer, including adoptive cell transfers of T cells, have shown promising results. It is now widely believed that recruitment of CD4(+) helper T cells to the tumor would be favorable, as CD4(+) cells play a pivotal role in cytokine secretion as well as promoting the survival, proliferation, and effector functions of tumor-specific CD8(+) cytotoxic T lymphocytes. Genetically engineered high-affinity T-cell receptors (TCRs) can be introduced into CD4(+) helper T cells to redirect them to recognize MHC-class I-restricted antigens, but it is not clear what affinity of the TCR will be optimal in this approach. Here, we show that CD4(+) T cells expressing a high-affinity TCR (nanomolar K (d) value) against a class I tumor antigen mediated more effective tumor treatment than the wild-type affinity TCR (micromolar K (d) value). High-affinity TCRs in CD4(+) cells resulted in enhanced survival and long-term persistence of effector memory T cells in a melanoma tumor model. The results suggest that TCRs with nanomolar affinity could be advantageous for tumor targeting when expressed in CD4(+) T cells.
Collapse
Affiliation(s)
- Carolina M Soto
- Neuroscience Program, University of Illinois, Urbana, IL 61801, USA
| | | | | | | | | | | | | |
Collapse
|
43
|
Schreiber K, Arina A, Engels B, Spiotto MT, Sidney J, Sette A, Karrison TG, Weichselbaum RR, Rowley DA, Schreiber H. Spleen cells from young but not old immunized mice eradicate large established cancers. Clin Cancer Res 2012; 18:2526-33. [PMID: 22415314 PMCID: PMC5354938 DOI: 10.1158/1078-0432.ccr-12-0127] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
PURPOSE Solid tumors that have grown two weeks or longer in mice and have diameters larger than 1 cm are histologically indistinguishable from autochthonous human cancers. When experimental tumors reach this clinically relevant size, they are usually refractory to most immunotherapies but may be destroyed by adoptive T-cell transfer. However, TCR-transgenic T cells and/or tumor cells overexpressing antigens are frequently used in these experiments. Here we studied the requirements for destroying clinical size, unmanipulated 8101 tumors by adoptive cell therapy. EXPERIMENTAL DESIGN 8101 arose in an old mouse after chronic exposure to UV light. A cancer line was established, which was never serially transplanted. The immunodominant CD8(+) T cell-recognized antigen of this tumor is caused by a somatic tumor-specific mutation in the RNA helicase p68. 8101 tumors were treated with spleen cells from young naive, or young and old immunized mice to ascertain the characteristics of immune cells that lead to rejection. RESULTS Here we show that the mutant p68 peptide has an exceptionally high affinity to the presenting MHC class I molecule K(b) and that spleen cells from immunized young syngeneic mice adoptively transferred to Rag(-/-) or cancer-suppressed euthymic mice eradicate 8101 tumors larger than 1 cm in average diameter and established for several weeks. Spleen cells from naive young mice or from old and boosted (reimmunized) mice were ineffective. CONCLUSIONS Relapse-free destruction of large and long-established tumors expressing a genuine very high-affinity tumor-specific antigen can be achieved by using adoptive transfer of lymphocytes from immunized young individuals.
Collapse
Affiliation(s)
- Karin Schreiber
- Department of Pathology, The University of Chicago, Chicago, Illinois 60637, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|