1
|
Yu Z, Shi J, Fang Y, Zhao Y, Xu A, Li N. Developing innovative strategies of tumor‑infiltrating lymphocyte therapy for tumor treatment. Oncol Rep 2024; 51:85. [PMID: 38666543 PMCID: PMC11082634 DOI: 10.3892/or.2024.8744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 04/15/2024] [Indexed: 05/04/2024] Open
Abstract
Cancer is the main cause of global mortality, and thus far, effective therapeutic strategies for cancer treatment are in high demand. Adoptive transfer of tumor‑infiltrating lymphocytes (TILs) represents a promising avenue in immunotherapy for the management of malignancies. The clinical safety and efficacy of TIL‑based therapy have been established through numerous rigorous clinical trials. However, the efficacy of TIL infusion in inducing an anti‑tumor response is limited to a subset of clinical patients with cancer. Therefore, there is an urgent need to develop innovative strategies aimed at enhancing the effectiveness of TIL‑based therapy. In the present review, the developmental history of TIL‑based therapy was systematically summarized and analyzed, while also presenting a unique perspective on enhancing the multi‑dimensional anti‑tumor capabilities of TILs. The insight and conclusions presented in this review may contribute to improving the efficacy of TIL‑based therapy and expediting its development.
Collapse
Affiliation(s)
- Zhongjie Yu
- R&D, Qingdao Sino-cell Biomedicine Co., Ltd., Qingdao, Shandong 266000, P.R. China
| | - Jianhua Shi
- Phase I Clinical Research Center Affiliated, Linyi Tumor Hospital, Linyi, Shandong 276000, P.R. China
| | - Yuan Fang
- Clinical Trials Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, P.R. China
| | - Yi Zhao
- R&D, Qingdao Sino-cell Biomedicine Co., Ltd., Qingdao, Shandong 266000, P.R. China
| | - Aotian Xu
- R&D, Qingdao Sino-cell Biomedicine Co., Ltd., Qingdao, Shandong 266000, P.R. China
| | - Ning Li
- Clinical Trials Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, P.R. China
| |
Collapse
|
2
|
Tumor Infiltrating Lymphocyte (TIL) Therapy for Solid Tumor Treatment: Progressions and Challenges. Cancers (Basel) 2022; 14:cancers14174160. [PMID: 36077696 PMCID: PMC9455018 DOI: 10.3390/cancers14174160] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 08/22/2022] [Accepted: 08/25/2022] [Indexed: 12/15/2022] Open
Abstract
Over the past decade, immunotherapy, especially cell-based immunotherapy, has provided new strategies for cancer therapy. Recent clinical studies demonstrated that adopting cell transfer of tumor-infiltrating lymphocytes (TILs) for advanced solid tumors showed good efficacy. TIL therapy is a type of cell-based immunotherapy using the patient’s own immune cells from the microenvironment of the solid tumor to kill tumor cells. In this review, we provide a comprehensive summary of the current strategies and challenges in TIL isolation and generation. Moreover, the current clinical experience of TIL therapy is summarized and discussed, with an emphasis on lymphodepletion regimen, the use of interleukin-2, and related toxicity. Furthermore, we highlight the clinical trials where TIL therapy is used independently and in combination with other types of therapy for solid cancers. Finally, the limitations, future potential, and directions of TIL therapy for solid tumor treatment are also discussed.
Collapse
|
3
|
Li M, Zhu Y, Bai B, Fang J, Yao W, Li Y, Li S, Li X, Jin N, Jiang R. Suppression effect of a dual cancer-specific oncolytic adenovirus on luciferase-labeled human melanoma cells in vitro and in vivo. Cancer Biomark 2021; 32:251-262. [PMID: 34459386 DOI: 10.3233/cbm-203150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND To explore the suppressive effect of Apoptin-loaded oncolytic adenovirus (Ad-VT) on luciferase-labeled human melanoma cells in vitro and in vivo. METHODS The stable luciferase-expressing human melanoma cells A375-luc or M14-luc were obtained by transfecting the plasmid pGL4.51 and selection with G418, followed by luciferase activity, genetic stability and bioluminescence intensity assays. In vitro, the inhibitory effects of Ad-VT on A375-luc or M14-luc were evaluated using the MTS cell proliferation, FITC-Annexin V apoptosis detection, transwell migration, Matrigel invasion and scratch assays. The inhibition pathway in Ad-VT-infected A375-luc and M14-luc cells were analyzed by JC-1 staining and Western-blot detection of mitochondrial apoptosis-related proteins. In vivo, the suppressive effects of Ad-VT on A375-luc or M14-luc were assessed by living imaging technology, tumor volume, bioluminescence intensity, survival curves and immunohistochemical analysis of the tumors from the xenograft tumor model BALB/c nude mice. RESULTS The growth and migration of A375-luc and M14-luc were significantly inhibited by Ad-VT in vitro. The evaluations of A375-luc and M14-luc tumor models in BALB/c nude mice were successfully performed using living imaging technology. Ad-VT had an anti-tumor effect by reducing tumor growth and increasing survival in vivo. Ad-VT significantly changed the mitochondrial membrane potential by triggering the the mitochondrial release of apoptosis-related proteins, AIF (apoptosis inducing factor), ARTS (Apoptosis-Related Proteins), and Cyto-c (cytochrome c) from the mitochondria. CONCLUSION Ad-VT reduced the mitochondrial membrane potential in A375-luc or M14-luc cells and induced the mitochondrial release of AIF, ARTS and Cyto-C. Ad-VT induced apoptosis in A375-luc or M14-luc cells via the mitochondrial apoptotic pathway.
Collapse
Affiliation(s)
- Min Li
- Department of Dermatology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Yilong Zhu
- Academicians Workstation of Jilin Province, Changchun University of Chinese Medicine, Changchun, Jilin, China.,Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Institute of Military Veterinary Medicine, Academy of Military Medical Sciences, Changchun, Jilin, China
| | - Bing Bai
- Academicians Workstation of Jilin Province, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Jinbo Fang
- Academicians Workstation of Jilin Province, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Wei Yao
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Institute of Military Veterinary Medicine, Academy of Military Medical Sciences, Changchun, Jilin, China.,Center for Disease Control and Prevention, Agency for Offices Administration, Central Military Commission, Beijing, China
| | - Yiquan Li
- Academicians Workstation of Jilin Province, Changchun University of Chinese Medicine, Changchun, Jilin, China.,Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Institute of Military Veterinary Medicine, Academy of Military Medical Sciences, Changchun, Jilin, China
| | - Shanzhi Li
- Academicians Workstation of Jilin Province, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Xiao Li
- Department of Dermatology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China.,Academicians Workstation of Jilin Province, Changchun University of Chinese Medicine, Changchun, Jilin, China.,Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Institute of Military Veterinary Medicine, Academy of Military Medical Sciences, Changchun, Jilin, China
| | - Ningyi Jin
- Department of Dermatology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China.,Academicians Workstation of Jilin Province, Changchun University of Chinese Medicine, Changchun, Jilin, China.,Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Institute of Military Veterinary Medicine, Academy of Military Medical Sciences, Changchun, Jilin, China
| | - Rihua Jiang
- Department of Dermatology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
4
|
Cetintas VB, Batada NN. Is there a causal link between PTEN deficient tumors and immunosuppressive tumor microenvironment? J Transl Med 2020; 18:45. [PMID: 32000794 PMCID: PMC6993356 DOI: 10.1186/s12967-020-02219-w] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 01/10/2020] [Indexed: 12/13/2022] Open
Abstract
The PTEN tumor suppressor is the second most commonly inactivated gene across cancer types. While it's role in PI3K/AKT and DNA damage pathways are clear, increasing evidences suggest that PTEN may also promote anti-tumor immunity. PTEN-deficient tumors are characterized by (i) reduced levels of cytotoxic T cells, helper T cells and NK cells, (ii) elevated pro-oncogenic inflammatory cytokines like CCL2 and (iii) increased levels of immunosuppressive cells such as MDSCs and Tregs. An intriguing possibility is that link between PTEN and anti-tumor immunity is mediated by the interferon signaling pathway. In this review, we summarize the evidences for the mechanistic link between PTEN deficiency and immunosuppressive tumor microenvironment and the interferon signaling pathway. We further discuss how the link between these pathways can be exploited for development of personalized immunotherapy for patients with PTEN deficient tumors.
Collapse
Affiliation(s)
- Vildan B Cetintas
- Department of Medical Biology, Faculty of Medicine, Ege University, Izmir, Turkey.,Centre for Genomic and Experimental Medicine, MRC Institute of Genetics & Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - Nizar N Batada
- Centre for Genomic and Experimental Medicine, MRC Institute of Genetics & Molecular Medicine, University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
5
|
Abstract
Skin cancer, as the most physically accessible malignancy, allows for the greatest variety in treatment innovation. The last 2 decades have seen striking increases in the life expectancies of those diagnosed with malignant melanoma. However, many cases remain in which disease prevails against standard treatment, and those patients rely on continuing ingenuity. Drugs that can be injected directly into patients' tumors have become increasingly promising, not least for the reduction in side effects observed. Intratumoral therapy encompasses a wide array of agents, from chemotherapeutic drugs to cancer vaccines. While each show some efficacy, those agents which regulate the immune system likely have the greatest potential for preventing disease progression or recurrence. Recent research has highlighted the importance of the presence of cytotoxic T cells and of keeping regulatory T cells in check. Thus, manipulating the tumor microenvironment is a need in skin cancer therapy, which intratumoral delivery can potentially address. In order to find the best approach to each person's disease, more studies are needed to test intralesional agents in combination with currently approved therapies and with each other.
Collapse
|
6
|
Santos JM, Havunen R, Hemminki A. Modulation of the tumor microenvironment with an oncolytic adenovirus for effective T-cell therapy and checkpoint inhibition. Methods Enzymol 2019; 635:205-230. [PMID: 32122546 DOI: 10.1016/bs.mie.2019.05.043] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Despite exciting proof-of-concept data mediated by adoptive T-cell transfer and checkpoint blockade, major challenges imposed by the tumor microenvironment restrict clinical benefits to a minority of patients with advanced or metastatic solid malignancies. While employment of toxic pre- and postconditioning regimens to circumvent the inefficacy of T-cell transfer presents a fundamental problem for heavily pretreated cancer patients, for checkpoint blockade, the main issue relates to low single-agent response rates. To overcome these hurdles, combination therapy with oncolytic adenovirus is becoming an attractive solution given multiple intrinsic modulatory effects on the intratumoral immune compartment, engineering capabilities and safety profile. Here, we provide a short overview on the tumor microenvironmental challenges in solid tumors, and how oncolytic adenoviruses can counteract these barriers. Finally, the immunotherapeutic potential of oncolytic adenoviruses will be discussed in the context of clinical experience with adoptive T-cell therapy and immune checkpoint inhibitors.
Collapse
Affiliation(s)
- João Manuel Santos
- Cancer Gene Therapy Group, Translational Immunology Research Program and Department of Oncology, University of Helsinki, Helsinki, Finland; TILT Biotherapeutics Ltd, Helsinki, Finland
| | - Riikka Havunen
- Cancer Gene Therapy Group, Translational Immunology Research Program and Department of Oncology, University of Helsinki, Helsinki, Finland; TILT Biotherapeutics Ltd, Helsinki, Finland
| | - Akseli Hemminki
- Cancer Gene Therapy Group, Translational Immunology Research Program and Department of Oncology, University of Helsinki, Helsinki, Finland; TILT Biotherapeutics Ltd, Helsinki, Finland; Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland.
| |
Collapse
|
7
|
Castro F, Pinto ML, Almeida R, Pereira F, Silva AM, Pereira CL, Santos SG, Barbosa MA, Gonçalves RM, Oliveira MJ. Chitosan/poly(γ-glutamic acid) nanoparticles incorporating IFN-γ for immune response modulation in the context of colorectal cancer. Biomater Sci 2019; 7:3386-3403. [PMID: 31233057 DOI: 10.1039/c9bm00393b] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
IFN-γ therapy has been approved by the Food and Drug Administration (FDA) for the treatment of chronic granulomatous disease and severe malignant osteopetrosis. Despite the promising IFN-γ-based therapeutic applications, its limited success in clinical trials is related with limitations inherent to its molecular properties and with the difficulties to deliver it locally or with adequate periodicity to achieve a therapeutic effect. We have previously shown that chitosan (Ch)/poly(γ-glutamic acid) (γ-PGA) nanoparticles (NPs) are immunostimulatory, impairing colorectal cancer cell invasion. Ch is a biocompatible cationic polysaccharide extensively studied and already approved for biomedical applications while γ-PGA is a poly(amino acid), biodegradable and negatively charged. Here, we evaluated the potential of Ch/γ-PGA NPs as vehicles for IFN-γ and their ability to modulate immune cells' phenotype. In this study, Ch/IFN-γ/γ-PGA nanoparticles (IFN-γ-NPs) prepared by a co-acervation method, presenting a size of approximately 180 nm and a low polydispersity index, were tested for their immunomodulatory activity. These IFN-γ-NPs induced an immunostimulatory profile on dendritic cells (DCs) with increased cell surface costimulatory molecules and secretion of pro-inflammatory cytokines, including IL-6, IL-12p40 and TNF-α. IFN-γ-NPs also modulated the IL-10-stimulated macrophage profile, increasing their ability to secrete the pro-inflammatory cytokines IL-6, IL-12p40 and TNF-α. Concomitantly, these phenotypic alterations enhanced T cell proliferation. In addition, the ability of DCs and macrophages to induce colorectal cancer cell invasion was hampered in the presence of IFN-γ-NPs. Although the major observations were mediated by Ch/γ-PGA NPs, the incorporation of IFN-γ into NPs potentiated the expression of CD40 and CD86, and the impairment of colorectal cancer cell invasion. This work bridges the previously reported immunostimulatory capacity of Ch/γ-PGA NPs with their potential as carriers for immunomodulatory molecules, like IFN-γ, opening new avenues for their use in clinical settings.
Collapse
Affiliation(s)
- Flávia Castro
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal and INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal. and ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Marta L Pinto
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal and INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal. and CNC - Centro de Neurociências e Biologia Celular, Universidade de Coimbra, Portugal
| | - Rui Almeida
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal and INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal.
| | - Flávia Pereira
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal and INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal. and CBMA - Centro de Biologia Molecular e Ambiental, Universidade do Minho, Braga, Portugal
| | - Andreia M Silva
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal and INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal. and ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Catarina L Pereira
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal and INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal. and ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Susana G Santos
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal and INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal. and ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Mário A Barbosa
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal and INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal. and ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Raquel M Gonçalves
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal and INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal.
| | - Maria J Oliveira
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal and INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal. and Departamento de Patologia e Oncologia, Faculdade de Medicina, Universidade do Porto, Porto, Portugal
| |
Collapse
|
8
|
Cervera-Carrascon V, Havunen R, Hemminki A. Oncolytic adenoviruses: a game changer approach in the battle between cancer and the immune system. Expert Opin Biol Ther 2019; 19:443-455. [PMID: 30905206 DOI: 10.1080/14712598.2019.1595582] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Oncolytic adenoviruses are among the most studied oncolytic viruses because of their tumor selectivity, safety, and transgene-delivery capability. With a growing number of different immunotherapies against cancer, the extraordinary immunogenicity of the adenovirus has emerged as a differentiating strength. Enabling T-cell related therapies with oncolytic adenoviruses appears a promising approach due to its inherent ability to elicit responses from the adaptive immune compartment. AREAS COVERED These viruses have successfully enhanced both adoptive T-cell therapies and immune-checkpoint therapies. Oncolytic viruses induce several effects at the tumor and on the systemic level that help to circumvent current limitations of T-cells and related therapies, such as T-cell trafficking, tumor immune suppressivity and antigen spreading EXPERT OPINION Taking into account the multitude of possibilities of treating cancer with immunotherapies, learning to optimize the combinations and administration strategies of these drugs, could lead to durable responses in patients with currently incurable cancers.
Collapse
Affiliation(s)
- Victor Cervera-Carrascon
- a Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine , University of Helsinki , Helsinki , Finland.,b TILT Biotherapeutics Ltd , Helsinki , Finland
| | - Riikka Havunen
- a Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine , University of Helsinki , Helsinki , Finland.,b TILT Biotherapeutics Ltd , Helsinki , Finland
| | - Akseli Hemminki
- a Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine , University of Helsinki , Helsinki , Finland.,b TILT Biotherapeutics Ltd , Helsinki , Finland.,c Hospital Comprehensive Cancer Center , Helsinki University , Helsinki , Finland
| |
Collapse
|
9
|
Combination immunotherapies implementing adoptive T-cell transfer for advanced-stage melanoma. Melanoma Res 2019. [PMID: 29521881 DOI: 10.1097/cmr.0000000000000436] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Immunotherapy is a promising method of treatment for a number of cancers. Many of the curative results have been seen specifically in advanced-stage melanoma. Despite this, single-agent therapies are only successful in a small percentage of patients, and relapse is very common. As chemotherapy is becoming a thing of the past for treatment of melanoma, the combination of cellular therapies with immunotherapies appears to be on the rise in in-vivo models and in clinical trials. These forms of therapies include tumor-infiltrating lymphocytes, T-cell receptor, or chimeric antigen receptor-modified T cells, cytokines [interleukin (IL-2), IL-15, IL-12, granulocyte-macrophage colony stimulating factor, tumor necrosis factor-α, interferon-α, interferon-γ], antibodies (αPD-1, αPD-L1, αTIM-3, αOX40, αCTLA-4, αLAG-3), dendritic cell-based vaccines, and chemokines (CXCR2). There are a substantial number of ongoing clinical trials using two or more of these combination therapies. Preliminary results indicate that these combination therapies are a promising area to focus on for cancer treatments, especially melanoma. The main challenges with the combination of cellular and immunotherapies are adverse events due to toxicities and autoimmunity. Identifying mechanisms for reducing or eliminating these adverse events remains a critical area of research. Many important questions still need to be elucidated in regard to combination cellular therapies and immunotherapies, but with the number of ongoing clinical trials, the future of curative melanoma therapies is promising.
Collapse
|
10
|
Adenovirus Coding for Interleukin-2 and Tumor Necrosis Factor Alpha Replaces Lymphodepleting Chemotherapy in Adoptive T Cell Therapy. Mol Ther 2018; 26:2243-2254. [PMID: 30017877 DOI: 10.1016/j.ymthe.2018.06.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 06/01/2018] [Accepted: 06/01/2018] [Indexed: 02/08/2023] Open
Abstract
Lymphodepleting preconditioning with high-dose chemotherapy is commonly used to increase the clinical efficacy of adoptive T cell therapy (ACT) strategies, however, with severe toxicity for patients. Conversely, oncolytic adenoviruses are safe and, when engineered to express interleukin-2 (IL-2) and tumor necrosis factor alpha (TNF-α), they can achieve antitumor immunomodulatory effects similar to lymphodepletion. Therefore, we compare the safety and efficacy of such adenoviruses with a cyclophosphamide- and fludarabine-containing lymphodepleting regimen in the setting of ACT. Human adenovirus (Ad5/3-E2F-D24-hTNF-α-IRES-hIL-2; TILT-123) replication was studied using a Syrian hamster pancreatic tumor model (HapT1) infused with tumor-infiltrating lymphocytes (TILs). Using the oncolytic virus instead of lymphodepletion resulted in superior efficacy and survival. Immune cells responsive to TNF-α IL-2 were studied using an immunocompetent mouse melanoma model (B16.OVA) infused with ovalbumin-specific T (OT-I) cells. Here, the adenovirus approach improved tumor control together with increased intratumoral Th1 cytokine levels and infiltration of CD8+ T cells and CD86+ dendritic cells. Similar to humans, lymphodepleting preconditioning caused severe cytopenias, systemic inflammation, and damage to vital organs. Toxicity was minimal in adenovirus- and OT-I-treated mice. These findings demonstrate that ACT can be effectively facilitated by cytokine-coding adenovirus without requiring lymphodepletion, a rationale being clinically investigated.
Collapse
|
11
|
Castro F, Cardoso AP, Gonçalves RM, Serre K, Oliveira MJ. Interferon-Gamma at the Crossroads of Tumor Immune Surveillance or Evasion. Front Immunol 2018; 9:847. [PMID: 29780381 PMCID: PMC5945880 DOI: 10.3389/fimmu.2018.00847] [Citation(s) in RCA: 787] [Impact Index Per Article: 112.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Accepted: 04/05/2018] [Indexed: 12/15/2022] Open
Abstract
Interferon-gamma (IFN-γ) is a pleiotropic molecule with associated antiproliferative, pro-apoptotic and antitumor mechanisms. This effector cytokine, often considered as a major effector of immunity, has been used in the treatment of several diseases, despite its adverse effects. Although broad evidence implicating IFN-γ in tumor immune surveillance, IFN-γ-based therapies undergoing clinical trials have been of limited success. In fact, recent reports suggested that it may also play a protumorigenic role, namely, through IFN-γ signaling insensitivity, downregulation of major histocompatibility complexes, and upregulation of indoleamine 2,3-dioxygenase and of checkpoint inhibitors, as programmed cell-death ligand 1. However, the IFN-γ-mediated responses are still positively associated with patient's survival in several cancers. Consequently, major research efforts are required to understand the immune contexture in which IFN-γ induces its intricate and highly regulated effects in the tumor microenvironment. This review discusses the current knowledge on the pro- and antitumorigenic effects of IFN-γ as part of the complex immune response to cancer, highlighting the relevance to identify IFN-γ responsive patients for the improvement of therapies that exploit associated signaling pathways.
Collapse
Affiliation(s)
- Flávia Castro
- i3S – Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- INEB – Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
- ICBAS – Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Ana Patrícia Cardoso
- i3S – Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- INEB – Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
| | - Raquel Madeira Gonçalves
- i3S – Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- INEB – Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
- ICBAS – Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Karine Serre
- IMM – Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Maria José Oliveira
- i3S – Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- INEB – Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
- Departamento de Patologia e Oncologia, Faculdade de Medicina, Universidade do Porto, Porto, Portugal
| |
Collapse
|
12
|
Adoptive Cell Therapy with Tumor-Infiltrating Lymphocytes in Advanced Melanoma Patients. J Immunol Res 2018; 2018:3530148. [PMID: 29750176 PMCID: PMC5883986 DOI: 10.1155/2018/3530148] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Revised: 01/16/2018] [Accepted: 01/29/2018] [Indexed: 12/02/2022] Open
Abstract
Immunotherapy for melanoma includes adoptive cell therapy with autologous tumor-infiltrating lymphocytes (TILs). This monocenter retrospective study was undertaken to evaluate the efficacy and safety of this treatment of patients with advanced melanoma. All advanced melanoma patients treated with TILs using the same TIL expansion methodology and same treatment interleukin-2 (IL-2) regimen between 2009 and 2012 were included. After sterile intralesional excision of a cutaneous or subcutaneous metastasis, TILs were produced according to a previously described method and then infused into the patient who also received a complementary subcutaneous IL-2 regimen. Nine women and 1 man were treated for unresectable stage IIIC (n = 4) or IV (n = 6) melanoma. All but 1 patient with unresectable stage III melanoma (1st line) had received at least 2 previous treatments, including anti-CTLA-4 antibody for 4. The number of TILs infused ranged from 0.23 × 109 to 22.9 × 109. Regarding safety, no serious adverse effect was reported. Therapeutic responses included a complete remission, a partial remission, 2 stabilizations, and 6 progressions. Among these 4 patients with clinical benefit, 1 is still alive with 9 years of follow-up and 1 died from another cause after 8 years of follow-up. Notably, patients treated with high percentages of CD4 + CD25 + CD127lowFoxp3+ T cells among their TILs had significantly shorter OS. The therapeutic effect of combining TILs with new immunotherapies needs further investigation.
Collapse
|
13
|
T-Cell Therapy Enabling Adenoviruses Coding for IL2 and TNFα Induce Systemic Immunomodulation in Mice With Spontaneous Melanoma. J Immunother 2017; 39:343-354. [PMID: 27741089 DOI: 10.1097/cji.0000000000000144] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The immunosuppressive microenvironment of solid tumors renders adoptively transferred T cells hypofunctional. However, adenoviral delivery of immunostimulatory cytokines IL2 and TNFα can significantly improve the efficacy of adoptive T-cell therapy. Using ret transgenic mice that spontaneously develop skin malignant melanoma, we analyzed the mechanism of action of adenoviruses coding for IL2 and TNFα in combination with adoptive transfer of TCR-transgenic TRP-2-specific T cells. Following T-cell therapy and intratumoral virus injection, a significant increase in antigen-experienced, tumor-reactive PD-1 CD8 T cells was seen in both cutaneous lesions and in metastatic lymph nodes. A reverse correlation between tumor weight and the number of tumor-reactive PD-1 tumor-infiltrating lymphocytes (TILs) was observed, suggesting that these T cells could target and kill tumor cells. It is interesting to note that, local expression of cytokines did not affect intratumoral levels of T-regulatory cells (Tregs), which had previously been associated with systemic IL2 therapy. Instead, Ad5-IL2 induced upregulation of IL2 receptor α-chain (CD25) on conventional CD4CD25Foxp3 cells, indicating that these CD4 T cells may contribute to CD8 T-cell activation and/or homing. Signs of therapy-induced resistance were also observed as the expression of PD-L1 on tumor-infiltrating granulocytic myeloid-derived suppressor cells was upregulated as a reaction to PD-1+ TILs. Finally, beneficial ratios between tumor-reactive PD-1 CD8 TILs and immunosuppressive cell subsets (Tregs and nitric oxide-producing myeloid-derived suppressor cells) were observed in primary and secondary tumor sites, indicating that local delivery of IL2 and TNFα coding adenoviruses can systemically modify the cellular composition of the tumor microenvironment in favor of adoptively transferred T cells.
Collapse
|
14
|
Holanda RA, Muñoz JE, Dias LS, Silva LBR, Santos JRA, Pagliari S, Vieira ÉLM, Paixão TA, Taborda CP, Santos DA, Bruña-Romero O. Recombinant vaccines of a CD4+ T-cell epitope promote efficient control of Paracoccidioides brasiliensis burden by restraining primary organ infection. PLoS Negl Trop Dis 2017; 11:e0005927. [PMID: 28938005 PMCID: PMC5627964 DOI: 10.1371/journal.pntd.0005927] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Revised: 10/04/2017] [Accepted: 09/02/2017] [Indexed: 11/19/2022] Open
Abstract
Paracoccidioidomycosis (PCM) is an infectious disease endemic to South America, caused by the thermally dimorphic fungi Paracoccidioides. Currently, there is no effective human vaccine that can be used in prophylactic or therapeutic regimes. We tested the hypothesis that the immunogenicity of the immunodominant CD4+ T-cell epitope (P10) of Paracoccidioides brasiliensis gp43 antigen might be significantly enhanced by using a hepatitis B virus-derived particle (VLP) as an antigen carrier. This chimera was administered to mice as a (His)6-purified protein (rPbT) or a replication-deficient human type 5 adenoviral vector (rAdPbT) in an immunoprophylaxis assay. The highly virulent Pb18 yeast strain was used to challenge our vaccine candidates. Fungal challenge evoked robust P10-specific memory CD4+ T cells secreting protective Th-1 cytokines in most groups of immunized mice. Furthermore, the highest level of fungal burden control was achieved when rAdPbT was inoculated in a homologous prime-boost regimen, with 10-fold less CFU recovering than in non-vaccinated mice. Systemic Pb18 spreading was only prevented when rAdPbT was previously inoculated. In summary, we present here VLP/P10 formulations as vaccine candidates against PCM, some of which have demonstrated for the first time their ability to prevent progression of this pernicious fungal disease, which represents a significant social burden in developing countries.
Collapse
MESH Headings
- Animals
- Antigens, Fungal/immunology
- CD4-Positive T-Lymphocytes/immunology
- Cytokines/immunology
- Cytokines/metabolism
- Epitopes, T-Lymphocyte/genetics
- Epitopes, T-Lymphocyte/immunology
- Fungal Proteins/immunology
- Fungal Vaccines/administration & dosage
- Fungal Vaccines/immunology
- Glycoproteins/immunology
- Hepatitis B virus/genetics
- Immunization
- Immunodominant Epitopes/immunology
- Immunogenicity, Vaccine
- Immunologic Memory
- Liver/microbiology
- Lung/microbiology
- Mice, Inbred BALB C
- Paracoccidioides/growth & development
- Paracoccidioides/immunology
- Paracoccidioidomycosis/immunology
- Paracoccidioidomycosis/microbiology
- Paracoccidioidomycosis/prevention & control
- Spleen/microbiology
- Th1 Cells/immunology
- Vaccines, Synthetic/administration & dosage
- Vaccines, Synthetic/genetics
- Vaccines, Synthetic/immunology
- Vaccines, Virus-Like Particle/administration & dosage
- Vaccines, Virus-Like Particle/genetics
- Vaccines, Virus-Like Particle/immunology
Collapse
Affiliation(s)
- Rodrigo Assunção Holanda
- Departamento de Microbiologia, Universidade Federal de Minas Gerais, Minas Gerais, Brazil
- Laboratório de Biologia Parasitária, Universidade CEUMA, Maranhão, Brazil
- * E-mail:
| | - Julián Esteban Muñoz
- Departamento de Microbiologia, Universidade de São Paulo, São Paulo, Brazil
- Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogotá, Colombia
| | - Lucas Santos Dias
- Departamento de Microbiologia, Universidade de São Paulo, São Paulo, Brazil
| | | | - Julliana Ribeiro Alves Santos
- Departamento de Microbiologia, Universidade Federal de Minas Gerais, Minas Gerais, Brazil
- Laboratório de Microbiologia Ambiental, Universidade CEUMA, Maranhão, Brazil
| | - Sthefany Pagliari
- Departamento de Microbiologia, Imunologia e Parasitologia, Universidade Federal de Santa Catarina, Santa Catarina, Brazil
| | | | - Tatiane Alves Paixão
- Departamento de Patologia Geral, Universidade Federal de Minas Gerais, Minas Gerais, Brazil
| | | | - Daniel Assis Santos
- Departamento de Microbiologia, Universidade Federal de Minas Gerais, Minas Gerais, Brazil
| | - Oscar Bruña-Romero
- Departamento de Microbiologia, Universidade Federal de Minas Gerais, Minas Gerais, Brazil
- Departamento de Microbiologia, Imunologia e Parasitologia, Universidade Federal de Santa Catarina, Santa Catarina, Brazil
| |
Collapse
|
15
|
Fournier C, Martin F, Zitvogel L, Kroemer G, Galluzzi L, Apetoh L. Trial Watch: Adoptively transferred cells for anticancer immunotherapy. Oncoimmunology 2017; 6:e1363139. [PMID: 29147628 DOI: 10.1080/2162402x.2017.1363139] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Revised: 07/29/2017] [Accepted: 07/30/2017] [Indexed: 12/22/2022] Open
Abstract
Immunotherapies aimed at strengthening immune effector responses against malignant cells are growing at exponential rates. Alongside, the impressive benefits obtained by patients with advanced melanoma who received adoptively transferred tumor-infiltrating lymphocytes (TILs) have encouraged the scientific community to pursue adoptive cell transfer (ACT)-based immunotherapy. ACT involves autologous or allogenic effector lymphocytes that are generally obtained from the peripheral blood or resected tumors, expanded and activated ex vivo, and administered to lymphodepleted patients. ACT may be optionally associated with chemo- and/or immunotherapeutics, with the overall aim of enhancing the proliferation, persistence and functionality of infused cells, as well as to ensure their evolution in an immunological permissive local and systemic microenvironment. In addition, isolated lymphocytes can be genetically engineered to endow them with the ability to target a specific tumor-associated antigen (TAA), to increase their lifespan, and/or to reduce their potential toxicity. The infusion of chimeric antigen receptor (CAR)-expressing cytotoxic T lymphocytes redirected against CD19 has shown promising clinical efficacy in patients with B-cell malignancies. Accordingly, the US Food and Drug Administration (FDA) has recently granted 'breakthrough therapy' designation to a CAR-based T-cell therapy (CTL019) for patients with B-cell malignancies. Considerable efforts are now being devoted to the development of efficient ACT-based immunotherapies for non-hematological neoplasms. In this Trial Watch, we summarize recent clinical advances on the use of ACT for oncological indications.
Collapse
Affiliation(s)
- Carole Fournier
- INSERM, U1231, Dijon, France.,Faculté de Médecine, Université de Bourgogne Franche Comté, Dijon, France
| | - François Martin
- INSERM, U1231, Dijon, France.,Faculté de Médecine, Université de Bourgogne Franche Comté, Dijon, France
| | - Laurence Zitvogel
- Gustave Roussy Comprehensive Cancer Institute, Villejuif, France.,INSERM, U1015, Villejuif, France.,Center of Clinical Investigations in Biotherapies of Cancer (CICBT), Villejuif, France.,Université Paris Sud/Paris XI, Le Kremlin-Bicêtre, France
| | - Guido Kroemer
- Université Paris Descartes/Paris V, France.,Université Pierre et Marie Curie/Paris VI, Paris.,Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, Paris, France.,INSERM, U1138, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, Villejuif, France.,Karolinska Institute, Department of Women's and Children's Health, Karolinska University Hospital, Stockholm, Sweden.,Pôle de Biologie, Hopitâl Européen George Pompidou, AP-HP; Paris, France
| | - Lorenzo Galluzzi
- Université Paris Descartes/Paris V, France.,Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA.,Sandra and Edward Meyer Cancer Center, New York, NY, USA
| | - Lionel Apetoh
- INSERM, U1231, Dijon, France.,Faculté de Médecine, Université de Bourgogne Franche Comté, Dijon, France.,Centre Georges François Leclerc, Dijon, France
| |
Collapse
|
16
|
Santos JM, Havunen R, Siurala M, Cervera‐Carrascon V, Tähtinen S, Sorsa S, Anttila M, Karell P, Kanerva A, Hemminki A. Adenoviral production of interleukin‐2 at the tumor site removes the need for systemic postconditioning in adoptive cell therapy. Int J Cancer 2017; 141:1458-1468. [DOI: 10.1002/ijc.30839] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2016] [Revised: 05/05/2017] [Accepted: 05/23/2017] [Indexed: 12/26/2022]
Affiliation(s)
- Joao Manuel Santos
- TILT Biotherapeutics LtdHelsinki Finland
- Department of PathologyFaculty of Medicine, Cancer Gene Therapy Group, University of Helsinki Finland
| | - Riikka Havunen
- TILT Biotherapeutics LtdHelsinki Finland
- Department of PathologyFaculty of Medicine, Cancer Gene Therapy Group, University of Helsinki Finland
| | - Mikko Siurala
- TILT Biotherapeutics LtdHelsinki Finland
- Department of PathologyFaculty of Medicine, Cancer Gene Therapy Group, University of Helsinki Finland
| | - Víctor Cervera‐Carrascon
- TILT Biotherapeutics LtdHelsinki Finland
- Department of PathologyFaculty of Medicine, Cancer Gene Therapy Group, University of Helsinki Finland
| | - Siri Tähtinen
- Department of PathologyFaculty of Medicine, Cancer Gene Therapy Group, University of Helsinki Finland
| | - Suvi Sorsa
- TILT Biotherapeutics LtdHelsinki Finland
- Department of PathologyFaculty of Medicine, Cancer Gene Therapy Group, University of Helsinki Finland
| | - Marjukka Anttila
- Pathology Unit, Finnish Food Safety Authority (EVIRA)Helsinki Finland
| | - Pauliina Karell
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki Finland
| | - Anna Kanerva
- Department of PathologyFaculty of Medicine, Cancer Gene Therapy Group, University of Helsinki Finland
- Department of Obstetrics and GynecologyHelsinki University Hospital Finland
| | - Akseli Hemminki
- TILT Biotherapeutics LtdHelsinki Finland
- Department of PathologyFaculty of Medicine, Cancer Gene Therapy Group, University of Helsinki Finland
- Helsinki University Hospital Comprehensive Cancer CenterHelsinki Finland
| |
Collapse
|
17
|
Kim J, Chhour P, Hsu J, Litt HI, Ferrari VA, Popovtzer R, Cormode DP. Use of Nanoparticle Contrast Agents for Cell Tracking with Computed Tomography. Bioconjug Chem 2017; 28:1581-1597. [PMID: 28485976 PMCID: PMC5481820 DOI: 10.1021/acs.bioconjchem.7b00194] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
![]()
Efforts
to develop novel cell-based therapies originated with the
first bone marrow transplant on a leukemia patient in 1956. Preclinical
and clinical examples of cell-based treatment strategies have shown
promising results across many disciplines in medicine, with recent
advances in immune cell therapies for cancer producing remarkable
response rates, even in patients with multiple treatment failures.
However, cell-based therapies suffer from inconsistent outcomes, motivating
the search for tools that allow monitoring of cell delivery and behavior
in vivo. Noninvasive cell imaging techniques, also known as cell tracking,
have been developed to address this issue. These tools can allow real-time,
quantitative, and long-term monitoring of transplanted cells in the
recipient, providing insight on cell migration, distribution, viability,
differentiation, and fate, all of which play crucial roles in treatment
efficacy. Understanding these parameters allows the optimization of
cell choice, delivery route, and dosage for therapy and advances cell-based
therapy for specific clinical uses. To date, most cell tracking work
has centered on imaging modalities such as MRI, radionuclide imaging,
and optical imaging. However, X-ray computed tomography (CT) is an
emerging method for cell tracking that has several strengths such
as high spatial and temporal resolution, and excellent quantitative
capabilities. The advantages of CT for cell tracking are enhanced
by its wide availability and cost effectiveness, allowing CT to become
one of the most popular clinical imaging modalities and a key asset
in disease diagnosis. In this review, we will discuss recent advances
in cell tracking methods using X-ray CT in various applications, in
addition to predictions on how the field will progress.
Collapse
Affiliation(s)
| | | | | | | | | | - Rachela Popovtzer
- Department of Engineering, Bar-Ilan University , Ramat Gan, 5290002, Israel
| | | |
Collapse
|
18
|
Zhang T, Suryawanshi YR, Woyczesczyk HM, Essani K. Targeting Melanoma with Cancer-Killing Viruses. Open Virol J 2017; 11:28-47. [PMID: 28567163 PMCID: PMC5420172 DOI: 10.2174/1874357901711010028] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 01/05/2017] [Accepted: 01/17/2017] [Indexed: 12/20/2022] Open
Abstract
Melanoma is the deadliest skin cancer with ever-increasing incidence. Despite the development in diagnostics and therapies, metastatic melanoma is still associated with significant morbidity and mortality. Oncolytic viruses (OVs) represent a class of novel therapeutic agents for cancer by possessing two closely related properties for tumor reduction: virus-induced lysis of tumor cells and induction of host anti-tumor immune responses. A variety of viruses, either in "natural" or in genetically modified forms, have exhibited a remarkable therapeutic efficacy in regressing melanoma in experimental and/or clinical studies. This review provides a comprehensive summary of the molecular and cellular mechanisms of action of these viruses, which involve manipulating and targeting the abnormalities of melanoma, and can be categorized as enhancing viral tropism, targeting the tumor microenvironment and increasing the innate and adaptive antitumor responses. Additionally, this review describes the "biomarkers" and deregulated pathways of melanoma that are responsible for melanoma initiation, progression and metastasis. Advances in understanding these abnormalities of melanoma have resulted in effective targeted and immuno-therapies, and could potentially be applied for engineering OVs with enhanced oncolytic activity in future.
Collapse
Affiliation(s)
- Tiantian Zhang
- Laboratory of Virology, Department of Biological Sciences, Western Michigan University, Kalamazoo, MI, 49008, U.S.A
| | - Yogesh R. Suryawanshi
- Laboratory of Virology, Department of Biological Sciences, Western Michigan University, Kalamazoo, MI, 49008, U.S.A
| | - Helene M. Woyczesczyk
- Laboratory of Virology, Department of Biological Sciences, Western Michigan University, Kalamazoo, MI, 49008, U.S.A
| | - Karim Essani
- Laboratory of Virology, Department of Biological Sciences, Western Michigan University, Kalamazoo, MI, 49008, U.S.A
| |
Collapse
|
19
|
Havunen R, Siurala M, Sorsa S, Grönberg-Vähä-Koskela S, Behr M, Tähtinen S, Santos JM, Karell P, Rusanen J, Nettelbeck DM, Ehrhardt A, Kanerva A, Hemminki A. Oncolytic Adenoviruses Armed with Tumor Necrosis Factor Alpha and Interleukin-2 Enable Successful Adoptive Cell Therapy. MOLECULAR THERAPY-ONCOLYTICS 2016; 4:77-86. [PMID: 28345026 PMCID: PMC5363700 DOI: 10.1016/j.omto.2016.12.004] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Accepted: 12/20/2016] [Indexed: 01/18/2023]
Abstract
Adoptive cell therapy holds much promise in the treatment of cancer but results in solid tumors have been modest. The notable exception is tumor-infiltrating lymphocyte (TIL) therapy of melanoma, but this approach only works with high-dose preconditioning chemotherapy and systemic interleukin (IL)-2 postconditioning, both of which are associated with toxicities. To improve and broaden the applicability of adoptive cell transfer, we constructed oncolytic adenoviruses coding for human IL-2 (hIL2), tumor necrosis factor alpha (TNF-α), or both. The viruses showed potent antitumor efficacy against human tumors in immunocompromised severe combined immunodeficiency (SCID) mice. In immunocompetent Syrian hamsters, we combined the viruses with TIL transfer and were able to cure 100% of the animals. Cured animals were protected against tumor re-challenge, indicating a memory response. Arming with IL-2 and TNF-α increased the frequency of both CD4+ and CD8+ TILs in vivo and augmented splenocyte proliferation ex vivo, suggesting that the cytokines were important for T cell persistence and proliferation. Cytokine expression was limited to tumors and treatment-related signs of systemic toxicity were absent, suggesting safety. To conclude, cytokine-armed oncolytic adenoviruses enhanced adoptive cell therapy by favorable alteration of the tumor microenvironment. A clinical trial is in progress to study the utility of Ad5/3-E2F-d24-hTNFa-IRES-hIL2 (TILT-123) in human patients with cancer.
Collapse
Affiliation(s)
- Riikka Havunen
- Cancer Gene Therapy Group, Faculty of Medicine, University of Helsinki, 00290 Helsinki, Finland
| | - Mikko Siurala
- Cancer Gene Therapy Group, Faculty of Medicine, University of Helsinki, 00290 Helsinki, Finland; TILT Biotherapeutics, Ltd., 00290 Helsinki, Finland
| | - Suvi Sorsa
- Cancer Gene Therapy Group, Faculty of Medicine, University of Helsinki, 00290 Helsinki, Finland; TILT Biotherapeutics, Ltd., 00290 Helsinki, Finland
| | | | - Michael Behr
- Cancer Gene Therapy Group, Faculty of Medicine, University of Helsinki, 00290 Helsinki, Finland
| | - Siri Tähtinen
- Cancer Gene Therapy Group, Faculty of Medicine, University of Helsinki, 00290 Helsinki, Finland
| | - João Manuel Santos
- Cancer Gene Therapy Group, Faculty of Medicine, University of Helsinki, 00290 Helsinki, Finland; TILT Biotherapeutics, Ltd., 00290 Helsinki, Finland
| | - Pauliina Karell
- Institute for Molecular Medicine Finland, University of Helsinki, 00290 Helsinki, Finland
| | - Juuso Rusanen
- Cancer Gene Therapy Group, Faculty of Medicine, University of Helsinki, 00290 Helsinki, Finland
| | | | - Anja Ehrhardt
- Faculty of Health, Institute for Virology and Microbiology, University Witten/Herdecke, 58448 Witten, Germany
| | - Anna Kanerva
- Cancer Gene Therapy Group, Faculty of Medicine, University of Helsinki, 00290 Helsinki, Finland; Department of Obstetrics and Gynecology, Helsinki University Central Hospital, 00610 Helsinki, Finland
| | - Akseli Hemminki
- Cancer Gene Therapy Group, Faculty of Medicine, University of Helsinki, 00290 Helsinki, Finland; TILT Biotherapeutics, Ltd., 00290 Helsinki, Finland; Comprehensive Cancer Center, Helsinki University Hospital, 00290 Helsinki, Finland
| |
Collapse
|
20
|
Dany M, Nganga R, Chidiac A, Hanna E, Matar S, Elston D. Advances in immunotherapy for melanoma management. Hum Vaccin Immunother 2016; 12:2501-2511. [PMID: 27454404 PMCID: PMC5085014 DOI: 10.1080/21645515.2016.1190889] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Revised: 04/26/2016] [Accepted: 05/13/2016] [Indexed: 12/31/2022] Open
Abstract
Melanoma remains a leading cause of death among young adults. Evidence that melanoma tumor cells are highly immunogenic and a better understanding of T-cell immune checkpoints have changed the therapeutic approach to advanced melanoma. Instead of targeting the tumor directly, immunotherapy targets and activates the immune response using checkpoint inhibitors, monoclonal antibodies, vaccines, and adoptive T cell therapy. This review focuses on the immune signaling and biological mechanisms of action of recent immune-based melanoma therapies as well as their clinical benefits.
Collapse
Affiliation(s)
- Mohammed Dany
- Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston, SC, USA
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA
| | - Rose Nganga
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA
| | - Alissar Chidiac
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA
| | - Edith Hanna
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA
| | - Sara Matar
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA
| | - Dirk Elston
- Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston, SC, USA
| |
Collapse
|
21
|
Kursunel MA, Esendagli G. The untold story of IFN-γ in cancer biology. Cytokine Growth Factor Rev 2016; 31:73-81. [DOI: 10.1016/j.cytogfr.2016.07.005] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Revised: 07/07/2016] [Accepted: 07/07/2016] [Indexed: 12/21/2022]
|
22
|
Zeltsman M, Mayor M, Jones DR, Adusumilli PS. Surgical immune interventions for solid malignancies. Am J Surg 2016; 212:682-690.e5. [PMID: 27659157 DOI: 10.1016/j.amjsurg.2016.06.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 06/17/2016] [Accepted: 06/17/2016] [Indexed: 12/19/2022]
Abstract
BACKGROUND The purpose of this study was to systematically review clinically translatable immunotherapeutic agents that are delivered regionally for solid malignancies. DATA SOURCES PubMed and ClinicalTrials.gov were searched for published and registered clinical trials, respectively. The search yielded 334 relevant publications, of which 116 articles were included for review after exclusion criteria were applied. CONCLUSIONS There has been an increase in the regional administration of cell-based and viral vector-based clinical trials over the last 5 years. Surgical interventions have been developed for intrapleural, intracranial, intraperitoneal, and intratumoral routes of access to enhance the local delivery of these therapies. Multimodality therapies that combine regional immunotherapy with other local and systemic therapies are demonstrating continued growth as the field of immunotherapy continues to expand.
Collapse
Affiliation(s)
- Masha Zeltsman
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY, USA
| | - Marissa Mayor
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY, USA
| | - David R Jones
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY, USA
| | - Prasad S Adusumilli
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY, USA; Center for Cell Engineering, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY, USA.
| |
Collapse
|
23
|
Adenoviral Delivery of Tumor Necrosis Factor-α and Interleukin-2 Enables Successful Adoptive Cell Therapy of Immunosuppressive Melanoma. Mol Ther 2016; 24:1435-43. [PMID: 27357626 DOI: 10.1038/mt.2016.137] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Accepted: 06/20/2016] [Indexed: 12/12/2022] Open
Abstract
Adoptive T-cell transfer is a promising treatment approach for metastatic cancer, but efficacy in solid tumors has only been achieved with toxic pre- and postconditioning regimens. Thus, adoptive T-cell therapies would benefit from complementary modalities that enable their full potential without excessive toxicity. We aimed to improve the efficacy and safety of adoptive T-cell transfer by using adenoviral vectors for direct delivery of immunomodulatory murine cytokines into B16.OVA melanoma tumors with concomitant T-cell receptor transgenic OT-I T-cell transfer. Armed adenoviruses expressed high local and low systemic levels of cytokine when injected into B16.OVA tumors, suggesting safety of virus-mediated cytokine delivery. Antitumor efficacy was significantly enhanced with adenoviruses coding for murine interleukin-2 (mIL-2) and tumor necrosis factor-α (mTNFα) when compared with T-cell transfer alone or viruses alone. Further improvement in efficacy was achieved with a triple combination of mIL-2, mTNFα, and OT-I T-cells. Mechanistic studies suggest that mIL-2 has an important role in activating T-cells at the tumor, while mTNFα induces chemokine expression. Furthermore, adenovirus treatments enhanced tumor-infiltration of OT-I T-cells as demonstrated by SPECT/CT imaging of (111)In-labeled cells. Our results suggest the utility of cytokine-coding adenoviruses for improving the efficacy of adoptive T-cell therapies.
Collapse
|
24
|
Jin P, Zhao Y, Liu H, Chen J, Ren J, Jin J, Bedognetti D, Liu S, Wang E, Marincola F, Stroncek D. Interferon-γ and Tumor Necrosis Factor-α Polarize Bone Marrow Stromal Cells Uniformly to a Th1 Phenotype. Sci Rep 2016; 6:26345. [PMID: 27211104 PMCID: PMC4876328 DOI: 10.1038/srep26345] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Accepted: 04/26/2016] [Indexed: 01/21/2023] Open
Abstract
Activated T cells polarize mesenchymal stromal cells (MSCs) to a proinflammatory Th1 phenotype which likely has an important role in amplifying the immune response in the tumor microenvironment. We investigated the role of interferon gamma (IFN-γ) and tumor necrosis factor alpha (TNF-α), two factors produced by activated T cells, in MSC polarization. Gene expression and culture supernatant analysis showed that TNF-α and IFN-γ stimulated MSCs expressed distinct sets of proinflammatory factors. The combination of IFN-γ and TNF-α was synergistic and induced a transcriptome most similar to that found in MSCs stimulated with activated T cells and similar to that found in the inflamed tumor microenvironment; a Th1 phenotype with the expression of the immunosuppressive factors IL-4, IL-10, CD274/PD-L1 and indoleamine 2,3 dioxygenase (IDO). Single cell qRT-PCR analysis showed that the combination of IFN-γ and TNF-α polarized uniformly to this phenotype. The combination of IFN-γ and TNF-α results in the synergist uniform polarization of MSCs toward a primarily Th1 phenotype. The stimulation of MSCs by IFN-γ and TNF-α released from activated tumor infiltrating T cells is likely responsible for the production of many factors that characterize the tumor microenvironment.
Collapse
Affiliation(s)
- Ping Jin
- Cell Processing Section, Department of Transfusion Medicine, Clinical Center, National Institutes of Health, Bethesda, Maryland 20892 USA
| | - Yuanlong Zhao
- Cell Processing Section, Department of Transfusion Medicine, Clinical Center, National Institutes of Health, Bethesda, Maryland 20892 USA
| | - Hui Liu
- Cell Processing Section, Department of Transfusion Medicine, Clinical Center, National Institutes of Health, Bethesda, Maryland 20892 USA
| | - Jinguo Chen
- Center for Human Immunology (CHI), National Institutes of Health, Bethesda, MD, 20892, USA
| | - Jiaqiang Ren
- Cell Processing Section, Department of Transfusion Medicine, Clinical Center, National Institutes of Health, Bethesda, Maryland 20892 USA
| | - Jianjian Jin
- Cell Processing Section, Department of Transfusion Medicine, Clinical Center, National Institutes of Health, Bethesda, Maryland 20892 USA
| | | | - Shutong Liu
- Cell Processing Section, Department of Transfusion Medicine, Clinical Center, National Institutes of Health, Bethesda, Maryland 20892 USA
| | - Ena Wang
- Research Branch, Sidra Medical and Research Center, Doha, Qatar
| | | | - David Stroncek
- Cell Processing Section, Department of Transfusion Medicine, Clinical Center, National Institutes of Health, Bethesda, Maryland 20892 USA
| |
Collapse
|